US20110097708A1 - Assessment of human papilloma virus-related disease - Google Patents

Assessment of human papilloma virus-related disease Download PDF

Info

Publication number
US20110097708A1
US20110097708A1 US12/976,428 US97642810A US2011097708A1 US 20110097708 A1 US20110097708 A1 US 20110097708A1 US 97642810 A US97642810 A US 97642810A US 2011097708 A1 US2011097708 A1 US 2011097708A1
Authority
US
United States
Prior art keywords
time point
range
low
hpv
gene expression
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US12/976,428
Inventor
Attila T. Lorincz
James G. Lazar
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Qiagen Gaithersburg LLC
Original Assignee
Qiagen Gaithersburg LLC
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=27371534&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20110097708(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Qiagen Gaithersburg LLC filed Critical Qiagen Gaithersburg LLC
Priority to US12/976,428 priority Critical patent/US20110097708A1/en
Publication of US20110097708A1 publication Critical patent/US20110097708A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/70Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving virus or bacteriophage
    • C12Q1/701Specific hybridization probes
    • C12Q1/708Specific hybridization probes for papilloma
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01NPRESERVATION OF BODIES OF HUMANS OR ANIMALS OR PLANTS OR PARTS THEREOF; BIOCIDES, e.g. AS DISINFECTANTS, AS PESTICIDES OR AS HERBICIDES; PEST REPELLANTS OR ATTRACTANTS; PLANT GROWTH REGULATORS
    • A01N1/00Preservation of bodies of humans or animals, or parts thereof
    • A01N1/02Preservation of living parts
    • A01N1/0205Chemical aspects
    • A01N1/021Preservation or perfusion media, liquids, solids or gases used in the preservation of cells, tissue, organs or bodily fluids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6806Preparing nucleic acids for analysis, e.g. for polymerase chain reaction [PCR] assay
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/112Disease subtyping, staging or classification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/118Prognosis of disease development

Definitions

  • the present invention is generally related to the field of cytological and 10 molecular assays and specifically to the area of assays for the assessment of disease using a sensitive assay for diagnosis and prognosis of HPV-induced carcinoma.
  • the detection and diagnosis of disease is of obvious importance for the treatment of disease. Numerous characteristics of diseases have been identified and many are used for the diagnosis of disease. Many diseases are preceded by, and are characterized by, changes in the state of the affected cells. Changes can include the expression of viral genes in infected cells, changes in the expression patterns of genes in affected cells, and changes in cell morphology. The detection, diagnosis, and monitoring of diseases can be aided by the assessment of such cell states.
  • An aspect of the present invention relates to human papilloma virus (HPV), which induces benign epithelial proliferations of the skin and mucosa in humans and is associated with anogenital neoplasias and carcinomas.
  • HPV human papilloma virus
  • the intact DNA of HPV is supercoiled and thus resembles an endless loop of twisted telephone handset cord.
  • the viral DNA is packaged in and around proteins from the cell nucleus, histones, and associated peptides, into a structure that resembles cellular chromatin. (Turek, (1994)).
  • Human papillomaviruses characterized to date are associated with lesions confined to the epithelial layers of skin, or oral, pharyngeal, respiratory, and, most importantly, anogenital mucosae.
  • Specific human papillomavirus types including HPV 6 and 11, frequently cause benign mucosal lesions, whereas other types, HPV 16, 18, and a host of other strains, are predominantly found in high-grade lesions and cancer. All human and animal papillomaviruses appear to share a similar genetic organization, although there are differences in the functions of individual viral genes and in their regulation. The most common genital HPV type associated with cervical carcinoma, HPV 16, has been studied most extensively.
  • All large open reading frames (ORFs) in HPV are on one DNA strand.
  • Papillomaviral mRNAs appear to be transcribed solely from a single strand in infected cells.
  • the viral genome can be divided into three regions, the upstream regulatory region (URR), or long control region (LCR), containing control sequences for HPV replication and gene expression, the viral early gene region, encoding, among others, the E2, E6 and E7 genes, and the late region, encoding the L1 and L2 genes. (Turek, (1994)).
  • HPV gene expression in high-grade premalignant disease or cancer appears restricted to the early genes, possibly due to cellular differentiation arrest induced by the viral E6 and E7 genes.
  • E6 and E7 gene control in cancer is deranged by mutations in the viral URR and, in integrated viral fragments, by the disruption of the viral E2 gene, stabilization of E6 and E7 mRNAs, and influences at the cellular integration site.
  • Genes E6 and E7 are considered to have oncogenic activity.
  • the encoded proteins interact with and disturb the physiologic functions of cellular proteins that are involved in cell cycle control.
  • the E6/E7 proteins of HPV 16, 18 or related types are most efficient in this regard. Some of these activities lead to genetic instability of the persistently infected human cell. This enhances the probability of mutations in cellular proto-oncogenes and tumor suppressor genes and thus contributes to tumor progression.
  • Mutations in cellular genes devoted to the intracellular surveillance of HPV infections, integration of viral DNA, and deletions or mutations of viral transcription control sequences lead to a significantly increased expression of the E6/E7 genes, which is a consistent characteristic of high-grade intraepithelial neoplasia and cancers.
  • the genetic instability caused by viral oncoproteins and the autocatalytic increase in oncoprotein expression caused by mutations in the viral and cellular genome identify the virus as a major driving force of progression to carcinoma.
  • HPV human papillomaviruses
  • Yet another object of the present invention to provide a method for monitoring the effectiveness of treatment of HPV-based disease.
  • a further object of the present invention to provide kits for assessing the stage of HPV-based disease.
  • Another object of the present invention to provide computer-based operation, analysis, and data management of assay data to assess the stage of HPV-based disease.
  • One embodiment of the present invention involves measuring the levels of expression of genes involved in a cell state, and comparing their expression to each other or to reference genes in a specific ratio, as an indication of the state of a disease in the cell sample.
  • the present invention can be used to assess the stage or risk of a disease as indicated by the state of the cells. It can also be used to guide or assess the effectiveness of a therapy for a disease by identifying appropriate therapy based on the indicated cell state or by indicating any change in the state of cells subjected to the therapy.
  • the stage and prognosis of a human papillomavirus (HPV) infection or HPV-based disease is assessed.
  • HPV human papillomavirus
  • This embodiment involves the measurement of the level of expression of two or more HPV genes. Genes for this purpose are the HPV E6, E7, L1, and E2 genes, although other HPV genes such as E1, E4, E5, and L2 can also be used. It has been discovered that the level of expression of these genes, the ratio of expression of these genes to each other or to reference genes, or both, are indicative of the stage of HPV-based disease.
  • Gene expression levels are used according to this invention to assess the progression of HPV infection from benign to malignant growth.
  • HPV infection progresses from CIN I through CIN TTT and finally to malignant cancer.
  • These stages can be identified by the ratios of HPV genes.
  • the transition from CIN I to CIN II/III i.e., a transition to pre-malignancy
  • a ratio of greater than 3 in the present invention indicates a transition from pre-malignancy to malignant cancer.
  • a ratio below 1 indicates a low-level of CIN in an HPV infected cell.
  • a ratio between one and about three indicates a high grade CIN (i.e., CIN III) or pre-malignant condition.
  • a ratio of over about three is an indication of an HPV induced malignancy.
  • FIG. 1 is a graph of detection of E6/E7 mRNA shown as the number of cells/well versus the number of E6/E7 mRNA/well.
  • FIG. 2 is a graph of the sensitivity of an HPV L1 assay shown as the number of RNA molecules/well versus the signal-to-noise ratio minus 1.
  • FIG. 3 is the probe sequence for HPV 16 L2 (SEQ ID NO: 2).
  • FIG. 4 is the probe sequence for HPV 16 L1 (SEQ ID NO: 2).
  • FIG. 5 is the probe sequence for HPV 16 E6/E7 (SEQ ID NO: 3).
  • FIG. 6 is the probe sequence for HPV 16 E2 (SEQ ID NO: 4).
  • FIG. 7 is the probe sequence for HPV 16 E4 (SEQ ID NO: 5).
  • the present invention relates to the identification and monitoring of diseased cells.
  • One method involves measuring the levels of expression of genes involved in a disease state, and comparing their expression to each other or to reference genes, as an indication of the state of the cells. Such measurements can be combined with other assays to increase the accuracy and reliability of the assessment of the disease state.
  • One method of the present invention can be used to assess the stage of a disease as indicated by the state of the cells. This method can also be used to guide or assess the effectiveness of a therapy for a disease by identifying appropriate therapy based on the indicated disease state or by indicating any change in the state of cells subjected to the therapy.
  • neoplastic and cancerous cells generally exhibit certain distinctive morphologies and growth characteristics. Molecular characteristics, such as gene mutations and gene expression patterns are also a good indicator of disease progression. Virally infected cells can exhibit different morphologies and gene expression patterns, including expression of viral genes. Using the present invention, the characteristics of the cell state, such as changes in cell morphology or expression of genes can be determined from a patient sample.
  • the characteristics to be detected are generally specific to the cell state of interest and the disease suspected of being present in the cell sample. Such characteristics can be generally divided into two types, cytological characteristics and molecular characteristics.
  • cytological characteristics are characteristics such as, for example, overall cell shape and appearance. The primary identification and classification of many neoplastic and cancerous cells has traditionally been accomplished using cytological characteristics. Identification of cytological characteristics is generally slow, requires a relatively high level of training, and generally cannot be easily automated.
  • molecular characteristics are the presence and state of particular molecular species, such as proteins, nucleic acids, and metabolites. Such molecular characteristics are generally identified by detecting and measuring the particular molecules of interest.
  • the characteristics assayed can include additional or surrogate marker characteristics that are not a direct cause or result of the disease but that are related to certain disease and cell states.
  • additional markers include polymorphic markers, human leukocyte antigens (HLA) such as B7 that predispose women for cervical carcinomas, oncogenes, p53 mutations, other cancer markers, oncosupressors, cytokines, growth factor receptors, and hormones.
  • HLA human leukocyte antigens
  • Such markers can be present in, or absent from, cells exhibiting state-or disease-specific characteristics, and such presence or absence can be indicative of for example, a more severe or less severe disease state.
  • markers can be used in conjunction with the disclosed method to infer either higher or lower risk of neoplastic disease depending on the number of abnormal scores or the magnitude of change in quantitative markers.
  • disease states for assessment using the present invention include, but are not limited to neoplasias and cancer.
  • Disease states of interest are HPV-based disease—including HPV infection, cervical intraepithelial neoplasia (CIN), and cancer, atypical squamous cells of undetermined significance (ASCUS), warts, condylomata, epidemic) dysplasia verruciformis and other skin diseases, laryngeal papilloma, oral papilloma and conjunctival papilloma.
  • HPV-based disease including HPV infection, cervical intraepithelial neoplasia (CIN), and cancer, atypical squamous cells of undetermined significance (ASCUS), warts, condylomata, epidemic) dysplasia verruciformis and other skin diseases, laryngeal papilloma, oral papilloma and conjunctival papilloma.
  • An embodiment of the present invention is detection and measurement of the expression levels of certain HPV genes.
  • An impressive amount of data has been accumulated over the past decade, showing that carcinoma of the cervix is associated with infection of certain types of HPV.
  • carcinoma of the cervix is associated with infection of certain types of HPV.
  • the presence of HPV DNA in a precancerous lesion is indicative of an increased relative risk for cervical dysplasia and invasive carcinoma, it is still difficult to predict the clinical behavior of precancerous cervical lesions. Tumors arise due to the accumulation of genetic alterations which can activate oncogenes and/or inactivate tumor suppressor genes and/or genes involved in DNA damage recognition and repair.
  • the expression levels of E6 and E7 oncoproteins encoded by high-risk HPV types are a more sensitive and accurate measure of potential risk of an HPV infection developing into a cancerous lesion.
  • the present invention measures the relative amounts of E6 and/or E7 expression levels and E2 and/or L1 in an HPV-infected lesion to determine the ratio of E6 and/or E7 to L1 and/or E2, where in this ratio is a direct measure of risk, and susceptibility to the development of a cancerous lesion.
  • HPV expression can be measured by mRNA or protein levels in the cell.
  • the stage and prognosis of a human papillomavirus (HPV) infection or HPV-based disease is assessed.
  • This embodiment of the present invention involves the measurement of the level of expression of one or more HPV genes discovered to be related to the stage and nature of HPV-based disease.
  • Genes 25 useful for this purpose include the HPV E6, E7, L1, and E2 genes. It has been discovered that the level of expression of these genes, the ratio of expression of these genes to each other or to one or more other genes, or both, are indicative of the stage of HPV-based disease.
  • the level of expression is relative to other HPV genes, or the level of expression relative to a non-HPV gene, referred to herein as a reference gene.
  • reference genes can be any appropriate gene (not encoded by HPV), and are, for example, housekeeping genes or other constitutively expressed genes.
  • reference genes include actin genes, cytoskeletal genes, histone genes, tubulin genes, epidermal growth factor receptor genes, the normal p53 gene, the normal pRB gene, cyclin genes, 13-globin genes, and glucose-6-phosphate dehydrogenase genes. Expression of reference genes can be measured in the same cell as the level of HPV genes are measured or in neighboring cells in the same cell sample. In such a case, the reference gene is an internal control for gene expression.
  • HPV-based high grade CIN referenced in the table is the premalignant stage leading to cancer and low grade CIN is a productive viral infection that has little malignant potential but is a public health concern with respect to the spread of HPV infection.
  • Normal tissue refers to cytologically normal tissue that is infected with HPV. Although not limited to this standard, one standard for establishing this lower limit of expression is a level below that detectable using the hybrid capture assay described in WO 93/10263 by Digene.
  • E6/E7 refers to E6, E7, or E6 +E7.
  • each major disease state is represented by a unique expression pattern of these three sets of genes. Both of these conditions are regarded as serious medical aliments.
  • Other relationships involving the relative level 15 expression of other HPV genes can also be used to assess cell state.
  • L2 and E4 are frequently associated with benign viral production diseases, and E1 is similar in profile to E2 and is often deleted in malignancies.
  • Other relationships of expression for these HPV proteins can exist for other HPV-based diseases, and the disclosed method can be used to assess the state of such other diseases using the appropriate levels and ratios for that disease.
  • the stage of the disease can be assessed in several ways.
  • the presence or absence of detectable expression is indicative of the disease state in the infected cells.
  • a lack of E2 expression is indicative of high grade CIN or cancer.
  • a change or difference in expression of an HPV gene product can be indicative of change occurring in the infected cell state.
  • an increased level of expression of E6 and E7 relative to, for example, an earlier sample or a reference sample—may be indicative of high grade CIN or cancer.
  • a change in the ratio of E6/E7 expression to E2 expression is used to identify low grade CIN or a shift from normal cells to low grade CIN.
  • Many other combinations of comparisons are also possible, and other combinations can be derived from the information in Table 1 for use in the method of the present invention.
  • group 1 genes or gene sets include E6, E7, and E6+E7.
  • Group 2 genes or gene sets include L1, L2, E4, and any combinations.
  • Group 3 genes or gene sets include E1, E2, E5, and any combinations.
  • Useful ratios of expression include ratios of members of group 1 to members of group 2 or group 3. Examples of theses ratios are (E6+E7)/(L1+L2), E6/L1, E7/L1, E6/L2, E7/L2, (E6+E7)/L1, (E6+E7)/L2), E6/(L1+L2), and E7/(L1+L2).
  • a value of less than two is indicative of benign human papillomavirus infection or low grade intra-epithelial neoplasia.
  • This type of infection is also classified as CIN I.
  • HPV infections which progress beyond CIN I indicate cell transformation has occurred and cancerous growth has begun.
  • CIN FM later infections
  • a ratio of expression of more than two is indicative of high grade intra-epithelial neoplasia or pre-malignant cancer.
  • a ratio of expression of much more than two is indicative of cancer.
  • Preferred ratios for use in this invention are (E6+E7)/L1, (E6+E7)/(L1+L2), (E6+E7+E2+E4)/(L1+L2), (E6+E7)/(E2+E4), (E6+E7)/E2, (E6+E7+E2-E4)/(L1+L2).
  • measured levels of expression of HPV genes can be compared and categorized. For example, where the presence or absence of expression is indicative of the cell state, expression of the HPV gene is analyzed without reference to the expression level of other genes. Where the relative level of expression of an HPV gene is indicative of the cell state, the measured level of expression is compared, for example, to the level of expression of the same type of HPV gene in a different cell sample (such as an earlier cell sample from the same source or reference cells harboring HPV), to the level of expression of a different type of HPV gene in the same or a different cell sample, to the level of expression of a non-HPV reference gene in the same cell sample, or to the level of expression of a non-HPV reference gene in reference cells.
  • a different cell sample such as an earlier cell sample from the same source or reference cells harboring HPV
  • levels and ratios of expression of HPV genes are compared to the levels of the same genes in a cell line that contains HPV (such as HeLa or CaSki).
  • HPV such as HeLa or CaSki
  • Such cell lines provide a standard against which levels of expression of HPV genes in cell samples are compared.
  • Such comparisons are used to assess and compare the absolute levels of expression of these HPV proteins with those in a standard or comparative cell line.
  • Other cell lines useful for this purpose are non-cancerous cell lines infected with HPV 16 (such as W12) or HPV 31 (such as ON612; Meyers (1992)).
  • the levels and ratios of expression of HPV genes are also compared to reference genes, such as housekeeping genes or other constitutively expressed genes, in the same cells or in reference cells, such as a cell line.
  • reference genes such as housekeeping genes or other constitutively expressed genes
  • the level of expression of the HPV gene and the reference gene is measured in the same cell sample.
  • Such measurements provide an internal control of the overall expression level in a cell sample and are used to calculate a corrected level of expression for the HPV gene to allow more accurate comparisons of the level of expression between different cell samples.
  • One form of correction is referred to as normalization.
  • the level of expression of one or more HPV genes can be measured in two or more cell samples along with the level of expression of the same reference gene in each of the cell samples.
  • the level of expression of the HPV genes is then normalized to each other based on differences, if any, between the measured level of expression of the reference gene in each of the samples.
  • the expression of a particular HPV gene is low or undetectable. Such lack of detectable expression is used herein to identify patterns of expression that serve as prognostic or diagnostic indicators.
  • the expression of other HPV genes is assessed in parallel or in the same assay to provide a control for the lack of expression of one or more of the genes to be assessed. This is accomplished by measuring the expression of the several HPV genes together or in parallel. For example, measuring the level of expression of the HPV E6, E7, L1, E4, and E2 genes in parallel is useful since at least one of these genes is expressed in all of the stages of HPV-based disease.
  • a ratio of E6/E7 to L1 /E2 provides an indicator of likelihood of developing cancer from the HPV infection. In this way, indicative information is collected as well as providing an internal control.
  • the presence of L1, L2, and E4 in combination is also indicative of benign disease while the absence of E1 and/or E2 is indicative of neoplastic potential.
  • the types of comparison described above can also be used with other genes and other disease states. That is, the measured level of expression of a gene of interest can be compared, for example, to the level of expression of the same type of gene in a different cell sample (such as an earlier cell sample from the same source or appropriate reference cells), to the level of expression of a different type of gene in the same or a different cell sample, to the level of expression of a reference gene in the same cell sample, or to the level of expression of a reference gene in reference cells.
  • a different cell sample such as an earlier cell sample from the same source or appropriate reference cells
  • RNA can be detected using hybridization, amplification, or sequencing techniques, and protein can be detected using specific antibodies. Many techniques for the specific detection of gene expression, by detection of expression products, are known and can be used with the disclosed method.
  • One technique for detecting and measuring the level of expression of genes of interest is detection of RNA transcribed from the genes of interest. For the most reliable comparisons, expression levels that are to be compared should be measured using the same technique and be performed in the same manner.
  • probes specific for these sequences from different HPV types can be used. This ensures that the method will detect expression regardless of the type of HPV involved.
  • probes designed for the sequence of a certain HPV type or a mixture of probes for only some HPV types. Such probes may or may not be type-specific depending on the differences between the sequences of the HPV nucleic acids to be detected.
  • One useful mixture for this purpose would include probes for HPV types more closely associated with a progression to cancer.
  • the HPV types most commonly associated with cervical cancer are types 16 and 18.
  • Useful techniques for measuring the level of expression of a gene of interest in a cell sample include the hybrid capture technique described in WO 93/10263 by Digene, PCR in situ hybridization techniques described by (Nuovo, 1997)), branched DNA assays (Chernoff (1997)), transcription-mediated amplification (TMA); Stoflet (1988)), and polymerase chain reaction (PCR), ligase chain reaction (LCR), self-sustained sequence replication (3SR), nucleic acid sequence based amplification (NASBA), strand displacement amplification (SDA), and amplification with QB replicase (Birkenmeyer and Mushahwar, (1991); Landegren, (1993)).
  • HPV detection and typing assays which can be used in the disclosed method, are known, including assays involving Southern blots, dot blots, in situ hybridization, polymerase chain reaction, and solution hybridization.
  • Mant describes PCR assays used to identify DNA from specific HPV types.
  • Cope describes a PCR-based test using a consensus primer and a Hybrid Capture assay (HCA) of detection of HPV types.
  • HCA Hybrid Capture assay
  • the hybrid capture assay is also described in WO 93/10263 by Digene.
  • the hybrid capture assay is a useful method for detection of HPV and for determining HPV type in combination with the disclosed assay.
  • Swan discloses an HPV detection assay exploiting the 5′ to 3′ exonucleolytic activity of Taq DNA polymerase to increase the signal from fluorescent dyes by releasing them from genotype-specific probes during PCR.
  • Lizardi describes a method of detecting HPV using in situ hybridization with non-radioactive probes and visualization with conventional bright-field or fluorescence microscopy, or laser scanning confocal microscopy.
  • Zehbe [1] (1997), describes a modified version of in situ hybridization for detection of HPV that involves signal amplification.
  • Leiserowitz describes use of reverse transcriptase-polymerase chain reaction to detect HPV.
  • Zehbe [2] describes a nonisotopic, enzyme-linked immunosorbent assay-based sandwich capture hybridization assay for HPV detection.
  • the cells were first lysed by adding a proteolytic enzyme to the cells contained in wells of a microtiter plate.
  • enzymes for use in the present invention include proteinase K or Pronase.
  • Cells can also be subjected to detergent lysis or osmotic lysis or a French Press. After incubation, biotinylated DNA probes were added to each well.
  • the RNA:DNA hybrids were captured onto a solid phase by transferring to streptavidin coated microplates.
  • RNA:DNA hybrids Alkaline phosphataseconjugated antibodies to RNA:DNA hybrids were added to each well in the hybridization microplate and signals were generated by adding a chemiluminescent reagent such as CDP-Star® with Emerald II (Tropix) to each well. The signal was read from the microplate.
  • the solution based DNA analysis was performed similarly to the RNA analysis except that the microtiter plates were coated with anti-RNA:DNA hybrid antibodies and the probes were RNA probes.
  • the disclosed assay and other assays can also be sequentially combined. That is, first one type of assay can be performed, and then, depending on the results, another assay can be performed. For example, an assay to detect HPV (or HPV type) can be performed first, then, if HPV is present, the disclosed molecular assay can be 25 performed. As another example, the disclosed molecular assay can be performed first, and if the results of the assay were indicative of a high grade CIN or cancer, a cytological assay or biopsy can be performed. Such sequential combinations are particularly useful for limiting more extensive testing to patients and samples that are identified as high risk.
  • Useful sequential orders for the assays are (1) an HPV assay, followed by an assay for one or more other markers, followed by a cytological or histological assay; (2) a cytological or histological assay, followed by an HPV assay or an assay for one or more other markers, followed by an HPV assay or an assay for one or more other markers (whichever had not been performed first); (3) a cytological or histological assay, followed by a combined or simultaneous HPV and marker assay; (4) a combined or simultaneous HPV and marker assay, followed by a cytological or histological assay; and (5) detection of HPV, detection of HPV type, an HPV assay, and a cytological or histological assay.
  • Each combination of assays is followed by an assessment, using the combined assay results, of the cell state, disease state, patient status, patient prognosis, or other assessment as described herein.
  • results of initial assays are either equivocal or suggest a more severe stage of disease
  • further assays are useful to clarify and confirm the initial results.
  • a cell sample is a mosaic with some cells benignly infected with HPV and others exhibiting high grade neoplasia or cancer
  • an assay measuring expression of HPV genes may give equivocal results.
  • the presence of the high grade neoplastic or cancerous cells can be established.
  • the benefit of the disclosed method is that only some of the cell samples assayed (those with either equivocal or severe results) need be tested further.
  • the disclosed method can also be combined with treatment regimes.
  • results in the disclosed assay or method indicative of high grade CIN or cancer suggest that antiviral therapy will be ineffective since these stages of disease are often 25 accompanied by integration of HPV into the genome.
  • assay or method results indicative of normal or low grade stages of disease suggest antiviral treatments since HPV is generally not integrated at these stages.
  • Antiviral treatments include, for example, drugs or therapeutic vaccines. Where results of the disclosed method indicate a benign cell state, treatment can be avoided altogether. The ability to make such assessments reliably and accurately is a significant benefit of the disclosed assay.
  • the disclosed method can include the combination of a molecular assay as described above with any other assay for assessing a disease or state of cells in a cell sample.
  • a molecular assay measuring the level or ratio of expression of HPV genes can be combined with cytological assays, histological assays, determination of the HPV type present, determination of the level of HPV present, assays detecting other cellular markers such as oncoproteins or tumor suppressors, or combinations of these assays.
  • Such assays are known and are used for the diagnosis of HPV infection or HPV-based disease and assessment of the stage of disease.
  • Results from a molecular assay and one or more additional assays can be combined to increase the reliability of any assessment, prognosis, diagnosis, or monitoring of HPV-based disease.
  • This possibility of combination is a particularly useful aspect of the disclosed method since the HPV molecular assays described above provide information about HPV-based disease that is distinct from other assays. Where multiple assays point in the same prognostic or diagnostic direction, the reliability of the assessment is increased.
  • Useful combinations include a cytological assay and the disclosed molecular assay, an assay for determining HPV type and the disclosed molecular assay, and a combination of a cytological assay, a typing assay, an assay for detecting cancer markers, and the disclosed molecular assay.
  • Combined assays can be performed in any order and in any temporal relationship. For example, various assays can be performed in parallel or simultaneously. Such assays can be performed in any manner such as on the same apparatus by the same person, with different apparatus, or in the same or different locations.
  • Cytological assays for use in assessing the stage of HPV-based disease are known and can be used in the disclosed method.
  • the well established Pap smear and Hematoxylin & Eosin stains (H&E) are preferred examples.
  • the use and analysis of Pap smears and H&E stains are well-known in the art.
  • a cell sample as the term is used herein is primarily a collection of cells from a patient.
  • One method of obtaining cells is through non-invasive means, which is defined herein as obtained without the puncturing of a patient.
  • non-invasive means are, for example, cell samples obtained from urine or a nasal, epithelial, cervical or other cell surface scrape.
  • Patient cells can also be obtained by other means including, for example, needle biopsy or tissue biopsy.
  • the cell sample can be preserved in a collection medium which allows for a combination of two or more assays of different characteristics related to a cell state of interest.
  • the assay or assays refer to detection or measurement of specific characteristics, the results of which may be combined with other such measurements of other characteristics to an overall assessment of a cell suspected of being infected with one or more diseases.
  • These assays may include, for example, a combination of morphological analysis and quantitation of a particular RNA or DNA or protein whose levels provide a specific indication of the presence or progression of a disease.
  • the collection medium can be used to combine an assay identifying the morphology of cells in a cell sample with one or more assays identifying the HPV type involved, and, for example, identifying whether the HPV type identified is a high risk or low risk HPV type for the development of HPV-induced cell transformation and cancer.
  • sources of cell samples for assessing HPV-based disease include cervix, vagina, vulva, anus, penis, larynx, buccal cavity, lymph nodes, malignant deposits in any part of the human body, and epidermis; all of which are known sites of HPV infection and pathology.
  • Cell samples for use in the present invention can be collected and stored in liquid medium.
  • useful cell collection media are STM (Digene), PreservCyt® (Cytyc), and CytoRichTM (Autocyte). These media (PreservCyt® and CytoRichTM) were developed for the collection of cytological samples but can be adapted for use with molecular assays.
  • Cell samples for use in the method of the present invention can be fixed or processed in any manner consistent with the assays to be performed.
  • both cytological and molecular assays can be performed using cells fixed on a solid substrate such as, for example, a slide.
  • the requirements of the assays to be performed will generally identify the sample processing to be used.
  • kits that include one or more of the materials needed for the method, such as reagents and sample collection and handling materials.
  • kits can include cell collection medium, sample preserving reagents, reagents for specific detection of DNA and/or expression products (RNA or proteins) of one or more of the E2, E4, E5, E6, E7, L1 or L2 genes, and sample handling containers.
  • Useful reagents for detection of expression of the HPV genes are nucleic acid probes for those genes.
  • a kit may also contain control samples or reagents, or reagents and materials for performing other assays to be combined with the disclosed assay.
  • the kits can contain reagents for the separation of RNA and/or DNA from other cellular components.
  • the present invention can be performed using devices adapted to the method.
  • Numerous devices for performing similar assays are known and in use and can be adapted for use with the disclosed assays and method.
  • devices are known for automating all or a part of sample assays and sample handling in assays.
  • All or part of the disclosed method can be controlled or managed using special purpose computer programs.
  • the data collected from the disclosed method, and data from any other assay used in combination, can be compiled, analyzed, and output in various forms and for various purposes using special purpose computer programs.
  • Such programs can be used with, or combined into, other patient or data management computer programs. The usefulness of such a program increases with the number of measurements or assessments to be combined, and the relative importance of each type of measurement to the overall assessment.
  • Computer programs for use with the disclosed method can be used on general purpose computers, or can be incorporated into special purpose computers or computerized devices for controlling the disclosed method, handling and analyzing data from the disclosed method or both.
  • the assay for nucleic acids follows in general principle the method for detecting HIV RNA by the Digene Hybrid Capture HW Test, described in WO 93/10263 by Digene. Briefly, following lysis, 50 ill of probe mix (containing DNA biotinylated probe) was added to each well. The plate was sealed and incubated at 65° C. for 2 hours for hybridization to occur. After hybridization, samples were transferred to a strepavidin-coated microplate, and 25 iaL of anti-hybrid antibody was added to each well. The plate was agitated at 1100 RPM, for 1 hour, at room temperature. Wells were washed 6 ⁇ times with 65° C. wash buffer, followed by one wash using distilled water.
  • chemiluminescent substrate 100 Ill was added to each well and the plate was incubated at room temperature for 30 minutes. The plate was then read in the DML 2000 luminometer. The data was then expressed as signal-to-noise. Using a calibration curve, the chemiluminescent signal generated by each specimen was converted into mRNA copies per cell.
  • the assay described above can be run on either whole lysed cells or nucleic acid separated from other cellular components.
  • This example illustrates the measurement of HPV E6/E7 expression for use in the disclosed method.
  • a method for detecting and quantitating HPV mRNA, including E6/E7 and mRNA has been developed. This example measures expression in CaSki cells, but the method is generally applicable to other cell lines and clinical specimens.
  • CaSki cells contain an integrated high-risk HPV-16 genome (about 600 copies/cell).
  • CaSki cells were maintained in subconfluence in RMPI 1640 media containing 10% FBS and 10 mM sodium pyruvate.
  • CaSki cells were grown to confluence and were removed from the dishes by treatment with 0.1% trypsin-0.5 mM EDTA. Using trypan blue, viable cells were counted under microscopy.
  • Cells were seeded, in 10 pl volumes, at final concentrations of 10, 10 2 , 10 3 , 10 4 , and 105 cells/well in a polystyrene, tissue culture treated, 96 well plate. Each dilution was performed and seeded in triplicate.
  • the cells were lysed with Proteinase K (30 units) in a Tris-EDTA-buffered SDS).
  • the plate solutions (20 mM Tris pH 7.4, 20 mM EDTA and 0.5′ was sealed, agitated for 30 seconds at 1100 RPM and incubated at 37° C. for 30 minutes.
  • the test for HPV mRNA follows in general principle the method for detecting HIV RNA by the Digene Hybrid Capture HIV Test, described in WO 93/10263 by Digene. Briefly, following lysis, 50 pl of probe mix (containing E6/E7 DNA biotinylated probe) was added to each well. The plate was sealed and incubated at 65° C. for 1.5 hours for hybridization to occur.
  • samples were transferred to a strepavidin-coated microplate, and 25 of anti-hybrid antibody was added to each well.
  • the plate was agitated at 1100 RPM, for 1 hour, at room temperature.
  • Wells were washed 6 ⁇ times with 65° C. wash buffer, followed by one wash using distilled water.
  • 100 ⁇ l of a chemiluminescent substrate was added to each well and the plate was incubated at room temperature for 30 minutes. The plate was then read in the DML 2000 luminometer. The data was then expressed as signal-to-noise. Using a calibration curve, the chemiluminescent signal generated by each specimen was converted into mRNA copies per cell.
  • FIG. 1 The data for the direct detection of HPV mRNA from CaSki cells is shown in FIG. 1 .
  • This method is exemplified by the detection and quantitation of E6/E7 mRNA, but has also been applied to quantitate other mRNA molecules (for example, HPV L1 mRNA) from CaSki cells ( FIG. 2 ).
  • CaSki cell line was trypsinized by incubating with 0.25% Trypsin-EDTA for 5 minutes at 37° C. Cells were pelleted from the suspension by centrifugation at 800 rpm for 3 minutes in Sorvall RT 6000 centrifuge. Cell pellet was resuspended in 500 ⁇ l of 1 ⁇ PBS and counted under microscope Trypan Blue solution. Cells were diluted to 50 and 500 cells/ ⁇ l in 1 ⁇ PBS. 10 ⁇ l of each cell concentration, including zero point (10 ⁇ l of 1 ⁇ PBS) were spiked in 3 ml of PreservCyt reagent. 100 ⁇ l of Sample Conversion Buffer were added into each tub to help visualize the cell pellet.
  • 50 ⁇ l of each E6/E7 RNA calibration were loaded in designated wells at 0, 10 3 , 10 4 , 10 5 , 10 6 , 10 7 and 10 8 molecules/well to construct calibration curve for mRNA in the specimen.
  • 50 ⁇ l of the probe mix was added into each well. Plate was covered with the plate sealer and was agitated for 1 minute at 1100 rpm on the bench top shaker. Samples were incubated at 65° C. for 1.5 hours (hybridization reaction) in the heat block. Plate was transferred into the bench top shaker and was incubated for 1 hour with agitation at 1100 rpm at room temperature (capture reaction). 25 ⁇ l of Detection Reagent 1 was added into each well and plate was incubated without agitation for 1 hour at room temperature.
  • the contents of the plate were discarded and the plate was washed vigorously six times with Wash Buffer at 65° C. and one time with deionized water at room temperature.
  • the plate was drained into Kimtowels and 100 ⁇ l of the Detection Reagent 2 was added into each well. Plate was incubated for 30 minutes at room temperature covered from the light. At the end of incubation time, plate was read on the Digene DML 2000 luminometer and the data were expressed as signal-to-noise.
  • HaCaT cells were infected with HPV 16 by the procedure of White et. al. (White (1998)) to produce an episomal (non-integrated, total sequence, not spliced) HPV infection. Approximately 1 copy of HPV16 was present for every 40 cells. These cells are considered a representative of early stage infection or CIN I (cervical intraepithelial neoplasia). W12 cells contain approximately 100 copies of episomal HPV 16 DNA and represent pre-malignant, immortalized cells or CIN II or CIN III. SiHa cells contain 1-2 copies of HPV 16 integrated into the genome. These cells are considered to represent cancer.
  • RNA analysis was done according to Example 1 or the following procedure. Single stranded, biotinylated, DNA probes containing the specific HPV16 gene sequences were prepared. For HaCaT and SiHa cell lines, cells were grown to confluency, cells were harvested, and the total RNA was purified using the RNeasy kit (Qiagen Inc., Santa Clarita, Calif.). For W12, whole cells were used for analysis. RNA calibrators containing the complete HPV genome were prepared by transcribing (+) sense RNA from a plasmid containing the complete HPV16 genome with T7 RNA polymerase. The RNA was then diluted to 10 3 , 10 4 , 10 5 , 10 6 , and 10 7 copies per 50 pl.
  • RNA:DNA hybrids were captured onto the streptavidin coated plate and were simultaneously reacted with the antihybrid antibody conjugate. After several wash steps, a chemiluminescent substrate (Tropix CDP-star with Emerald) was added to the wells, and the light output was measured in a microplate luminometer after 30 minutes incubation at room temperature.
  • Probe mix containing the biotinylated, single-stranded DNA probe
  • the quantitation of HPV mRNA was performed as follows. The results from the RNA calibrators were used to construct standard curves. The regression equation was calculated from the logarithm of the copies versus the logarithm of signal to noise minus one [(S/N) ⁇ 1)]. The regression equations were then used to calculate the number of copies of mRNA in the cellular RNA samples.

Abstract

This invention provides novel methods for assessing HPV infection. Gene expression levels are used to assess the progression of HPV infection from benign to malignant growth. Also provided are kits for carrying out the methods of this invention.

Description

  • This application is a continuation application of U.S. patent application Ser. No. 11/681,539, filed Mar. 2, 2007, which is a continuation of Ser. No. 09/970,477, filed Oct. 4, 2001, which is a continuation of Ser. No. 09/210,168, filed Dec. 11, 1998, issued as Pat. No. 6,355,424 on Feb. 21, 2002, to which priority under 35 U.S.C. §120 is claimed. This application also claims benefit of U.S. patent application Ser. Nos. 60/082,167, filed Apr. 17, 1998, 60/070,486, filed Jan. 5, 1998, and 60/069,426, filed Dec. 12, 1997.
  • FIELD OF THE INVENTION
  • The present invention is generally related to the field of cytological and 10 molecular assays and specifically to the area of assays for the assessment of disease using a sensitive assay for diagnosis and prognosis of HPV-induced carcinoma.
  • BACKGROUND OF THE INVENTION
  • The detection and diagnosis of disease is of obvious importance for the treatment of disease. Numerous characteristics of diseases have been identified and many are used for the diagnosis of disease. Many diseases are preceded by, and are characterized by, changes in the state of the affected cells. Changes can include the expression of viral genes in infected cells, changes in the expression patterns of genes in affected cells, and changes in cell morphology. The detection, diagnosis, and monitoring of diseases can be aided by the assessment of such cell states.
  • An aspect of the present invention relates to human papilloma virus (HPV), which induces benign epithelial proliferations of the skin and mucosa in humans and is associated with anogenital neoplasias and carcinomas. The intact DNA of HPV is supercoiled and thus resembles an endless loop of twisted telephone handset cord. Inside this shell, the viral DNA is packaged in and around proteins from the cell nucleus, histones, and associated peptides, into a structure that resembles cellular chromatin. (Turek, (1994)). Human papillomaviruses characterized to date are associated with lesions confined to the epithelial layers of skin, or oral, pharyngeal, respiratory, and, most importantly, anogenital mucosae. Specific human papillomavirus types, including HPV 6 and 11, frequently cause benign mucosal lesions, whereas other types, HPV 16, 18, and a host of other strains, are predominantly found in high-grade lesions and cancer. All human and animal papillomaviruses appear to share a similar genetic organization, although there are differences in the functions of individual viral genes and in their regulation. The most common genital HPV type associated with cervical carcinoma, HPV 16, has been studied most extensively.
  • All large open reading frames (ORFs) in HPV are on one DNA strand. Papillomaviral mRNAs appear to be transcribed solely from a single strand in infected cells. The viral genome can be divided into three regions, the upstream regulatory region (URR), or long control region (LCR), containing control sequences for HPV replication and gene expression, the viral early gene region, encoding, among others, the E2, E6 and E7 genes, and the late region, encoding the L1 and L2 genes. (Turek, (1994)).
  • HPV gene expression in high-grade premalignant disease or cancer appears restricted to the early genes, possibly due to cellular differentiation arrest induced by the viral E6 and E7 genes. In comparison to active HPV infection, E6 and E7 gene control in cancer is deranged by mutations in the viral URR and, in integrated viral fragments, by the disruption of the viral E2 gene, stabilization of E6 and E7 mRNAs, and influences at the cellular integration site.
  • Because the E2 gene is disrupted or inactivated in integrated HPV fragments in invasive cervical carcinomas (Cullen, (1991); Diirst, (1985); Matsukura, (1989); Schneider-Gadicke, (1986); Schwarz, (1985); Wilczynski, (1988)), it has been predicted that loss of E2 bestows a selective growth advantage to the infected cell because of uncontrolled E6 and E7 expression (Schneider-Gadicke, (1986); Schwarz, (1985)). Indeed, cervical cells containing replicating HPV genomes rapidly segregate and are outgrown in culture by cells that contain integrated viral genomes (Jeon (1995)), but the underlying mechanism(s) have remained unclear until recently. The full-length HPV 16 E2 gene products are strong transcriptional activators comparable to HPV 1 E2 at some viral as well as at simple, synthetic promoters (Demeret (1994); Ushikai (1994)).
  • Genes E6 and E7 are considered to have oncogenic activity. The encoded proteins interact with and disturb the physiologic functions of cellular proteins that are involved in cell cycle control. The E6/E7 proteins of HPV 16, 18 or related types are most efficient in this regard. Some of these activities lead to genetic instability of the persistently infected human cell. This enhances the probability of mutations in cellular proto-oncogenes and tumor suppressor genes and thus contributes to tumor progression. Mutations in cellular genes devoted to the intracellular surveillance of HPV infections, integration of viral DNA, and deletions or mutations of viral transcription control sequences lead to a significantly increased expression of the E6/E7 genes, which is a consistent characteristic of high-grade intraepithelial neoplasia and cancers. The genetic instability caused by viral oncoproteins and the autocatalytic increase in oncoprotein expression caused by mutations in the viral and cellular genome identify the virus as a major driving force of progression to carcinoma.
  • Individual types of human papillomaviruses (HPV) which infect mucosal surfaces have been implicated as the causative agents for carcinomas of the cervix, anus, penis, larynx and the buccal cavity, occasional periungal carcinomas, as well as benign anogenital warts. The identification of particular HPV types is used for identifying patients with premalignant lesions who are at risk of progression to malignancy. Although visible anogenital lesions are present in some persons infected with human papillomavirus, the majority of individuals with HPV genital tract infection do not have clinically apparent disease, but analysis of cytomorphological traits present in cervical smears can be used to detect HPV infection. Conventional viral detection assays, including serologic assays and growth in cell culture, are not commercially available and/or are not suitable for the diagnosis and tracking of HPV infection. Papanicolaou tests are a valuable screening tool, but they miss a large proportion of HPV-infected persons.
  • Thus, it is an object of the present invention to provide a method for assessing the stage of HPV-based disease.
  • It is another object of the present invention to provide an assay that can be combined with other assays to improve the accuracy and reliability of prognostic and diagnostic assessments of HPV-based disease.
  • It is a further object of the present invention to provide a method for assessing the risk that a patient infected with HPV will develop HPV-based disease.
  • It is another object of the present invention to provide a method for stratifying patients who are currently HPV-infected but without detectable HPV-based disease into those at risk for progression to disease and those not at risk for progression to disease.
  • It is also an object of the present invention to provide a method for identifying treatment regimes for patients having HPV-based disease.
  • Yet another object of the present invention to provide a method for monitoring the effectiveness of treatment of HPV-based disease.
  • A further object of the present invention to provide kits for assessing the stage of HPV-based disease.
  • Another object of the present invention to provide computer-based operation, analysis, and data management of assay data to assess the stage of HPV-based disease.
  • SUMMARY OF THE INVENTION
  • One embodiment of the present invention involves measuring the levels of expression of genes involved in a cell state, and comparing their expression to each other or to reference genes in a specific ratio, as an indication of the state of a disease in the cell sample. The present invention can be used to assess the stage or risk of a disease as indicated by the state of the cells. It can also be used to guide or assess the effectiveness of a therapy for a disease by identifying appropriate therapy based on the indicated cell state or by indicating any change in the state of cells subjected to the therapy.
  • In one form of the present invention, the stage and prognosis of a human papillomavirus (HPV) infection or HPV-based disease is assessed. This embodiment involves the measurement of the level of expression of two or more HPV genes. Genes for this purpose are the HPV E6, E7, L1, and E2 genes, although other HPV genes such as E1, E4, E5, and L2 can also be used. It has been discovered that the level of expression of these genes, the ratio of expression of these genes to each other or to reference genes, or both, are indicative of the stage of HPV-based disease.
  • Gene expression levels are used according to this invention to assess the progression of HPV infection from benign to malignant growth. HPV infection progresses from CIN I through CIN TTT and finally to malignant cancer. These stages can be identified by the ratios of HPV genes. In particular, the transition from CIN I to CIN II/III, i.e., a transition to pre-malignancy, can be predicted when the ratio of the present invention exceeds one. A ratio of greater than 3 in the present invention indicates a transition from pre-malignancy to malignant cancer. Thus, a ratio below 1 indicates a low-level of CIN in an HPV infected cell. A ratio between one and about three indicates a high grade CIN (i.e., CIN III) or pre-malignant condition. And a ratio of over about three is an indication of an HPV induced malignancy.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is a graph of detection of E6/E7 mRNA shown as the number of cells/well versus the number of E6/E7 mRNA/well.
  • FIG. 2 is a graph of the sensitivity of an HPV L1 assay shown as the number of RNA molecules/well versus the signal-to-noise ratio minus 1.
  • FIG. 3 is the probe sequence for HPV 16 L2 (SEQ ID NO: 2).
  • FIG. 4 is the probe sequence for HPV 16 L1 (SEQ ID NO: 2).
  • FIG. 5 is the probe sequence for HPV 16 E6/E7 (SEQ ID NO: 3).
  • FIG. 6 is the probe sequence for HPV 16 E2 (SEQ ID NO: 4).
  • FIG. 7 is the probe sequence for HPV 16 E4 (SEQ ID NO: 5).
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention relates to the identification and monitoring of diseased cells. One method involves measuring the levels of expression of genes involved in a disease state, and comparing their expression to each other or to reference genes, as an indication of the state of the cells. Such measurements can be combined with other assays to increase the accuracy and reliability of the assessment of the disease state. One method of the present invention can be used to assess the stage of a disease as indicated by the state of the cells. This method can also be used to guide or assess the effectiveness of a therapy for a disease by identifying appropriate therapy based on the indicated disease state or by indicating any change in the state of cells subjected to the therapy.
  • Many diseases are characterized by specific cellular phenotypes and gene expression patterns. For example, neoplastic and cancerous cells generally exhibit certain distinctive morphologies and growth characteristics. Molecular characteristics, such as gene mutations and gene expression patterns are also a good indicator of disease progression. Virally infected cells can exhibit different morphologies and gene expression patterns, including expression of viral genes. Using the present invention, the characteristics of the cell state, such as changes in cell morphology or expression of genes can be determined from a patient sample.
  • The characteristics to be detected are generally specific to the cell state of interest and the disease suspected of being present in the cell sample. Such characteristics can be generally divided into two types, cytological characteristics and molecular characteristics. As used herein, cytological characteristics are characteristics such as, for example, overall cell shape and appearance. The primary identification and classification of many neoplastic and cancerous cells has traditionally been accomplished using cytological characteristics. Identification of cytological characteristics is generally slow, requires a relatively high level of training, and generally cannot be easily automated. As used herein, molecular characteristics are the presence and state of particular molecular species, such as proteins, nucleic acids, and metabolites. Such molecular characteristics are generally identified by detecting and measuring the particular molecules of interest.
  • The characteristics assayed can include additional or surrogate marker characteristics that are not a direct cause or result of the disease but that are related to certain disease and cell states. Examples of such additional markers include polymorphic markers, human leukocyte antigens (HLA) such as B7 that predispose women for cervical carcinomas, oncogenes, p53 mutations, other cancer markers, oncosupressors, cytokines, growth factor receptors, and hormones. Such markers can be present in, or absent from, cells exhibiting state-or disease-specific characteristics, and such presence or absence can be indicative of for example, a more severe or less severe disease state. These markers can be used in conjunction with the disclosed method to infer either higher or lower risk of neoplastic disease depending on the number of abnormal scores or the magnitude of change in quantitative markers.
  • Examples of disease states for assessment using the present invention include, but are not limited to neoplasias and cancer. Disease states of interest are HPV-based disease—including HPV infection, cervical intraepithelial neoplasia (CIN), and cancer, atypical squamous cells of undetermined significance (ASCUS), warts, condylomata, epidemic) dysplasia verruciformis and other skin diseases, laryngeal papilloma, oral papilloma and conjunctival papilloma.
  • An embodiment of the present invention is detection and measurement of the expression levels of certain HPV genes. An impressive amount of data has been accumulated over the past decade, showing that carcinoma of the cervix is associated with infection of certain types of HPV. Though the presence of HPV DNA in a precancerous lesion is indicative of an increased relative risk for cervical dysplasia and invasive carcinoma, it is still difficult to predict the clinical behavior of precancerous cervical lesions. Tumors arise due to the accumulation of genetic alterations which can activate oncogenes and/or inactivate tumor suppressor genes and/or genes involved in DNA damage recognition and repair.
  • The expression levels of E6 and E7 oncoproteins encoded by high-risk HPV types are a more sensitive and accurate measure of potential risk of an HPV infection developing into a cancerous lesion. The present invention measures the relative amounts of E6 and/or E7 expression levels and E2 and/or L1 in an HPV-infected lesion to determine the ratio of E6 and/or E7 to L1 and/or E2, where in this ratio is a direct measure of risk, and susceptibility to the development of a cancerous lesion. HPV expression can be measured by mRNA or protein levels in the cell.
  • In one aspect of the invention, the stage and prognosis of a human papillomavirus (HPV) infection or HPV-based disease is assessed. This embodiment of the present invention involves the measurement of the level of expression of one or more HPV genes discovered to be related to the stage and nature of HPV-based disease. Genes 25 useful for this purpose include the HPV E6, E7, L1, and E2 genes. It has been discovered that the level of expression of these genes, the ratio of expression of these genes to each other or to one or more other genes, or both, are indicative of the stage of HPV-based disease. The level of expression is relative to other HPV genes, or the level of expression relative to a non-HPV gene, referred to herein as a reference gene. Such reference genes can be any appropriate gene (not encoded by HPV), and are, for example, housekeeping genes or other constitutively expressed genes. Examples of reference genes include actin genes, cytoskeletal genes, histone genes, tubulin genes, epidermal growth factor receptor genes, the normal p53 gene, the normal pRB gene, cyclin genes, 13-globin genes, and glucose-6-phosphate dehydrogenase genes. Expression of reference genes can be measured in the same cell as the level of HPV genes are measured or in neighboring cells in the same cell sample. In such a case, the reference gene is an internal control for gene expression.
  • The relationship of the relative level of expression of these genes to the state of cells infected with HPV is generally illustrated in Table 1 below.
  • TABLE 1
    Medical HPV-Based disease E6, E7,
    Character state E6 + E7 E2 Ll
    benign HPV-infected low low to high low to
    normal tissue undetectable
    benign Low grade ClN low to low to high medium to
    i.e., CIN I medium high
    neoplastic High grade CIN medium to low to low to
    i.e., CIN HMI high undetectable undetectable
    neoplastic Cancer medium to low to low to
    high undetectable undetectable
  • The HPV-based high grade CIN referenced in the table is the premalignant stage leading to cancer and low grade CIN is a productive viral infection that has little malignant potential but is a public health concern with respect to the spread of HPV infection. Normal tissue refers to cytologically normal tissue that is infected with HPV. Although not limited to this standard, one standard for establishing this lower limit of expression is a level below that detectable using the hybrid capture assay described in WO 93/10263 by Digene. As used herein, E6/E7 refers to E6, E7, or E6 +E7. The addition of genes, as with “E6 +E7,” for example, refers to the combined expression of the added genes.
  • As can be readily discerned, each major disease state is represented by a unique expression pattern of these three sets of genes. Both of these conditions are regarded as serious medical aliments. Other relationships involving the relative level 15 expression of other HPV genes (such as E1, E4, E5, and L2), and other, non-HPV genes, can also be used to assess cell state. For example, L2 and E4 are frequently associated with benign viral production diseases, and E1 is similar in profile to E2 and is often deleted in malignancies. Other relationships of expression for these HPV proteins can exist for other HPV-based diseases, and the disclosed method can be used to assess the state of such other diseases using the appropriate levels and ratios for that disease.
  • Using information about the levels and ratios of HPV genes in different cell states, the stage of the disease can be assessed in several ways. In some cases, the presence or absence of detectable expression is indicative of the disease state in the infected cells. For example, a lack of E2 expression (when HPV is present) is indicative of high grade CIN or cancer. In other cases, a change or difference in expression of an HPV gene product can be indicative of change occurring in the infected cell state. For example, an increased level of expression of E6 and E7—relative to, for example, an earlier sample or a reference sample—may be indicative of high grade CIN or cancer. A change in the ratio of E6/E7 expression to E2 expression is used to identify low grade CIN or a shift from normal cells to low grade CIN. Many other combinations of comparisons are also possible, and other combinations can be derived from the information in Table 1 for use in the method of the present invention.
  • One way in which ratios of HPV genes can be related to HPV-based disease states is by reference to groups of HPV genes. For this purpose, group 1 genes or gene sets include E6, E7, and E6+E7. Group 2 genes or gene sets include L1, L2, E4, and any combinations. Group 3 genes or gene sets include E1, E2, E5, and any combinations. Useful ratios of expression include ratios of members of group 1 to members of group 2 or group 3. Examples of theses ratios are (E6+E7)/(L1+L2), E6/L1, E7/L1, E6/L2, E7/L2, (E6+E7)/L1, (E6+E7)/L2), E6/(L1+L2), and E7/(L1+L2). For such ratios, a value of less than two is indicative of benign human papillomavirus infection or low grade intra-epithelial neoplasia. This type of infection is also classified as CIN I. HPV infections which progress beyond CIN I indicate cell transformation has occurred and cancerous growth has begun. These later infections (CIN FM) have ratios of greater than 2. A ratio of expression of more than two is indicative of high grade intra-epithelial neoplasia or pre-malignant cancer. A ratio of expression of much more than two (i.e., exceeding 4 and up to infinity) is indicative of cancer. Preferred ratios for use in this invention are (E6+E7)/L1, (E6+E7)/(L1+L2), (E6+E7+E2+E4)/(L1+L2), (E6+E7)/(E2+E4), (E6+E7)/E2, (E6+E7+E2-E4)/(L1+L2).
  • There are several ways in which measured levels of expression of HPV genes can be compared and categorized. For example, where the presence or absence of expression is indicative of the cell state, expression of the HPV gene is analyzed without reference to the expression level of other genes. Where the relative level of expression of an HPV gene is indicative of the cell state, the measured level of expression is compared, for example, to the level of expression of the same type of HPV gene in a different cell sample (such as an earlier cell sample from the same source or reference cells harboring HPV), to the level of expression of a different type of HPV gene in the same or a different cell sample, to the level of expression of a non-HPV reference gene in the same cell sample, or to the level of expression of a non-HPV reference gene in reference cells.
  • In one embodiment of the present invention, levels and ratios of expression of HPV genes are compared to the levels of the same genes in a cell line that contains HPV (such as HeLa or CaSki). Such cell lines provide a standard against which levels of expression of HPV genes in cell samples are compared. Such comparisons are used to assess and compare the absolute levels of expression of these HPV proteins with those in a standard or comparative cell line. Other cell lines useful for this purpose are non-cancerous cell lines infected with HPV 16 (such as W12) or HPV 31 (such as ON612; Meyers (1992)).
  • The levels and ratios of expression of HPV genes are also compared to reference genes, such as housekeeping genes or other constitutively expressed genes, in the same cells or in reference cells, such as a cell line. For example, the level of expression of the HPV gene and the reference gene is measured in the same cell sample. Such measurements provide an internal control of the overall expression level in a cell sample and are used to calculate a corrected level of expression for the HPV gene to allow more accurate comparisons of the level of expression between different cell samples. One form of correction is referred to as normalization. Thus, the level of expression of one or more HPV genes can be measured in two or more cell samples along with the level of expression of the same reference gene in each of the cell samples. The level of expression of the HPV genes is then normalized to each other based on differences, if any, between the measured level of expression of the reference gene in each of the samples.
  • In some stages of HPV-based disease, the expression of a particular HPV gene is low or undetectable. Such lack of detectable expression is used herein to identify patterns of expression that serve as prognostic or diagnostic indicators. The expression of other HPV genes is assessed in parallel or in the same assay to provide a control for the lack of expression of one or more of the genes to be assessed. This is accomplished by measuring the expression of the several HPV genes together or in parallel. For example, measuring the level of expression of the HPV E6, E7, L1, E4, and E2 genes in parallel is useful since at least one of these genes is expressed in all of the stages of HPV-based disease. In a preferred embodiment of the present invention, a ratio of E6/E7 to L1 /E2 provides an indicator of likelihood of developing cancer from the HPV infection. In this way, indicative information is collected as well as providing an internal control. The presence of L1, L2, and E4 in combination is also indicative of benign disease while the absence of E1 and/or E2 is indicative of neoplastic potential.
  • The types of comparison described above can also be used with other genes and other disease states. That is, the measured level of expression of a gene of interest can be compared, for example, to the level of expression of the same type of gene in a different cell sample (such as an earlier cell sample from the same source or appropriate reference cells), to the level of expression of a different type of gene in the same or a different cell sample, to the level of expression of a reference gene in the same cell sample, or to the level of expression of a reference gene in reference cells.
  • Expression of genes of interest, such as the HPV E6, E7, L1, E4, and E2 genes, can be assessed using any suitable method. For example, RNA can be detected using hybridization, amplification, or sequencing techniques, and protein can be detected using specific antibodies. Many techniques for the specific detection of gene expression, by detection of expression products, are known and can be used with the disclosed method. One technique for detecting and measuring the level of expression of genes of interest is detection of RNA transcribed from the genes of interest. For the most reliable comparisons, expression levels that are to be compared should be measured using the same technique and be performed in the same manner.
  • For hybridization detection of HPV nucleic acids, a mixture of probes specific for these sequences from different HPV types can be used. This ensures that the method will detect expression regardless of the type of HPV involved. For some purposes, it may be desirable to use probes designed for the sequence of a certain HPV type, or a mixture of probes for only some HPV types. Such probes may or may not be type-specific depending on the differences between the sequences of the HPV nucleic acids to be detected. One useful mixture for this purpose would include probes for HPV types more closely associated with a progression to cancer. The HPV types most commonly associated with cervical cancer are types 16 and 18.
  • Useful techniques for measuring the level of expression of a gene of interest in a cell sample include the hybrid capture technique described in WO 93/10263 by Digene, PCR in situ hybridization techniques described by (Nuovo, 1997)), branched DNA assays (Chernoff (1997)), transcription-mediated amplification (TMA); Stoflet (1988)), and polymerase chain reaction (PCR), ligase chain reaction (LCR), self-sustained sequence replication (3SR), nucleic acid sequence based amplification (NASBA), strand displacement amplification (SDA), and amplification with QB replicase (Birkenmeyer and Mushahwar, (1991); Landegren, (1993)).
  • Numerous assays for the detection and measurement of gene expression products are known and can be adapted for the determination of the level of expression of genes of interest in the disclosed assay. For example, many of the techniques for the detection of HPV in general or expression of other HPV genes described below can also be adapted for use in the disclosed assay for the detection of expression of HPV genes E6, E7, L1, E4,and E2.
  • Many HPV detection and typing assays, which can be used in the disclosed method, are known, including assays involving Southern blots, dot blots, in situ hybridization, polymerase chain reaction, and solution hybridization. Mant (1997) describes PCR assays used to identify DNA from specific HPV types. Cope (1997) describes a PCR-based test using a consensus primer and a Hybrid Capture assay (HCA) of detection of HPV types. The hybrid capture assay is also described in WO 93/10263 by Digene. The hybrid capture assay is a useful method for detection of HPV and for determining HPV type in combination with the disclosed assay.
  • Swan (1997), discloses an HPV detection assay exploiting the 5′ to 3′ exonucleolytic activity of Taq DNA polymerase to increase the signal from fluorescent dyes by releasing them from genotype-specific probes during PCR. Lizardi (1997), describes a method of detecting HPV using in situ hybridization with non-radioactive probes and visualization with conventional bright-field or fluorescence microscopy, or laser scanning confocal microscopy. Zehbe [1] (1997), describes a modified version of in situ hybridization for detection of HPV that involves signal amplification. Leiserowitz (1997), describes use of reverse transcriptase-polymerase chain reaction to detect HPV. Zehbe [2] (1997), describes a nonisotopic, enzyme-linked immunosorbent assay-based sandwich capture hybridization assay for HPV detection.
  • In one embodiment of the present invention RNA was analyzed directly by solution based procedures. The cells were first lysed by adding a proteolytic enzyme to the cells contained in wells of a microtiter plate. Non-limiting examples of enzymes for use in the present invention include proteinase K or Pronase. Cells can also be subjected to detergent lysis or osmotic lysis or a French Press. After incubation, biotinylated DNA probes were added to each well. The RNA:DNA hybrids were captured onto a solid phase by transferring to streptavidin coated microplates. Alkaline phosphataseconjugated antibodies to RNA:DNA hybrids were added to each well in the hybridization microplate and signals were generated by adding a chemiluminescent reagent such as CDP-Star® with Emerald II (Tropix) to each well. The signal was read from the microplate. The solution based DNA analysis was performed similarly to the RNA analysis except that the microtiter plates were coated with anti-RNA:DNA hybrid antibodies and the probes were RNA probes.
  • Other methods for detection and assessment of HPV infections that can be used in the disclosed method are described in U.S. Pat. Nos. 5,415,995, 4,777,239, 5,484,699, 4,983,728, 5,527,898, 5,364,758, 5,639,871, 5,501,947, 5,665,533, 4,748,109, 5,623,932, 5,665,571 and 5,648,459. These are just examples of HPV detection and typing assays that may be used in combination with the disclosed molecular assay. Many of the techniques described above can also be adapted for use in the disclosed assay for the detection of expression of HPV genes.
  • The disclosed assay and other assays can also be sequentially combined. That is, first one type of assay can be performed, and then, depending on the results, another assay can be performed. For example, an assay to detect HPV (or HPV type) can be performed first, then, if HPV is present, the disclosed molecular assay can be 25 performed. As another example, the disclosed molecular assay can be performed first, and if the results of the assay were indicative of a high grade CIN or cancer, a cytological assay or biopsy can be performed. Such sequential combinations are particularly useful for limiting more extensive testing to patients and samples that are identified as high risk.
  • Useful sequential orders for the assays are (1) an HPV assay, followed by an assay for one or more other markers, followed by a cytological or histological assay; (2) a cytological or histological assay, followed by an HPV assay or an assay for one or more other markers, followed by an HPV assay or an assay for one or more other markers (whichever had not been performed first); (3) a cytological or histological assay, followed by a combined or simultaneous HPV and marker assay; (4) a combined or simultaneous HPV and marker assay, followed by a cytological or histological assay; and (5) detection of HPV, detection of HPV type, an HPV assay, and a cytological or histological assay. Each combination of assays is followed by an assessment, using the combined assay results, of the cell state, disease state, patient status, patient prognosis, or other assessment as described herein.
  • Where the results of initial assays are either equivocal or suggest a more severe stage of disease, further assays are useful to clarify and confirm the initial results. For example, where a cell sample is a mosaic with some cells benignly infected with HPV and others exhibiting high grade neoplasia or cancer, an assay measuring expression of HPV genes may give equivocal results. By following up with a morphological assay, the presence of the high grade neoplastic or cancerous cells can be established. In this case, the benefit of the disclosed method is that only some of the cell samples assayed (those with either equivocal or severe results) need be tested further.
  • The disclosed method can also be combined with treatment regimes. For example, results in the disclosed assay or method indicative of high grade CIN or cancer suggest that antiviral therapy will be ineffective since these stages of disease are often 25 accompanied by integration of HPV into the genome. On the other hand, assay or method results indicative of normal or low grade stages of disease suggest antiviral treatments since HPV is generally not integrated at these stages. Antiviral treatments include, for example, drugs or therapeutic vaccines. Where results of the disclosed method indicate a benign cell state, treatment can be avoided altogether. The ability to make such assessments reliably and accurately is a significant benefit of the disclosed assay.
  • The disclosed method can include the combination of a molecular assay as described above with any other assay for assessing a disease or state of cells in a cell sample. For example, a molecular assay measuring the level or ratio of expression of HPV genes can be combined with cytological assays, histological assays, determination of the HPV type present, determination of the level of HPV present, assays detecting other cellular markers such as oncoproteins or tumor suppressors, or combinations of these assays. Such assays are known and are used for the diagnosis of HPV infection or HPV-based disease and assessment of the stage of disease. Results from a molecular assay and one or more additional assays can be combined to increase the reliability of any assessment, prognosis, diagnosis, or monitoring of HPV-based disease. This possibility of combination is a particularly useful aspect of the disclosed method since the HPV molecular assays described above provide information about HPV-based disease that is distinct from other assays. Where multiple assays point in the same prognostic or diagnostic direction, the reliability of the assessment is increased. Useful combinations include a cytological assay and the disclosed molecular assay, an assay for determining HPV type and the disclosed molecular assay, and a combination of a cytological assay, a typing assay, an assay for detecting cancer markers, and the disclosed molecular assay. Combined assays can be performed in any order and in any temporal relationship. For example, various assays can be performed in parallel or simultaneously. Such assays can be performed in any manner such as on the same apparatus by the same person, with different apparatus, or in the same or different locations.
  • Cytological assays for use in assessing the stage of HPV-based disease are known and can be used in the disclosed method. The well established Pap smear and Hematoxylin & Eosin stains (H&E) are preferred examples. The use and analysis of Pap smears and H&E stains are well-known in the art.
  • A cell sample as the term is used herein is primarily a collection of cells from a patient. One method of obtaining cells is through non-invasive means, which is defined herein as obtained without the puncturing of a patient. Examples of non-invasive means are, for example, cell samples obtained from urine or a nasal, epithelial, cervical or other cell surface scrape. Patient cells can also be obtained by other means including, for example, needle biopsy or tissue biopsy.
  • The cell sample can be preserved in a collection medium which allows for a combination of two or more assays of different characteristics related to a cell state of interest. As used herein, the assay or assays refer to detection or measurement of specific characteristics, the results of which may be combined with other such measurements of other characteristics to an overall assessment of a cell suspected of being infected with one or more diseases. These assays may include, for example, a combination of morphological analysis and quantitation of a particular RNA or DNA or protein whose levels provide a specific indication of the presence or progression of a disease. Alternatively, for example, the collection medium can be used to combine an assay identifying the morphology of cells in a cell sample with one or more assays identifying the HPV type involved, and, for example, identifying whether the HPV type identified is a high risk or low risk HPV type for the development of HPV-induced cell transformation and cancer.
  • For example, sources of cell samples for assessing HPV-based disease include cervix, vagina, vulva, anus, penis, larynx, buccal cavity, lymph nodes, malignant deposits in any part of the human body, and epidermis; all of which are known sites of HPV infection and pathology.
  • Cell samples for use in the present invention can be collected and stored in liquid medium. Examples of useful cell collection media are STM (Digene), PreservCyt® (Cytyc), and CytoRich™ (Autocyte). These media (PreservCyt® and CytoRich™) were developed for the collection of cytological samples but can be adapted for use with molecular assays.
  • Cell samples for use in the method of the present invention can be fixed or processed in any manner consistent with the assays to be performed. For example, both cytological and molecular assays can be performed using cells fixed on a solid substrate such as, for example, a slide. The requirements of the assays to be performed will generally identify the sample processing to be used.
  • The present invention can be conveniently performed using kits that include one or more of the materials needed for the method, such as reagents and sample collection and handling materials. For example, kits can include cell collection medium, sample preserving reagents, reagents for specific detection of DNA and/or expression products (RNA or proteins) of one or more of the E2, E4, E5, E6, E7, L1 or L2 genes, and sample handling containers. Useful reagents for detection of expression of the HPV genes are nucleic acid probes for those genes. A kit may also contain control samples or reagents, or reagents and materials for performing other assays to be combined with the disclosed assay. In addition, the kits can contain reagents for the separation of RNA and/or DNA from other cellular components.
  • The present invention can be performed using devices adapted to the method. Numerous devices for performing similar assays are known and in use and can be adapted for use with the disclosed assays and method. For example, devices are known for automating all or a part of sample assays and sample handling in assays.
  • All or part of the disclosed method can be controlled or managed using special purpose computer programs. The data collected from the disclosed method, and data from any other assay used in combination, can be compiled, analyzed, and output in various forms and for various purposes using special purpose computer programs. Such programs can be used with, or combined into, other patient or data management computer programs. The usefulness of such a program increases with the number of measurements or assessments to be combined, and the relative importance of each type of measurement to the overall assessment. Computer programs for use with the disclosed method can be used on general purpose computers, or can be incorporated into special purpose computers or computerized devices for controlling the disclosed method, handling and analyzing data from the disclosed method or both.
  • EXAMPLES
  • The examples herein are meant to exemplify the various aspects of 10 carrying out the invention and are not intended to limit the invention in any way.
  • Example 1 General Methods for Nucleic Acid Analysis
  • The assay for nucleic acids follows in general principle the method for detecting HIV RNA by the Digene Hybrid Capture HW Test, described in WO 93/10263 by Digene. Briefly, following lysis, 50 ill of probe mix (containing DNA biotinylated probe) was added to each well. The plate was sealed and incubated at 65° C. for 2 hours for hybridization to occur. After hybridization, samples were transferred to a strepavidin-coated microplate, and 25 iaL of anti-hybrid antibody was added to each well. The plate was agitated at 1100 RPM, for 1 hour, at room temperature. Wells were washed 6× times with 65° C. wash buffer, followed by one wash using distilled water. 100 Ill of a chemiluminescent substrate was added to each well and the plate was incubated at room temperature for 30 minutes. The plate was then read in the DML 2000 luminometer. The data was then expressed as signal-to-noise. Using a calibration curve, the chemiluminescent signal generated by each specimen was converted into mRNA copies per cell. The assay described above can be run on either whole lysed cells or nucleic acid separated from other cellular components.
  • Example 2 Quantitation of HPV
  • This example illustrates the measurement of HPV E6/E7 expression for use in the disclosed method. A method for detecting and quantitating HPV mRNA, including E6/E7 and mRNA has been developed. This example measures expression in CaSki cells, but the method is generally applicable to other cell lines and clinical specimens. CaSki cells contain an integrated high-risk HPV-16 genome (about 600 copies/cell). CaSki cells were maintained in subconfluence in RMPI 1640 media containing 10% FBS and 10 mM sodium pyruvate. For this example, CaSki cells were grown to confluence and were removed from the dishes by treatment with 0.1% trypsin-0.5 mM EDTA. Using trypan blue, viable cells were counted under microscopy. Cells were seeded, in 10 pl volumes, at final concentrations of 10, 102, 103, 104, and 105 cells/well in a polystyrene, tissue culture treated, 96 well plate. Each dilution was performed and seeded in triplicate.
  • The cells were lysed with Proteinase K (30 units) in a Tris-EDTA-buffered SDS). The plate solutions (20 mM Tris pH 7.4, 20 mM EDTA and 0.5′ was sealed, agitated for 30 seconds at 1100 RPM and incubated at 37° C. for 30 minutes. The test for HPV mRNA follows in general principle the method for detecting HIV RNA by the Digene Hybrid Capture HIV Test, described in WO 93/10263 by Digene. Briefly, following lysis, 50 pl of probe mix (containing E6/E7 DNA biotinylated probe) was added to each well. The plate was sealed and incubated at 65° C. for 1.5 hours for hybridization to occur. After hybridization, samples were transferred to a strepavidin-coated microplate, and 25 of anti-hybrid antibody was added to each well. The plate was agitated at 1100 RPM, for 1 hour, at room temperature. Wells were washed 6× times with 65° C. wash buffer, followed by one wash using distilled water. 100 μl of a chemiluminescent substrate was added to each well and the plate was incubated at room temperature for 30 minutes. The plate was then read in the DML 2000 luminometer. The data was then expressed as signal-to-noise. Using a calibration curve, the chemiluminescent signal generated by each specimen was converted into mRNA copies per cell. The data for the direct detection of HPV mRNA from CaSki cells is shown in FIG. 1. This method is exemplified by the detection and quantitation of E6/E7 mRNA, but has also been applied to quantitate other mRNA molecules (for example, HPV L1 mRNA) from CaSki cells (FIG. 2).
  • Example 3 Quantitation of HPV mRNA Using Preservative Collection Medium
  • CaSki cell line was trypsinized by incubating with 0.25% Trypsin-EDTA for 5 minutes at 37° C. Cells were pelleted from the suspension by centrifugation at 800 rpm for 3 minutes in Sorvall RT 6000 centrifuge. Cell pellet was resuspended in 500 μl of 1× PBS and counted under microscope Trypan Blue solution. Cells were diluted to 50 and 500 cells/μl in 1× PBS. 10 μl of each cell concentration, including zero point (10 μl of 1× PBS) were spiked in 3 ml of PreservCyt reagent. 100 μl of Sample Conversion Buffer were added into each tub to help visualize the cell pellet. All samples were mixed well and were spun down at 3800 rpm for 15 minutes in Sorvall RT 6000 centrifuge. Supernatants were discarded and tubes were drained by inversion on the Kimtowels for 2 to 5 minutes on the bench. All pellets were resuspended with 50 μl of the lysis reagent (50 units of Proteinase K) and mixture was transferred into the plate coated with streptavidin. Plate was covered with the plate sealer and was incubated at 37° C. for 30 minutes (heat block) with agitation every 15 minutes.
  • 50 μl of each E6/E7 RNA calibration were loaded in designated wells at 0, 103, 104, 105, 106, 107 and 108 molecules/well to construct calibration curve for mRNA in the specimen. 50 μl of the probe mix was added into each well. Plate was covered with the plate sealer and was agitated for 1 minute at 1100 rpm on the bench top shaker. Samples were incubated at 65° C. for 1.5 hours (hybridization reaction) in the heat block. Plate was transferred into the bench top shaker and was incubated for 1 hour with agitation at 1100 rpm at room temperature (capture reaction). 25 μl of Detection Reagent 1 was added into each well and plate was incubated without agitation for 1 hour at room temperature.
  • The contents of the plate were discarded and the plate was washed vigorously six times with Wash Buffer at 65° C. and one time with deionized water at room temperature. The plate was drained into Kimtowels and 100 μl of the Detection Reagent 2 was added into each well. Plate was incubated for 30 minutes at room temperature covered from the light. At the end of incubation time, plate was read on the Digene DML 2000 luminometer and the data were expressed as signal-to-noise.
  • Example 4 Comparison of the Expression of E6/E7 RNA and L1 RNA in Cell Lines Containing Episomal and Integrated HPV 16 DNA Cell Lines Tested
  • The following cell lines were examined according to the procedures outlined above.
      • HaCaT: an immortalized human keratinocyte cell line (Boukamp (1988)) SiHa: a human cancer cell line (Fried', (1970))
      • W12: a non-tumorigenic human cervical keratinocyte cell line (Stanley, (1989))
    HPV Infection Status
  • HaCaT cells were infected with HPV 16 by the procedure of White et. al. (White (1998)) to produce an episomal (non-integrated, total sequence, not spliced) HPV infection. Approximately 1 copy of HPV16 was present for every 40 cells. These cells are considered a representative of early stage infection or CIN I (cervical intraepithelial neoplasia). W12 cells contain approximately 100 copies of episomal HPV 16 DNA and represent pre-malignant, immortalized cells or CIN II or CIN III. SiHa cells contain 1-2 copies of HPV 16 integrated into the genome. These cells are considered to represent cancer.
  • Procedure
  • The RNA analysis was done according to Example 1 or the following procedure. Single stranded, biotinylated, DNA probes containing the specific HPV16 gene sequences were prepared. For HaCaT and SiHa cell lines, cells were grown to confluency, cells were harvested, and the total RNA was purified using the RNeasy kit (Qiagen Inc., Santa Clarita, Calif.). For W12, whole cells were used for analysis. RNA calibrators containing the complete HPV genome were prepared by transcribing (+) sense RNA from a plasmid containing the complete HPV16 genome with T7 RNA polymerase. The RNA was then diluted to 103, 104, 105, 106, and 107 copies per 50 pl. Aliquots of cellular RNA were diluted to 50 μl and then 50 μl of Probe mix (containing the biotinylated, single-stranded DNA probe) was added and hybridized to the RNA specimens for 2 hours at 65° C. The hybridization reactions were transferred to a streptavidin coated microplate and 25 μl of Detection Reagent 1 was added to each well. (Detection Reagent 1 contains the alkaline-phosphatase—anti-RNA:DNA monoclonal antibody conjugate.) During a 1 hour incubation with shaking, RNA:DNA hybrids were captured onto the streptavidin coated plate and were simultaneously reacted with the antihybrid antibody conjugate. After several wash steps, a chemiluminescent substrate (Tropix CDP-star with Emerald) was added to the wells, and the light output was measured in a microplate luminometer after 30 minutes incubation at room temperature.
  • Quantitation
  • The quantitation of HPV mRNA was performed as follows. The results from the RNA calibrators were used to construct standard curves. The regression equation was calculated from the logarithm of the copies versus the logarithm of signal to noise minus one [(S/N)−1)]. The regression equations were then used to calculate the number of copies of mRNA in the cellular RNA samples.
  • Ratio Results
  • The ratios of HPV 16 E6, E7, E2, E4, L1 and L2 were calculated for each cell type. The results are shown in Table 2. These results demonstrate that in an episomal, early stage infection the ratio of (E6+E7)/L1 is about 0.7, in the pre-malignant immortalized cell line the ratio is about 4 and in the cancerous cell line the ratio approaches infinity.
  • TABLE 2
    HaCaT W12 SiHa
    HPV Status Episomal Episomal Integrated
    Cell Status Early stage Pre- Malignant
    infection malignant,
    immortalized
    (E6 + E7)/L1 0.68 4.00 ∞*
    (E6 + E7)/(L1 + L2) ND 3.47 59.9
    (E7 + E6 + E2 + E4)/(L1 + L2) ND 6.96 70.6
    (E6 + E7)/(E2 + E4) ND 1.00  5.60
    (E6 + E7)/E2 ND 4.40 12.00
    (E6 + E7 + E2 − E4)/(L1 + L2) ND 1.58 59.10
    *L1 gene transcripts were undetectable in SiHa cells. Therefore, ratios of other gene transcripts to the Ll gene transcript are infinitely large.
  • Publications cited herein and the material for which they are cited are specifically incorporated by reference.
  • Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.
  • REFERENCES
    • Birkenmeyer & Mushahwar, J. Virol. Meth., 35:117-126 (1991)
    • Boukamp, et al., J. Cell Biol. 106:761-771 (1988).
    • Chernoff et al. J. Clinical Microbiology 35(10:2740-2744 (1997)
    • Cope et al. J. Clin. Microbiol. 35(9):2262-2265 (1997)
    • Cullen et al., J. Virol. 65(2):606-612 (1991)
    • Demeret et al., J. Virol. 68(1):7075-7082 (1994)
    • Dürst et al., J. Gen. Virol. 66:1515-1522 (1985)
    • Friedl et al., Proc. Soc. Exp. Biol. Med. 135(3):543-5 (1970)
    • Jeon et al., J. Virol. 69(5):2989-2997 (1995)
    • Kongsamul et al., Biochem. Biophys. Res. Commun. 127(1):71-9 (1985).
    • Landegren, Trends Genetics, 9(6):199-204 (1993)
    • Leiserowitz et al. Gynecol. Oncol. 66(2):295-299 (1997)
    • Lizard et al. Histochem J. 29(7):545-554 (1997)
    • Mant et al. J. Virol. Meth. 66(2):169-178 (1997)
    • Matsukura et al., Virology 172(1):63-72 (1989)
    • Meyers et al., Science 14; 257:971-3 (1992)
    • Nuovo, PCR In Situ Hybridization: Protocols and Applications, 3rd Edition, Lippencott-Raven Publishers, Philadelphia 1997
    • Pattillo et al., Science 196(4297):1456-8 (1977)
    • Schneider-Gädicke et al. EMBO J. 5:2285-2292 (1986)
    • Schwarz, et al., Nature 314:111-114 (1985)
    • Stanley et al., Int. J. Cancer 15;43(4):672-6 (1989)
    • Stoflet et al. Science 239:491-494 (1988)
    • Swan et al. J. Clin. Microbiol. 35(4):886-891 (1997)
    • Turek, Adv Virus Res. 44:305-356 (1994)
    • Ushikai et al., J. Virol. 68(1):6655-6666 (1994)
    • White et al., J. Virol. 72(2): 959-964 (1998).
    • Wilczynski et al., Virology 166:624-267 (1988)
    • Zehbe (1) et al. Am. J. Pathol. 150(5): 1553-1561 (1997)
    • Zehbe (2) et al. Mod. Pathol. 10(3):188-91(1997)

Claims (51)

1. A method of determining the medical character of an HPV-based disease comprising measuring the expression level of an HPV gene selected from the group consisting of E6, E7, E2, and L1 and correlating the expression level of the gene to the medical character of the HPV-based disease.
2. The method of claim 1 wherein low expression of E6 and/or E7 indicates a benign HPV infection.
3. The method of claim 1 wherein low to medium expression of E6 and/or E7 indicates a benign HPV infection characterized as CIN I.
4. The method of claim 1 wherein high expression of E6 and/or E7 indicates a neoplastic HPV infection.
5. The method of claim 1 wherein medium to high expression of E6 and/or E7 indicates a neoplastic HPV infection.
6. The method of claim 1 wherein high expression of E2 and/or L1 indicates a benign HPV infection.
7. The method of claim 1 wherein low to high expression of E2 indicates a benign HPV infection.
8. The method of claim 1 wherein medium to high expression of L1 indicates a benign HPV infection characterized as CIN I.
9. The method of claim 1 wherein low to undetectable expression of E2 indicates a neoplastic HPV infection.
10. The method of claim 1 wherein low to medium expression of E6 and/or E7 and medium to high expression of L1 indicates a benign HPV infection characterized as CIN I.
11. The method of claim 1 wherein low to medium expression of E6 and/or E7 and low to high expression of E2 indicates a benign HPV infection characterized as CIN I.
12. The method of claim 1 wherein medium to high expression of E6 and/or E7 and low to undetectable expression of E2 and/or L1 indicates a neoplastic HPV infection characterized as CIN II/III or cancer.
13. A method of determining progression of an HPV-based disease comprising measuring the expression level of an HPV gene selected from the group consisting of E6, E7, E2, and L1 at a first time point and a second later time point, and correlating the change in expression level to the medical character of the HPV-based disease.
14. The method of claim 13 wherein an increase in E6 and/or E7 gene expression from the first time point to the second time point indicates progression of the HPV-based disease.
15. The method of claim 13 wherein a decrease in E6 and/or E7 gene expression from the first time point to the second time point indicates regression of the HPV-based disease.
16. The method of claim 13 wherein a change in E6 and/or E7 gene expression from low at the first time point to high at the second time point to indicates progression from a benign to a neoplastic HPV infection.
17. The method of claim 13 wherein a change in E6 and/or E7 gene expression from high at the first time point to low at the second time point indicates regression from a neoplastic to a benign HPV infection.
18. The method of claim 13 wherein a change in E6 and/or E7 gene expression from low at the first time point to a range of medium to high at the second time point indicates progression from a benign to a neoplastic HPV infection.
19. The method of claim 13 wherein a change in E6 and/or E7 gene expression from a range of medium to high at the first time point to low at the second time point indicates regression from a neoplastic to a benign HPV infection.
20. The method of claim 13 wherein a change in E6 and/or E7 gene expression from a range of low to medium at the first time point to a range of medium to high at the second time point indicates progression from a benign to a neoplastic HPV infection.
21. The method of claim 13 wherein a change in E6 and/or E7 gene expression from a range of medium to high at the first time point to a range of low to medium at the second time point indicates regression from a neoplastic to a benign HPV infection.
22. The method of claim 13 wherein a decrease in E2 and/or L1 gene expression from the first time point to the second time point indicates progression of the HPV-based disease.
23. The method of claim 13 wherein an increase in E2 and/or L1 gene expression from the first time point to the second time point indicates regression of the HPV-based disease.
24. The method of claim 13 wherein a change in E2 gene expression from a range of low to high at the first time point to a range of low to undetectable at the second time point indicates progression from a benign to a neoplastic HPV infection.
25. The method of claim 13 wherein a change in E2 gene expression from a range of low to undetectable at the first time point to a range of low to high at the second time point indicates regression from a neoplastic to a benign HPV infection.
26. The method of claim 13 wherein a change in E2 gene expression from high at the first time point to undetectable at the second time point indicates progression from a benign to a neoplastic HPV infection.
27. The method of claim 13 wherein a change in E2 gene expression from undetectable at the first time point to high at the second time point indicates regression from a neoplastic to a benign HPV infection.
28. The method of claim 13 wherein a change in L1 gene expression from a range of medium to high at the first time point to a range of low to undetectable at the second time point indicates progression from a benign to a neoplastic HPV infection.
29. The method of claim 13 wherein a change in L1 gene expression from a range of low to undetectable at the first time point to a range of medium to high at the second time point indicates regression from a neoplastic to a benign HPV infection.
30. The method of claim 13 wherein progression of the HPV-based disease is indicated by:
(a) an increase in E6 and/or E7 gene expression from the first time point to the second time point; and
(b) a decrease in E2 and/or L1 gene expression from the first time point to the second time point.
31. The method of claim 13 wherein regression of the HPV-based disease is indicated by:
(a) a decrease in E6 and/or E7 gene expression from the first time point to the second time point;
(b) an increase in E2 and/or L1 gene expression from the first time point to the second time point.
32. The method of claim 13 wherein progression from a benign to a neoplastic HPV infection is indicated by:
(a) an increase in E6 and/or E7 gene expression from low at the first time point to high at the second time point; and
(b) a decrease in E2 gene expression from high at the first time point to undetectable at the second time point.
33. The method of claim 13 wherein progression from a benign to a neoplastic HPV infection is indicated by:
(a) an increase in E6 and/or E7 gene expression from low at the first time point to high at the second time point; and
(b) a decrease in L1 gene expression from a range of medium to high at the first time point to a range of low to undetectable at the second time point.
34. The method of claim 13 wherein progression from a benign to a neoplastic HPV infection is indicated by:
(a) an increase in E6 and/or E7 gene expression from low at the first time point to high at the second time point;
(b) a decrease in E2 gene expression from high at the first time point to undetectable at the second time point; and
(c) a decrease in L1 gene expression from a range of medium to high at the first time point to a range of low to undetectable at the second time point.
35. The method of claim 13 wherein regression from a neoplastic to a benign HPV infection is indicated by:
(a) a decrease in E6 and/or E7 gene expression from high at the first time point to low at the second time point; and
(b) an increase in E2 gene expression from undetectable at the first time point to high at the second time point.
36. The method of claim 13 wherein regression from a neoplastic to a benign HPV infection is indicated by:
(a) a decrease in E6 and/or E7 gene expression from high at the first time point to low at the second time point; and
(b) an increase in L1 gene expression from a range of low to undetectable at the first time point to a range of medium to high at the second time point.
37. The method of claim 13 wherein regression from a neoplastic to a benign HPV infection is indicated by:
(a) a decrease in E6 and/or E7 gene expression from high at the first time point to low at the second time point;
(b) an increase in E2 gene expression from undetectable at the first time point to high at the second time point; and
(c) an increase in L1 gene expression from a range of low to undetectable at the first time point to a range of medium to high at the second time point.
38. The method of claim 13 wherein progression from a benign to a neoplastic HPV infection is indicated by:
(a) an increase in E6 and/or E7 gene expression from low at the first time point to a range of medium to high at the second time point; and
(b) a decrease in E2 gene expression from a range of low to high at the first time point to a range of low to undetectable at the second time point.
39. The method of claim 13 wherein progression from a benign to a neoplastic HPV infection is indicated by:
(a) an increase in E6 and/or E7 gene expression from low at the first time point to a range of medium to high at the second time point; and
(b) a decrease in L1 gene expression from a range of medium to high at the first time point to a range of low to undetectable at the second time point.
40. The method of claim 13 wherein progression from a benign to a neoplastic HPV infection is indicated by:
(a) an increase in E6 and/or E7 gene expression from low at the first time point to a range of medium to high at the second time point;
(b) a decrease in E2 gene expression from a range of low to high at the first time point to a range of low to undetectable at the second time point; and
(c) a decrease in L1 gene expression from a range of medium to high at the first time point to a range of low to undetectable at the second time point.
41. The method of claim 13 wherein regression from a neoplastic to a benign HPV infection is indicated by:
(a) a decrease in E6 and/or E7 gene expression from a range of medium to high at the first time point to low at the second time point; and
(b) an increase in E2 gene expression from a range of low to undetectable at the first time point to a range of low to high at the second time point.
42. The method of claim 13 wherein regression from a neoplastic to a benign HPV infection is indicated by:
(a) a decrease in E6 and/or E7 gene expression from a range of medium to high at the first time point to low at the second time point; and
(b) an increase in L1 gene expression from a range of low to undetectable at the first time point to a range of medium to high at the second time point.
43. The method of claim 13 wherein regression from a neoplastic to a benign HPV infection is indicated by:
(a) a decrease in E6 and/or E7 gene expression from a range of medium to high at the first time point to low at the second time point;
(b) an increase in E2 gene expression from a range of low to undetectable at the first time point to a range of low to high at the second time point; and
(c) an increase in L1 gene expression from a range of low to undetectable at the first time point to a range of medium to high at the second time point.
44. The method of claim 13 wherein progression from a benign to a neoplastic HPV infection is indicate by:
(a) an increase in E6 and/or E7 gene expression from a range of low to medium at the first time point to a range of medium to high at the second time point; and
(b) a decrease in E2 gene expression from a range of low to high at the first time point to a range of low to undetectable at the second time point.
45. The method of claim 13 wherein progression from a benign to a neoplastic HPV infection is indicate by:
(a) an increase in E6 and/or E7 gene expression from a range of low to medium at the first time point to a range of medium to high at the second time point; and
(b) a decrease in L1 gene expression from a range of medium to high at the first time point to a range of low to undetectable at the second time point.
46. The method of claim 13 wherein progression from a benign to a neoplastic HPV infection is indicate by:
(a) an increase in E6 and/or E7 gene expression from a range of low to medium at the first time point to a range of medium to high at the second time point;
(b) a decrease in E2 gene expression from a range of low to high at the first time point to a range of low to undetectable at the second time point; and
(c) a decrease in L1 gene expression from a range of medium to high at the first time point to a range of low to undetectable at the second time point.
47. The method of claim 13 wherein regression from a neoplastic to a benign HPV infection is indicated by:
(a) a decrease in E6 and/or E7 gene expression from a range of medium to high at the first time point to a range of low to medium at the second time point; and
(b) an increase in E2 gene expression from a range of low to undetectable at the first time point to a range of low to high at the second time point.
48. The method of claim 13 wherein regression from a neoplastic to a benign HPV infection is indicated by:
(a) a decrease in E6 and/or E7 gene expression from a range of medium to high at the first time point to a range of low to medium at the second time point; and
(b) an increase in L1 gene expression from a range of low to undetectable at the first time point to a range of medium to high at the second time point.
49. The method of claim 13 wherein regression from a neoplastic to a benign HPV infection is indicated by:
(a) a decrease in E6 and/or E7 gene expression from a range of medium to high at the first time point to a range of low to medium at the second time point;
(b) an increase in E2 gene expression from a range of low to undetectable at the first time point to a range of low to high at the second time point; and
(c) an increase in L1 gene expression from a range of low to undetectable at the first time point to a range of medium to high at the second time point.
50. The method of claim 13 wherein progression from HPV-infected normal tissue to CIN-I is indicated by:
(a) an increase in L1 gene expression from a range of low to undetectable at the first time point to a range of medium to high at the second time point; and
(b) low to medium E6 and/or E7 gene expression at the second time point.
51. The method of claim 13 wherein progression from CIN-I to CIN II/II or cancer is indicated by:
(a) a decrease in L1 gene expression from a range of medium to high at the first time point to a range of low to undetectable at the second time point; and
(b) a medium to high level of E6 and/or E7 gene expression at the second time point.
US12/976,428 1997-12-12 2010-12-22 Assessment of human papilloma virus-related disease Abandoned US20110097708A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/976,428 US20110097708A1 (en) 1997-12-12 2010-12-22 Assessment of human papilloma virus-related disease

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US6942697P 1997-12-12 1997-12-12
US7048698P 1998-01-05 1998-01-05
US8216798P 1998-04-17 1998-04-17
US09/210,168 US6355424B1 (en) 1997-12-12 1998-12-11 Assessment of human papillomavirus-related disease
US09/970,477 US7291455B2 (en) 1997-12-12 2001-10-04 Assessment of human papilloma virus-related disease
US11/681,539 US7879546B2 (en) 1997-12-12 2007-03-02 Assessment of human papilloma virus-related disease
US12/976,428 US20110097708A1 (en) 1997-12-12 2010-12-22 Assessment of human papilloma virus-related disease

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/681,539 Continuation US7879546B2 (en) 1997-12-12 2007-03-02 Assessment of human papilloma virus-related disease

Publications (1)

Publication Number Publication Date
US20110097708A1 true US20110097708A1 (en) 2011-04-28

Family

ID=27371534

Family Applications (5)

Application Number Title Priority Date Filing Date
US09/210,168 Expired - Lifetime US6355424B1 (en) 1997-12-12 1998-12-11 Assessment of human papillomavirus-related disease
US09/210,031 Expired - Lifetime US6969585B2 (en) 1997-12-12 1998-12-11 Universal collection medium
US09/970,477 Expired - Fee Related US7291455B2 (en) 1997-12-12 2001-10-04 Assessment of human papilloma virus-related disease
US11/681,539 Expired - Fee Related US7879546B2 (en) 1997-12-12 2007-03-02 Assessment of human papilloma virus-related disease
US12/976,428 Abandoned US20110097708A1 (en) 1997-12-12 2010-12-22 Assessment of human papilloma virus-related disease

Family Applications Before (4)

Application Number Title Priority Date Filing Date
US09/210,168 Expired - Lifetime US6355424B1 (en) 1997-12-12 1998-12-11 Assessment of human papillomavirus-related disease
US09/210,031 Expired - Lifetime US6969585B2 (en) 1997-12-12 1998-12-11 Universal collection medium
US09/970,477 Expired - Fee Related US7291455B2 (en) 1997-12-12 2001-10-04 Assessment of human papilloma virus-related disease
US11/681,539 Expired - Fee Related US7879546B2 (en) 1997-12-12 2007-03-02 Assessment of human papilloma virus-related disease

Country Status (13)

Country Link
US (5) US6355424B1 (en)
EP (2) EP1038029A2 (en)
JP (3) JP2002508190A (en)
AT (1) ATE299946T1 (en)
AU (2) AU748022B2 (en)
BR (2) BR9814271A (en)
CA (2) CA2313641A1 (en)
DE (1) DE69830927T2 (en)
DK (1) DK1038022T3 (en)
ES (1) ES2246546T3 (en)
NO (2) NO20002980L (en)
PT (1) PT1038022E (en)
WO (2) WO1999029890A2 (en)

Families Citing this family (78)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BR9814271A (en) * 1997-12-12 2001-03-20 Digene Corp Determination of diseases related to human papilloma virus
JP2003519829A (en) 1999-10-13 2003-06-24 シークエノム・インコーポレーテツド Methods for creating a database and a database for identifying polymorphic genetic markers
KR100382703B1 (en) * 2000-03-15 2003-05-09 주식회사 바이오메드랩 diagnosis kit for genotyping of Human Papillomavirus and manufacturing method for thereof
AU2001249526A1 (en) * 2000-03-28 2001-10-08 Biosearch International Pty Ltd Approaches for hpv detection and staging by targeting the e6 gene region of the viral genome
US20040115692A1 (en) * 2000-04-03 2004-06-17 Cytyc Corporation Methods, compositions and apparatuses for detecting a target in a preservative solution
JP2003529376A (en) * 2000-04-03 2003-10-07 サイティック コーポレイション Detection and typing of human papillomavirus using PNA probes
IT1316070B1 (en) * 2000-05-05 2003-03-28 Bioanalisi Ct Sud Snc Di Perse METHOD AND MEANS FOR IDENTIFICATION OF VIRUS HPV
US7601497B2 (en) * 2000-06-15 2009-10-13 Qiagen Gaithersburg, Inc. Detection of nucleic acids by target-specific hybrid capture method
US7439016B1 (en) * 2000-06-15 2008-10-21 Digene Corporation Detection of nucleic acids by type-specific hybrid capture method
EP1294853B1 (en) * 2000-06-21 2010-05-05 QIAGEN Gaithersburg, Inc. Universal collection medium
GB0018140D0 (en) * 2000-07-24 2000-09-13 Medical Res Council Screening for abnormalities
US20040229298A1 (en) * 2000-11-11 2004-11-18 Lu Peter S. Methods and compositions for treating cervical cancer
US20040121310A1 (en) * 2002-12-18 2004-06-24 Ecker David J. Methods for rapid detection and identification of bioagents in forensic studies
KR100437626B1 (en) * 2001-06-21 2004-06-26 주식회사 마이진 Methods for Dectecting Human Papillomavirus and Detection Kit Thereof
DK1463839T3 (en) 2002-01-07 2007-07-02 Norchip As Method for detecting human papillomavirus mRNA
EP1507148B1 (en) 2003-08-25 2005-02-23 MTM Laboratories AG Method for detecting carcinomas in a solubilized cervical body sample
US20060247190A1 (en) 2002-10-21 2006-11-02 Kathleen Beach Compositions and methods for treating human papillomavirus mediated disease
ATE382099T1 (en) * 2003-03-31 2008-01-15 Stichting Res Fonds Pathologie DETECTION OF INVASIVE CANCER INDUCED BY HPV AND ITS PRECURSOR LESIONS WITH INVASION POTENTIAL
US20040203004A1 (en) * 2003-04-10 2004-10-14 Bernard Hans Ulrich Diagnostic apparatus and method
US7361460B2 (en) 2003-04-11 2008-04-22 Digene Corporation Approach to molecular diagnosis of human papillomavirus-related diseases
WO2005008248A2 (en) * 2003-07-18 2005-01-27 Georgetown University Diagnosis and treatment of cervical cancer
EP1510820B1 (en) * 2003-08-25 2010-03-17 MTM Laboratories AG Method for detecting medically relevant conditions in a solubilized LBC sample
US7482142B1 (en) 2004-05-07 2009-01-27 Roche Molecular Systems, Inc. High-risk human papillomavirus detection
US7316904B1 (en) 2004-06-30 2008-01-08 Chromodynamics, Inc. Automated pap screening using optical detection of HPV with or without multispectral imaging
KR100572721B1 (en) * 2004-07-26 2006-04-24 이미영 RNA preservation medium comprising cathodic electrolyzed water and method of preserving RNA using the same
CA2588581C (en) 2004-12-08 2011-05-31 Gen-Probe Incorporated Detection of nucleic acids from multiple types of human papillomaviruses
US20060189893A1 (en) * 2005-01-06 2006-08-24 Diamics, Inc. Systems and methods for detecting abnormal cells
US20060161076A1 (en) * 2005-01-06 2006-07-20 Diamics, Inc. Systems and methods for collection of cell clusters
GB0500996D0 (en) * 2005-01-18 2005-02-23 Delfts Diagnostic Labaratory B Detection method and materials therefor
US7524631B2 (en) * 2005-02-02 2009-04-28 Patterson Bruce K HPV E6, E7 mRNA assay and methods of use thereof
US20070111960A1 (en) * 2005-03-04 2007-05-17 Advandx, Inc. High affinity probes for analysis of human papillomavirus expression
DE102005015005A1 (en) * 2005-04-01 2006-10-05 Qiagen Gmbh Process for treating a sample containing biomolecules
US7972776B2 (en) * 2005-11-15 2011-07-05 Oncohealth Corporation Protein chips for HPV detection
US7732166B2 (en) * 2005-11-15 2010-06-08 Oncohealth Corporation Detection method for human pappilomavirus (HPV) and its application in cervical cancer
WO2007149350A2 (en) * 2006-06-19 2007-12-27 Becton, Dickinson And Company Methods and compositions for obtaining amplifiable nucleic acids from tissues, cells, or viruses exposed to transport media
EP2484781B1 (en) * 2006-08-01 2017-08-23 Applied Biosystems, LLC Detection of analytes and nucleic acids
US20100003704A1 (en) * 2008-06-13 2010-01-07 Shuling Cheng IN SITU detection of early stages and late stages HPV infection
US8968995B2 (en) * 2008-11-12 2015-03-03 Oncohealth Corp. Detection, screening, and diagnosis of HPV-associated cancers
WO2008071998A2 (en) * 2006-12-15 2008-06-19 Oncomethylome Sciences Sa Diagnostic methods for diseases caused by a hpv infection comprising determining the methylation status of the hpv genome
JP5241100B2 (en) * 2006-12-28 2013-07-17 シスメックス株式会社 Antigen activation liquid, antigen activation method, and cell detection method
US20090311392A1 (en) * 2007-09-20 2009-12-17 Paul Bernard Newman Novel approach to the controlled decontamination and or detoxification of nuts, grains, fruits and vegetables
WO2009092017A1 (en) * 2008-01-16 2009-07-23 Medical College Of Georgia Research Institute, Inc. Biomarkers for hpv-induced cancer
AU2009238247B2 (en) * 2008-04-17 2014-12-11 Qiagen Gaithersburg, Inc. Compositions, methods, and kits using synthetic probes for determining the presence of a target nucleic acid
US9186128B2 (en) 2008-10-01 2015-11-17 Covidien Lp Needle biopsy device
US11298113B2 (en) 2008-10-01 2022-04-12 Covidien Lp Device for needle biopsy with integrated needle protection
US8968210B2 (en) 2008-10-01 2015-03-03 Covidien LLP Device for needle biopsy with integrated needle protection
US9782565B2 (en) 2008-10-01 2017-10-10 Covidien Lp Endoscopic ultrasound-guided biliary access system
US9332973B2 (en) 2008-10-01 2016-05-10 Covidien Lp Needle biopsy device with exchangeable needle and integrated needle protection
TWI417389B (en) * 2008-10-27 2013-12-01 Qiagen Gaithersburg Inc Fast results hybrid capture assay on an automated platform
EP2184368A1 (en) 2008-11-07 2010-05-12 DKFZ Deutsches Krebsforschungszentrum Diagnostic transcript and splice patterns of HPV16 in different cervical lesions
GB0820822D0 (en) * 2008-11-13 2008-12-24 Inst Catala D Oncologia Novel product and processes
JPWO2010064628A1 (en) * 2008-12-05 2012-05-10 オリンパス株式会社 Nucleic acid-containing sample preparation method, sample preparation solution, and nucleic acid analysis method
SG173140A1 (en) 2009-01-27 2011-08-29 Genetic Technologies Ltd Biological sampling device
ES2644516T3 (en) * 2009-01-28 2017-11-29 Qiagen Gaithersburg, Inc. Method and test for sample preparation of large sequence specific volume
CN102414327B (en) 2009-05-01 2015-04-08 奇亚根盖瑟斯堡股份有限公司 A non-target amplification method for detection of RNA splice-forms in a sample
CN102449479B (en) 2009-05-07 2015-11-25 安科健康公司 Differentiate height or >=CIN2 be used for the early stage of Human infectious warts virus (HPV) and HPV associated cancer and detect late period, screening and diagnosis
EP2473596B1 (en) 2009-09-03 2017-12-13 Becton, Dickinson and Company Methods and compositions for direct chemical lysis
JP5826752B2 (en) 2009-09-14 2015-12-02 キアジェン ゲイサーズバーグ インコーポレイテッド Compositions and methods for recovering nucleic acids or proteins from tissue samples fixed in cytological media
US8722328B2 (en) * 2010-01-04 2014-05-13 Qiagen Gaithersburg, Inc. Methods, compositions, and kits for recovery of nucleic acids or proteins from fixed tissue samples
WO2011084598A1 (en) 2010-01-08 2011-07-14 Oncohealth Corporation High throughput cell-based hpv immunoassays for diagnosis and screening of hpv-associated cancers
ES2615728T3 (en) * 2010-01-29 2017-06-08 Qiagen Gaithersburg, Inc. Methods and compositions for purification and specific multiple nucleic acid sequence analysis
BR112012018545A2 (en) 2010-01-29 2016-05-03 Qiagen Gaithersburg Inc method of determining and confirming the presence of an hpv in a sample
AU2011255638B2 (en) 2010-05-19 2016-08-25 Qiagen Gaithersburg, Inc. Methods and compositions for sequence-specific purification and multiplex analysis of nucleic acids
CA2799205A1 (en) 2010-05-25 2011-12-01 Qiagen Gaithersburg, Inc. Fast results hybrid capture assay and associated strategically-truncated probes
JP2014511182A (en) * 2011-02-24 2014-05-15 キアゲン ガイサーズバーグ アイエヌシー. Materials and methods for detecting HPV nucleic acids
US20140127745A1 (en) * 2011-06-24 2014-05-08 Biotechnology Developers, S.A. Method, compositions and device for preparing cytological specimens
CA2854165C (en) 2011-11-03 2022-11-22 Qiagen Gaithersburg, Inc. Materials and method for immobilizing, isolating, and concentrating cells using carboxylated surfaces
JP5960421B2 (en) * 2011-12-05 2016-08-02 株式会社日立ハイテクノロジーズ Cell measuring method and cell measuring reagent
WO2013162756A1 (en) 2012-04-28 2013-10-31 Cytocore, Inc. Methods, packaging and apparatus for collection of biological samples
US8734364B1 (en) 2013-11-07 2014-05-27 Genetic Technologies Limited Device and method for obtaining a biological sample
WO2016005789A1 (en) * 2014-07-07 2016-01-14 Institut Pasteur Broad range gene and genotype papillomavirus transcriptome as a biomarker of papillomavirus- associated cancer stages
EP3225685B1 (en) 2015-06-30 2018-12-12 Sysmex Corporation Cell preservative solution, use of same, and method for producing cell preservative solution
AU2017338827B2 (en) 2016-10-03 2023-08-31 Juno Therapeutics, Inc. HPV-specific binding molecules
KR20200104284A (en) 2017-10-03 2020-09-03 주노 쎄러퓨티크스 인코퍼레이티드 HPV-specific binding molecule
CA3095027A1 (en) 2018-04-05 2019-10-10 Juno Therapeutics, Inc. T cell receptors and engineered cells expressing same
PL240016B1 (en) * 2019-09-09 2022-02-07 Genomtec Spolka Akcyjna Set of primers for the detection of human papillomavirus type 16 (HPV16 Human papillomavirus type 16) and human papillomavirus type 18 (HPV18 Human papillomavirus type 18), method of detecting HPV16 and HPV18 infections, the application of a set of primers for the detection of HPV16 and HPV18 infections
WO2021156404A2 (en) * 2020-02-07 2021-08-12 Isa Pharmaceuticals Treatment of hpv-related diseases
WO2023081900A1 (en) 2021-11-08 2023-05-11 Juno Therapeutics, Inc. Engineered t cells expressing a recombinant t cell receptor (tcr) and related systems and methods

Citations (87)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4465078A (en) * 1982-09-30 1984-08-14 Medtest Corporation Method for cell sampling in a body cavity
US4486539A (en) * 1981-10-16 1984-12-04 Orioon Corporation Ltd. Detection of microbial nucleic acids by a one-step sandwich hybridization test
US4563417A (en) * 1984-08-31 1986-01-07 Miles Laboratories, Inc. Nucleic acid hybridization assay employing antibodies to intercalation complexes
US4731325A (en) * 1984-02-17 1988-03-15 Orion-Yhtyma Arrays of alternating nucleic acid fragments for hybridization arrays
US4732847A (en) * 1981-06-09 1988-03-22 University Of Hawaii Monoclonal antibodies for DNA-RNA hybrid complexes and their uses
US4743535A (en) * 1984-11-07 1988-05-10 Miles Inc. Hybridization assay employing labeled probe and anti-hybrid
US4751177A (en) * 1985-06-13 1988-06-14 Amgen Methods and kits for performing nucleic acid hybridization assays
US4775619A (en) * 1984-10-16 1988-10-04 Chiron Corporation Polynucleotide determination with selectable cleavage sites
US4851330A (en) * 1983-01-10 1989-07-25 Kohne David E Method for detection, identification and quantitation of non-viral organisms
US4865980A (en) * 1982-12-29 1989-09-12 University Of Hawaii Monoclonal antibodies for DNA-RNA hybrid complexes and their uses
US4868105A (en) * 1985-12-11 1989-09-19 Chiron Corporation Solution phase nucleic acid sandwich assay
US4883084A (en) * 1987-07-21 1989-11-28 Ti Interlock Limited Pressure fluid control system
US4889798A (en) * 1984-02-09 1989-12-26 Enzo Biochem, Inc. Heterologous system for the detection of chemically labeled DNA and other biological materials providing a receptor or target moiety thereon
US4894325A (en) * 1984-04-27 1990-01-16 Enzo Biochem, Inc. Hybridization method for the detection of genetic material
US5082830A (en) * 1988-02-26 1992-01-21 Enzo Biochem, Inc. End labeled nucleotide probe
US5106727A (en) * 1989-04-27 1992-04-21 Life Technologies, Inc. Amplification of nucleic acid sequences using oligonucleotides of random sequences as primers
US5116734A (en) * 1989-09-01 1992-05-26 Digene Diagnostics, Inc. Highly sensitive method for detecting peroxidase
US5200313A (en) * 1983-08-05 1993-04-06 Miles Inc. Nucleic acid hybridization assay employing detectable anti-hybrid antibodies
US5256571A (en) * 1991-05-01 1993-10-26 Cytyc Corporation Cell preservative solution
US5288611A (en) * 1983-01-10 1994-02-22 Gen-Probe Incorporated Method for detecting, identifying, and quantitating organisms and viruses
US5357977A (en) * 1993-04-23 1994-10-25 St. Mary's Hospital And Medical Center, Inc. Cytological sampling method and device
US5370128A (en) * 1993-12-02 1994-12-06 Wainwright; Sharon R. Pap brush and pap unit container systems
US5374524A (en) * 1988-05-10 1994-12-20 E. I. Du Pont De Nemours And Company Solution sandwich hybridization, capture and detection of amplified nucleic acids
US5424413A (en) * 1992-01-22 1995-06-13 Gen-Probe Incorporated Branched nucleic acid probes
US5437977A (en) * 1990-04-03 1995-08-01 David Segev DNA probe signal amplification
US5474895A (en) * 1990-11-14 1995-12-12 Siska Diagnostics Inc. Non-isotopic detection of nucleic acids using a polystyrene support-based sandwich hybridization assay and compositions useful therefor
US5484699A (en) * 1990-09-28 1996-01-16 Abbott Laboratories Nucleotide sequences useful as type specific probes, PCR primers and LCR probes for the amplification and detection of human papilloma virus, and related kits and methods
US5527898A (en) * 1988-09-09 1996-06-18 Hoffmann-La Roche Inc. Detection of human papillomavirus by the polymerase chain reaction
US5543294A (en) * 1991-07-19 1996-08-06 The Trustees Of Columbia University In The City Of New York Polymerase chain reaction/restriction fragment length polymorphism method for the detection and typing of myobacteria
US5556748A (en) * 1991-07-30 1996-09-17 Xenopore Corporation Methods of sandwich hybridization for the quantitative analysis of oligonucleotides
US5580970A (en) * 1989-12-01 1996-12-03 Amoco Corporation Detection of HPV transcripts
US5614362A (en) * 1987-10-15 1997-03-25 Chiron Corporation Nucleic acid hybridization assay for hepatitis B virus DNA
US5627030A (en) * 1994-06-28 1997-05-06 Kalyx Biosciences Inc. Method of amplification for increasing the sensitivity of detecting nucleic acid-probe target hybrids
US5629156A (en) * 1992-10-09 1997-05-13 Amoco Corporation Multiple capture probe sandwich assays
US5629153A (en) * 1990-01-10 1997-05-13 Chiron Corporation Use of DNA-dependent RNA polymerase transcripts as reporter molecules for signal amplification in nucleic acid hybridization assays
US5641630A (en) * 1985-06-13 1997-06-24 Amgen Inc. Method and kit for performing nucleic acid hybridization assays
US5656731A (en) * 1987-10-15 1997-08-12 Chiron Corporation Nucleic acid-amplified immunoassay probes
US5681697A (en) * 1993-12-08 1997-10-28 Chiron Corporation Solution phase nucleic acid sandwich assays having reduced background noise and kits therefor
US5695926A (en) * 1990-06-11 1997-12-09 Bio Merieux Sandwich hybridization assays using very short capture probes noncovalently bound to a hydrophobic support
US5702893A (en) * 1989-06-30 1997-12-30 Chiron Corporation Hydrophobic nucleic acid probe
US5728531A (en) * 1994-09-30 1998-03-17 Kabushiki Kaisha Toyota Chuo Kenkyusho Method of detecting nucleic acid
US5731153A (en) * 1996-08-26 1998-03-24 The Regents Of The University Of California Identification of random nucleic acid sequence aberrations using dual capture probes which hybridize to different chromosome regions
US5736316A (en) * 1991-12-23 1998-04-07 Chiron Corporation HBV capture and amplifiers probes for use in solution phase sandwich hybridization assays
US5747248A (en) * 1994-12-05 1998-05-05 Chiron Corporation Discontinuous probe design using hybritope mapping
US5747244A (en) * 1991-12-23 1998-05-05 Chiron Corporation Nucleic acid probes immobilized on polystyrene surfaces
US5750338A (en) * 1986-10-23 1998-05-12 Amoco Corporation Target and background capture methods with amplification for affinity assays
US5759773A (en) * 1993-01-15 1998-06-02 The Public Health Research Institute Of The City Of New York, Inc. Sensitive nucleic acid sandwich hybridization assay
US5792606A (en) * 1990-02-26 1998-08-11 Boehringer Mannheim Gmbh Nucleic acid hybridization based assay for determining a substance of interest
US5800994A (en) * 1994-04-04 1998-09-01 Chiron Diagnostics Corporation Hybridization-ligation assays for the detection of specific nucleic acid sequences
US5821339A (en) * 1993-05-20 1998-10-13 Dana Farber Cancer Institute Compositions and methods for treatment of herpesvirus infections
US5827661A (en) * 1994-12-23 1998-10-27 Kalyx Biosciences Incorporated Enhancing detection polymerase chain reaction assays by RNA transcription and immunodetection of RNA:DNA hybrids
US5853993A (en) * 1996-10-21 1998-12-29 Hewlett-Packard Company Signal enhancement method and kit
US5888724A (en) * 1995-02-17 1999-03-30 The Trustees Of Columbia University In The City Of New York Detection of high oncogenic-risk papilloma virus in high grade cervical lesions and cancers by a PCR/ELISA assay
US5981179A (en) * 1991-11-14 1999-11-09 Digene Diagnostics, Inc. Continuous amplification reaction
US5994079A (en) * 1998-02-06 1999-11-30 Digene Corporation Direct detection of RNA mediated by reverse transcriptase lacking RNAse H function
US6043038A (en) * 1998-03-31 2000-03-28 Tularik, Inc. High-throughput screening assays for modulators of primase activity
US6057099A (en) * 1994-12-02 2000-05-02 Intelligene Ltd. Detection of nucleic acid sequences
US6083925A (en) * 1995-06-07 2000-07-04 Connaught Laboratories Limited Nucleic acid respiratory syncytial virus vaccines
US6110676A (en) * 1996-12-04 2000-08-29 Boston Probes, Inc. Methods for suppressing the binding of detectable probes to non-target sequences in hybridization assays
US6221581B1 (en) * 1984-04-27 2001-04-24 Enzo Diagnostics, Inc. Processes for detecting polynucleotides, determining genetic mutations or defects in genetic material, separating or isolating nucleic acid of interest from samples, and useful compositions of matter and multihybrid complex compositions
US6228578B1 (en) * 1991-11-14 2001-05-08 Digene Corporation Non-radioactive hybridization assay and kit
US6232462B1 (en) * 1994-08-30 2001-05-15 Bayer Corporation Reduction of nonspecific hybridization by using novel base-pairing schemes
US6268128B1 (en) * 1989-03-10 2001-07-31 Vysis, Inc. Immobilized oligonucleotide probes and uses thereof
US6326136B1 (en) * 1988-04-01 2001-12-04 Digene Corporation Macromolecular conjugate made using unsaturated aldehydes
US20010055766A1 (en) * 1999-04-02 2001-12-27 Alexander Aristarkhov Immunosorbant assay using branched bis-biotin/avidin/multiple label complex as a detection reagent
US6355424B1 (en) * 1997-12-12 2002-03-12 Digene Corporation Assessment of human papillomavirus-related disease
US6436662B1 (en) * 2000-04-04 2002-08-20 Digene Corporation Device and method for cytology slide preparation
US6521190B1 (en) * 2000-05-19 2003-02-18 Digene Corporation Cell collection apparatus
US6544732B1 (en) * 1999-05-20 2003-04-08 Illumina, Inc. Encoding and decoding of array sensors utilizing nanocrystals
US20030096232A1 (en) * 1997-12-19 2003-05-22 Kris Richard M. High throughput assay system
US20030108897A1 (en) * 1997-01-16 2003-06-12 Drmanac Radoje T. Methods and compositions for detection or quantification of nucleic acid species
US6583278B1 (en) * 1995-11-15 2003-06-24 Gen-Probe Incorporated Nucleic acid probes complementary to human papilloma virus nucleic acid
US20030175765A1 (en) * 1997-11-04 2003-09-18 Christoph Kessler Specific and sensitive nucleic acid detection method
US6686151B1 (en) * 1998-02-06 2004-02-03 Digene Corporation Immunological detection of RNA:DNA hybrids on microarrays
US20040214302A1 (en) * 2000-06-15 2004-10-28 James Anthony Detection of nucleic acids by type-specific hybrid capture method
US6828098B2 (en) * 2000-05-20 2004-12-07 The Regents Of The University Of Michigan Method of producing a DNA library using positional amplification based on the use of adaptors and nick translation
US20050147996A1 (en) * 2000-11-21 2005-07-07 Sorge Joseph A. Methods for detection of a nucleic acid by sequential amplification
US6977148B2 (en) * 2001-10-15 2005-12-20 Qiagen Gmbh Multiple displacement amplification
US20060051809A1 (en) * 2000-06-15 2006-03-09 Irina Nazarenko Detection of nucleic acids by target-specific hybrid capture method
US20060160188A1 (en) * 2005-01-14 2006-07-20 Kurnit David M Systems, methods , and compositions for detection of human papilloma virus in biological samples
US7371518B2 (en) * 2000-06-21 2008-05-13 Digene Corporation Universal collection medium
US20080200344A1 (en) * 2005-11-15 2008-08-21 Shu-Ling Cheng Protein chips for HPV detection
US20080247914A1 (en) * 2007-04-06 2008-10-09 Ted Carl Edens Sample Preparation System And Method for Processing Clinical Specimens
US20090032445A1 (en) * 1999-04-29 2009-02-05 Mss, Inc. Multi-Grade Object Sorting System And Method
US20090263819A1 (en) * 2007-02-27 2009-10-22 Nuclea Biomarkers, Llc Gene and protein expression profiles associated with the therapeutic efficacy of egfr-tk inhibitors
US20090298187A1 (en) * 2008-04-17 2009-12-03 Qiagen Gaithersburg, Inc. Compositions, methods, and kits using synthetic probes for determining the presence of a target nucleic acid
US20100081124A1 (en) * 2008-09-30 2010-04-01 Abbott Laboratories Primers and probes for detecting human papillomavirus and human beta globin sequences in test samples

Family Cites Families (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4578282A (en) * 1982-02-04 1986-03-25 Harrison James S Composition for diagnostic reagents
EP0144914A3 (en) 1983-12-12 1986-08-13 Miles Inc. Hybridization assay employing labeled pairs of hybrid binding reagents
CA1253777A (en) 1984-06-01 1989-05-09 Robert J. Carrico Nucleic acid hybridization assay employing immobilized rna probes
ZA853756B (en) 1984-06-01 1986-01-29 Miles Lab Nucleic acid hybridization assay employing immobilized rna probes
US4888278A (en) * 1985-10-22 1989-12-19 University Of Massachusetts Medical Center In-situ hybridization to detect nucleic acid sequences in morphologically intact cells
IL85551A0 (en) 1987-04-01 1988-08-31 Miles Inc Rapid hybridization assay and reagent system used therein
AU6966991A (en) * 1989-12-01 1991-06-26 Gene-Trak Systems Detection of hpv transcripts
IL106273A0 (en) 1992-07-17 1993-11-15 Res Dev Foundation Rapid detection of biopolymers in stained specimens
KR100189229B1 (en) 1993-07-23 1999-06-01 다니엘 엘. 캐시앙, 헨리 엘. 노르호프 Methods for enhancing nucleic acid amplification
DE4331012A1 (en) 1993-09-13 1995-03-16 Bayer Ag Nucleic acid-binding oligomers with N-branching for therapy and diagnostics
DE4445769C1 (en) * 1994-12-21 1995-11-09 Biochrom Kg Protective media for storing vital tissues
US5679333A (en) * 1996-10-25 1997-10-21 Dunphy; Brian William Formaldehyde-free tissue preservative compositions
US7399589B2 (en) 1998-02-06 2008-07-15 Digene Corporation Immunological detection of RNA:DNA hybrids on microarrays

Patent Citations (95)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4732847A (en) * 1981-06-09 1988-03-22 University Of Hawaii Monoclonal antibodies for DNA-RNA hybrid complexes and their uses
US4486539A (en) * 1981-10-16 1984-12-04 Orioon Corporation Ltd. Detection of microbial nucleic acids by a one-step sandwich hybridization test
US4563419A (en) * 1981-10-16 1986-01-07 Orion Corporation Ltd. Detection of microbial nucleic acids by a one-step sandwich hybridization test
US4465078A (en) * 1982-09-30 1984-08-14 Medtest Corporation Method for cell sampling in a body cavity
US4865980A (en) * 1982-12-29 1989-09-12 University Of Hawaii Monoclonal antibodies for DNA-RNA hybrid complexes and their uses
US4851330A (en) * 1983-01-10 1989-07-25 Kohne David E Method for detection, identification and quantitation of non-viral organisms
US5288611A (en) * 1983-01-10 1994-02-22 Gen-Probe Incorporated Method for detecting, identifying, and quantitating organisms and viruses
US5200313A (en) * 1983-08-05 1993-04-06 Miles Inc. Nucleic acid hybridization assay employing detectable anti-hybrid antibodies
US4889798A (en) * 1984-02-09 1989-12-26 Enzo Biochem, Inc. Heterologous system for the detection of chemically labeled DNA and other biological materials providing a receptor or target moiety thereon
US4731325A (en) * 1984-02-17 1988-03-15 Orion-Yhtyma Arrays of alternating nucleic acid fragments for hybridization arrays
US4894325A (en) * 1984-04-27 1990-01-16 Enzo Biochem, Inc. Hybridization method for the detection of genetic material
US6221581B1 (en) * 1984-04-27 2001-04-24 Enzo Diagnostics, Inc. Processes for detecting polynucleotides, determining genetic mutations or defects in genetic material, separating or isolating nucleic acid of interest from samples, and useful compositions of matter and multihybrid complex compositions
US4563417A (en) * 1984-08-31 1986-01-07 Miles Laboratories, Inc. Nucleic acid hybridization assay employing antibodies to intercalation complexes
US4775619A (en) * 1984-10-16 1988-10-04 Chiron Corporation Polynucleotide determination with selectable cleavage sites
US4743535A (en) * 1984-11-07 1988-05-10 Miles Inc. Hybridization assay employing labeled probe and anti-hybrid
US5641630A (en) * 1985-06-13 1997-06-24 Amgen Inc. Method and kit for performing nucleic acid hybridization assays
US4751177A (en) * 1985-06-13 1988-06-14 Amgen Methods and kits for performing nucleic acid hybridization assays
US4868105A (en) * 1985-12-11 1989-09-19 Chiron Corporation Solution phase nucleic acid sandwich assay
US5750338A (en) * 1986-10-23 1998-05-12 Amoco Corporation Target and background capture methods with amplification for affinity assays
US4883084A (en) * 1987-07-21 1989-11-28 Ti Interlock Limited Pressure fluid control system
US5656731A (en) * 1987-10-15 1997-08-12 Chiron Corporation Nucleic acid-amplified immunoassay probes
US5614362A (en) * 1987-10-15 1997-03-25 Chiron Corporation Nucleic acid hybridization assay for hepatitis B virus DNA
US5082830A (en) * 1988-02-26 1992-01-21 Enzo Biochem, Inc. End labeled nucleotide probe
US6326136B1 (en) * 1988-04-01 2001-12-04 Digene Corporation Macromolecular conjugate made using unsaturated aldehydes
US5374524A (en) * 1988-05-10 1994-12-20 E. I. Du Pont De Nemours And Company Solution sandwich hybridization, capture and detection of amplified nucleic acids
US5527898A (en) * 1988-09-09 1996-06-18 Hoffmann-La Roche Inc. Detection of human papillomavirus by the polymerase chain reaction
US6268128B1 (en) * 1989-03-10 2001-07-31 Vysis, Inc. Immobilized oligonucleotide probes and uses thereof
US5106727A (en) * 1989-04-27 1992-04-21 Life Technologies, Inc. Amplification of nucleic acid sequences using oligonucleotides of random sequences as primers
US5702893A (en) * 1989-06-30 1997-12-30 Chiron Corporation Hydrophobic nucleic acid probe
US5116734A (en) * 1989-09-01 1992-05-26 Digene Diagnostics, Inc. Highly sensitive method for detecting peroxidase
US5580970A (en) * 1989-12-01 1996-12-03 Amoco Corporation Detection of HPV transcripts
US5629153A (en) * 1990-01-10 1997-05-13 Chiron Corporation Use of DNA-dependent RNA polymerase transcripts as reporter molecules for signal amplification in nucleic acid hybridization assays
US5792606A (en) * 1990-02-26 1998-08-11 Boehringer Mannheim Gmbh Nucleic acid hybridization based assay for determining a substance of interest
US5437977A (en) * 1990-04-03 1995-08-01 David Segev DNA probe signal amplification
US5695926A (en) * 1990-06-11 1997-12-09 Bio Merieux Sandwich hybridization assays using very short capture probes noncovalently bound to a hydrophobic support
US5484699A (en) * 1990-09-28 1996-01-16 Abbott Laboratories Nucleotide sequences useful as type specific probes, PCR primers and LCR probes for the amplification and detection of human papilloma virus, and related kits and methods
US5474895A (en) * 1990-11-14 1995-12-12 Siska Diagnostics Inc. Non-isotopic detection of nucleic acids using a polystyrene support-based sandwich hybridization assay and compositions useful therefor
US5256571A (en) * 1991-05-01 1993-10-26 Cytyc Corporation Cell preservative solution
US5543294A (en) * 1991-07-19 1996-08-06 The Trustees Of Columbia University In The City Of New York Polymerase chain reaction/restriction fragment length polymorphism method for the detection and typing of myobacteria
US5556748A (en) * 1991-07-30 1996-09-17 Xenopore Corporation Methods of sandwich hybridization for the quantitative analysis of oligonucleotides
US6228578B1 (en) * 1991-11-14 2001-05-08 Digene Corporation Non-radioactive hybridization assay and kit
US20020012936A1 (en) * 1991-11-14 2002-01-31 Attila Lorincz Non-radioactive hybridization assay and kit
US6027897A (en) * 1991-11-14 2000-02-22 Digene Corporation Promoter-primer mediated nucleic acid amplification
US5981179A (en) * 1991-11-14 1999-11-09 Digene Diagnostics, Inc. Continuous amplification reaction
US5736316A (en) * 1991-12-23 1998-04-07 Chiron Corporation HBV capture and amplifiers probes for use in solution phase sandwich hybridization assays
US5747244A (en) * 1991-12-23 1998-05-05 Chiron Corporation Nucleic acid probes immobilized on polystyrene surfaces
US5424413A (en) * 1992-01-22 1995-06-13 Gen-Probe Incorporated Branched nucleic acid probes
US5629156A (en) * 1992-10-09 1997-05-13 Amoco Corporation Multiple capture probe sandwich assays
US5759773A (en) * 1993-01-15 1998-06-02 The Public Health Research Institute Of The City Of New York, Inc. Sensitive nucleic acid sandwich hybridization assay
US5357977A (en) * 1993-04-23 1994-10-25 St. Mary's Hospital And Medical Center, Inc. Cytological sampling method and device
US5821339A (en) * 1993-05-20 1998-10-13 Dana Farber Cancer Institute Compositions and methods for treatment of herpesvirus infections
US5370128A (en) * 1993-12-02 1994-12-06 Wainwright; Sharon R. Pap brush and pap unit container systems
US5681697A (en) * 1993-12-08 1997-10-28 Chiron Corporation Solution phase nucleic acid sandwich assays having reduced background noise and kits therefor
US5800994A (en) * 1994-04-04 1998-09-01 Chiron Diagnostics Corporation Hybridization-ligation assays for the detection of specific nucleic acid sequences
US5627030A (en) * 1994-06-28 1997-05-06 Kalyx Biosciences Inc. Method of amplification for increasing the sensitivity of detecting nucleic acid-probe target hybrids
US6232462B1 (en) * 1994-08-30 2001-05-15 Bayer Corporation Reduction of nonspecific hybridization by using novel base-pairing schemes
US5728531A (en) * 1994-09-30 1998-03-17 Kabushiki Kaisha Toyota Chuo Kenkyusho Method of detecting nucleic acid
US6057099A (en) * 1994-12-02 2000-05-02 Intelligene Ltd. Detection of nucleic acid sequences
US5747248A (en) * 1994-12-05 1998-05-05 Chiron Corporation Discontinuous probe design using hybritope mapping
US5827661A (en) * 1994-12-23 1998-10-27 Kalyx Biosciences Incorporated Enhancing detection polymerase chain reaction assays by RNA transcription and immunodetection of RNA:DNA hybrids
US5888724A (en) * 1995-02-17 1999-03-30 The Trustees Of Columbia University In The City Of New York Detection of high oncogenic-risk papilloma virus in high grade cervical lesions and cancers by a PCR/ELISA assay
US6083925A (en) * 1995-06-07 2000-07-04 Connaught Laboratories Limited Nucleic acid respiratory syncytial virus vaccines
US6583278B1 (en) * 1995-11-15 2003-06-24 Gen-Probe Incorporated Nucleic acid probes complementary to human papilloma virus nucleic acid
US5731153A (en) * 1996-08-26 1998-03-24 The Regents Of The University Of California Identification of random nucleic acid sequence aberrations using dual capture probes which hybridize to different chromosome regions
US5853993A (en) * 1996-10-21 1998-12-29 Hewlett-Packard Company Signal enhancement method and kit
US6110682A (en) * 1996-10-21 2000-08-29 Agilent Technologies Inc. Signal enhancement method and kit
US6110676A (en) * 1996-12-04 2000-08-29 Boston Probes, Inc. Methods for suppressing the binding of detectable probes to non-target sequences in hybridization assays
US20030108897A1 (en) * 1997-01-16 2003-06-12 Drmanac Radoje T. Methods and compositions for detection or quantification of nucleic acid species
US20030175765A1 (en) * 1997-11-04 2003-09-18 Christoph Kessler Specific and sensitive nucleic acid detection method
US6969585B2 (en) * 1997-12-12 2005-11-29 Digene Corporation Universal collection medium
US6355424B1 (en) * 1997-12-12 2002-03-12 Digene Corporation Assessment of human papillomavirus-related disease
US20030096232A1 (en) * 1997-12-19 2003-05-22 Kris Richard M. High throughput assay system
US6686151B1 (en) * 1998-02-06 2004-02-03 Digene Corporation Immunological detection of RNA:DNA hybrids on microarrays
US5994079A (en) * 1998-02-06 1999-11-30 Digene Corporation Direct detection of RNA mediated by reverse transcriptase lacking RNAse H function
US6277579B1 (en) * 1998-02-06 2001-08-21 Digene Corporation Direct detection of RNA mediated by reverse transcriptase lacking RNAse H function
US6043038A (en) * 1998-03-31 2000-03-28 Tularik, Inc. High-throughput screening assays for modulators of primase activity
US20010055766A1 (en) * 1999-04-02 2001-12-27 Alexander Aristarkhov Immunosorbant assay using branched bis-biotin/avidin/multiple label complex as a detection reagent
US20090032445A1 (en) * 1999-04-29 2009-02-05 Mss, Inc. Multi-Grade Object Sorting System And Method
US6544732B1 (en) * 1999-05-20 2003-04-08 Illumina, Inc. Encoding and decoding of array sensors utilizing nanocrystals
US6890729B2 (en) * 2000-04-04 2005-05-10 Digene Corporation Device and method for cytology slide preparation
US6436662B1 (en) * 2000-04-04 2002-08-20 Digene Corporation Device and method for cytology slide preparation
US7001776B2 (en) * 2000-04-04 2006-02-21 Digene Corporation Device and method for cytology slide preparation
US6521190B1 (en) * 2000-05-19 2003-02-18 Digene Corporation Cell collection apparatus
US6828098B2 (en) * 2000-05-20 2004-12-07 The Regents Of The University Of Michigan Method of producing a DNA library using positional amplification based on the use of adaptors and nick translation
US20060051809A1 (en) * 2000-06-15 2006-03-09 Irina Nazarenko Detection of nucleic acids by target-specific hybrid capture method
US20040214302A1 (en) * 2000-06-15 2004-10-28 James Anthony Detection of nucleic acids by type-specific hybrid capture method
US7371518B2 (en) * 2000-06-21 2008-05-13 Digene Corporation Universal collection medium
US20050147996A1 (en) * 2000-11-21 2005-07-07 Sorge Joseph A. Methods for detection of a nucleic acid by sequential amplification
US6977148B2 (en) * 2001-10-15 2005-12-20 Qiagen Gmbh Multiple displacement amplification
US20060160188A1 (en) * 2005-01-14 2006-07-20 Kurnit David M Systems, methods , and compositions for detection of human papilloma virus in biological samples
US20080200344A1 (en) * 2005-11-15 2008-08-21 Shu-Ling Cheng Protein chips for HPV detection
US20090263819A1 (en) * 2007-02-27 2009-10-22 Nuclea Biomarkers, Llc Gene and protein expression profiles associated with the therapeutic efficacy of egfr-tk inhibitors
US20080247914A1 (en) * 2007-04-06 2008-10-09 Ted Carl Edens Sample Preparation System And Method for Processing Clinical Specimens
US20090298187A1 (en) * 2008-04-17 2009-12-03 Qiagen Gaithersburg, Inc. Compositions, methods, and kits using synthetic probes for determining the presence of a target nucleic acid
US20100081124A1 (en) * 2008-09-30 2010-04-01 Abbott Laboratories Primers and probes for detecting human papillomavirus and human beta globin sequences in test samples

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Reid, R. and Lorincz, A.T. Bailliere's Clinical Obstetrics and Gynaecology 9(1):65 (March 1995). *
Stoler, M.H. et al. Human Pathology 23(2):117 (1992). *

Also Published As

Publication number Publication date
CA2313641A1 (en) 1999-06-24
BR9814271A (en) 2001-03-20
AU1911799A (en) 1999-06-28
WO1999031273A3 (en) 1999-10-07
US7879546B2 (en) 2011-02-01
EP1038029A2 (en) 2000-09-27
US6355424B1 (en) 2002-03-12
NO20002980D0 (en) 2000-06-09
EP1038022A2 (en) 2000-09-27
NO20002980L (en) 2000-08-08
DE69830927T2 (en) 2006-05-24
US6969585B2 (en) 2005-11-29
NO20002979D0 (en) 2000-06-09
JP2002508190A (en) 2002-03-19
US20070154884A1 (en) 2007-07-05
DE69830927D1 (en) 2005-08-25
US20020127545A1 (en) 2002-09-12
JP2010136729A (en) 2010-06-24
PT1038022E (en) 2005-11-30
AU1723299A (en) 1999-07-05
CA2313483A1 (en) 1999-06-17
WO1999031273A2 (en) 1999-06-24
EP1038022B1 (en) 2005-07-20
US20030091992A1 (en) 2003-05-15
NO20002979L (en) 2000-08-09
AU746061B2 (en) 2002-04-11
US7291455B2 (en) 2007-11-06
CA2313483C (en) 2017-12-05
NO330906B1 (en) 2011-08-15
WO1999029890A2 (en) 1999-06-17
ES2246546T3 (en) 2006-02-16
AU748022B2 (en) 2002-05-30
ATE299946T1 (en) 2005-08-15
BR9814272A (en) 2000-10-03
DK1038022T3 (en) 2005-10-24
WO1999029890A3 (en) 1999-09-10
JP2001526045A (en) 2001-12-18

Similar Documents

Publication Publication Date Title
US7879546B2 (en) Assessment of human papilloma virus-related disease
JP5204132B2 (en) Method for detecting human papillomavirus mRNA
JP5773955B2 (en) HPVE6, E7 mRNA assay and methods of use thereof
US8227190B2 (en) Approach to molecular diagnosis of human papillomavirus-related diseases
Narimatsu et al. High-throughput cervical cancer screening using intracellular human papillomavirus E6 and E7 mRNA quantification by flow cytometry
Williams et al. Molecular detection methods in HPV-related cancers
JP2007524421A (en) Detection of human papillomavirus
US20130196308A1 (en) Multiparameter assay
US20070037137A1 (en) Method and kit for quantitative and qualitative determination of human papillomavirus
US20060134615A1 (en) Method for the simultaneous detection of HPV RNA and DNA in a sample
AU2002300070B2 (en) Assessment of Human Papilloma Virus-Related disease
WO2009011472A1 (en) Primers and proves for detection of high risk group geno-type human papillomavirus dna, a qualitative assay method of the same dna using them and a qualitative assay kit of the same dna
Abdulhamit et al. FDA-Approved Molecular Tests Used to Define Human Papillomavirus (HPV) Infections which Cause Cervix Cancer
Li et al. HPV Testing and Molecular Biomarkers in Cervical Cytology
Feng et al. Human Papillomavirus and its role in cervical carcinoma
Enache et al. Human papillomaviruses and cervical cancer
Rosini et al. Management and triage of women in follow-up for low-grade cervical disease: association of DNA and RNA-based methods

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION