WO2000020641A1 - Tissue analysis and kits therefor - Google Patents

Tissue analysis and kits therefor Download PDF

Info

Publication number
WO2000020641A1
WO2000020641A1 PCT/US1999/022909 US9922909W WO0020641A1 WO 2000020641 A1 WO2000020641 A1 WO 2000020641A1 US 9922909 W US9922909 W US 9922909W WO 0020641 A1 WO0020641 A1 WO 0020641A1
Authority
WO
WIPO (PCT)
Prior art keywords
nucleic acid
tissue sample
section
probe
tissue
Prior art date
Application number
PCT/US1999/022909
Other languages
French (fr)
Inventor
Robert L. Cohen
Mary Beth Gardiner
Mark X. Sliwkowski
Gregory T. Stelzer
Original Assignee
Genentech, Inc.
Cytometry Associates
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Genentech, Inc., Cytometry Associates filed Critical Genentech, Inc.
Priority to AU64097/99A priority Critical patent/AU6409799A/en
Publication of WO2000020641A1 publication Critical patent/WO2000020641A1/en

Links

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
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • C12Q1/6841In situ hybridisation
    • 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
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57415Specifically defined cancers of breast
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • G01N33/57492Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites involving compounds localized on the membrane of tumor or cancer cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/58Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances
    • G01N33/582Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances with fluorescent label
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6872Intracellular protein regulatory factors and their receptors, e.g. including ion channels
    • 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
    • C12Q1/6804Nucleic acid analysis using immunogens
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/71Assays involving receptors, cell surface antigens or cell surface determinants for growth factors; for growth regulators

Definitions

  • the present invention relates generally to the field of tissue analysis. Specifically, the invention combines morphological staining and/or immunohistochemistry (IHC) with fluorescence in situ hybridization (FISH) within the same section of a tissue sample thereby allowing for accurate and simplified prognostic, diagnostic, or research applications on a subject's tissue sample.
  • the invention provides kits for analysis of a tissue sample utilizing the present methods.
  • FISH Fluorescence in situ hybridization
  • Immunohistochemical staining of tissue sections has been shown to be a reliable method of assessing alteration of proteins in a heterogeneous tissue.
  • Immunohistochemistry (IHC) techniques utilize an antibody to probe and visualize cellular antigens in situ, generally by chromagenic or fluorescent methods. This technique excels because it avoids the unwanted effects of disaggregation and allows for evaluation of individual cells in the context of morphology. In addition, the target protein is not altered by the freezing process.
  • IHC Immunohistochemistry
  • the HER2/neu gene encodes a protein product, often identified as pi 85 .
  • the native pi 85 protein is a membrane receptor-like molecule with homology to the epidermal growth factor receptor (EGFR).
  • EGFR epidermal growth factor receptor
  • HER2 overexpression was determined using Southern and Northern blotting which correlate with the HER2 oncoprotein expression evaluated by Western blotting and IHC [Slamon et al. Science 235:177 (1987); Slamon et al. Science 244:707 (1989)].
  • the present invention combines cellular morphological analysis with fluorescence in situ hybridization (FISH) to provide for a correlation of genetic abnormalities and cellular morphology within the same section of a subject's tissue sample. Accordingly, one may identify and score by FISH cancer cells (e.g. invasive ductal carcinoma cells) as distinct from other normal cells (e.g. stromal and inflammatory elements found in the biopsy). Alternatively, or additionally, the invention combines immunohistochemistry (IHC) with FISH to provide for a correlation of genetic abnormalities with protein expression in the same tissue section.
  • FISH cancer cells e.g. invasive ductal carcinoma cells
  • IHC immunohistochemistry
  • Morphologic assessment, or evaluation of protein expression, in a tissue prior to quantitative FISH analysis provides for accurate, specific evaluation of that tissue in a timely and cost-efficientmanner.
  • the invention provides a method of correlating cellular morphology with the presence of a cellular target nucleic acid sequence in a section of a tissue sample comprising the following steps:
  • step (e) correlating step (b) with step (d).
  • the invention pertains to a method of correlating the presenceof a cellular target protein with the presence of a cellular target nucleic acid sequence in a section of a tissue sample comprising the following steps:
  • step (e) correlating step (b) with step (d).
  • kits comprising: (a) a primary antibody which specifically binds a cellular target protein; (b) a fluorescently labeled probe complementary to a genetic abnormality; and (c) instructions for applying the antibody (a) and probe (b) to the same section of tissue sample.
  • FIGURES Figure 1 shows the effect of HEMA 3® staining on the ability to score FISH in formalin fixed, paraffin- embedded cells harvested from each of three cell lines of known HER2/neu amplification status; SKBR3 (highly amplified HER2), MDA175 (barely amplified HER2), and MDA231 (non-amplified HER2).
  • Figure 2 shows the effect of HEMA 3® mo ⁇ hologic stain on FISH with respect to tumor biopsy sections.
  • Duplicate sections from 13 tumor specimens were analyzed for HER2 and chromosome 17 (Chr 17) by FISH either with or without prior staining for mo ⁇ hologic analysis prior to FISH. The same area of tumor was scored for FISH on each section.
  • Figure 3 shows the mean HER2/neu:Ch ⁇ 17 ratio for blinded vs non-blinded assessment of normal tissue.
  • Figure 4 shows the mean HER2/neu:Chr 17 ratio for blinded vs non-blinded assessment of tumor tissue.
  • Figure 5 shows the relationship between the scoring systems used for HER2lneu amplification by FISH and overexpression by IHC.
  • subject or “patient” is meant any single subject for which therapy is desired, including humans, cattle, dogs, guinea pigs, rabbits, chickens, insects and so on. Also intended to be included as a subject are any subjects involved in clinical research trials not showing any clinical sign of disease, or subjects involved in epidemiological studies, or subjects used as controls.
  • the tissue sample may contain compounds which are not naturally intermixed with the tissue in nature such as preservatives, anticoagulants, buffers, fixatives, nutrients, antibiotics, or the like.
  • the tissue sample is "non-hemato logic tissue" (i.e. not blood or bone marrow tissue).
  • disease gene is meant a gene that results in altered protein product (i.e., protein different from native protein in terms of sequence, structure and/or amount expressed) and results in a disease.
  • mo ⁇ hological stain is meant a dye that stains different cellular components, in order to facilitate identification of cell type and/or disease status by light microscopy.
  • the mo ⁇ hological stain is readily distinguishable from any label used in the FISH analysis, e.g., a stain which will not autofluoresce at the same wavelength as the fluorochrome used in the FISH analysis.
  • the term "monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to conventional (polyclonal) antibody preparations that typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen.
  • the modifier "monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies herein specifically include "chimeric" antibodies (immunoglobulins) in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity [U.S. Patent No. 4,816,567; and Morrison et al, Proc. Natl. Acad. Sci. USA 81 :6851-6855 (1984)].
  • chimeric antibodies immunoglobulins in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in
  • HER2 Unless indicated otherwise, the terms "HER2", “pi 85 " and “ErbB2” when used herein refer to human HER2 protein or a portion thereof and “HER2 " , “HER2/neu “ and “c-er/>B2” refer to the human HER2 gene or a portion thereof.
  • the human HER2 gene and HER2 protein are, for example, described in Semba et al, PNAS (USA) 82:6497-6501 (1985) and Yamamoto et al. Nature 319:230-234 (1986) (Genebank accession number X03363).
  • the present invention relates to a method which combines mo ⁇ hological staining and/or immunohistochemical analysis with fluorescence in situ hybridization (FISH) within the same section of a tissue sample.
  • FISH fluorescence in situ hybridization
  • the present methods allow for a highly accurate and simplified method of correlating mo ⁇ hological criteria or protein expression with genetic abnormalities. Because many tissue types, such as breast tissue, are characterized by significant cellular heterogeneity, inaccurate results may be obtained when serial sections from a tissue block are analyzed by two different methods. According to the present invention, both types of analysis are carried out on the same tissue section, thereby reducing error when analyzing such heterogeneous tissue. As evidenced by the following, the method of this application is useful in a variety of prognostic, diagnostic and research applications. Also disclosed are kits for use in the disclosed methods.
  • tissue sample from a subject may be used.
  • tissue samples that may be used include, but are not limited to, breast, prostate, ovary, colon, lung, endometrium, stomach, salivary gland or pancreas.
  • the tissue sample can be obtained by a variety of procedures including, but not limited to surgical excision, aspiration or biopsy.
  • the tissue may be fresh or frozen.
  • the tissue sample is fixed and embedded in paraffin or the like.
  • the tissue sample may be fixed (i.e. preserved) by conventional methodology [See e.g., "Manual of Histological Staining Method of the Armed Forces Institute of Pathology," 3 rd edition (1960) Lee G. Luna, HT (ASCP) Editor, The Blakston Division McGraw-Hill Book Company, New York; The Armed Forces Institute of Pathology Advanced Laboratory Methods in Histology and Pathology (1994) Ulreka V. Mikel, Editor, Armed Forces Institute of Pathology, American Registry of Pathology, Washington, D.C.].
  • a fixative is determined by the pu ⁇ ose for which the tissue is to be histologically stained or otherwise analyzed.
  • the sample may be sectioned by a microtome or the like (See e.g., "Manual of Histological Staining Method of the Armed Forces Institute of Pathology", supra). By way of example for this procedure, sections may range from about three microns to about five microns in thickness.
  • the sections may be attached to slides by several standard methods. Examples of slide adhesives include, but are not limited to, silane, gelatin, poly-L-lysine and the like.
  • the paraffin embedded sections may be attached to positively charged slides and/or slides coated with poly-L-lysine.
  • the sections mounted on slides may be stained with a mo ⁇ hological stain for evaluation.
  • the mo ⁇ hological stain to be used in the instant method provides for accurate mo ⁇ hological evaluation of a tissue section and also allows for accurate quantification of fluorescently labeled (e.g., SPECTRUM ORANGE® and SPECTRUM GREEN®) nucleic acid probes when the sections are subsequently processed for FISH.
  • fluorescently labeled e.g., SPECTRUM ORANGE® and SPECTRUM GREEN®
  • Many mo ⁇ hological stains fluoresce when illuminated by light of the same wavelength required to visualize the probe fluorophore of interest. The disclosed method solves this problem.
  • the section is stained with one or more dyes each of which distinctly stains different cellular components.
  • xanthine dye or the functional equivalent thereof and/or a thiazine dye or the functional equivalent thereof are used to enhance and make distinguishable the nucleus, cytoplasm, and "granular" structures within each.
  • Such dyes are commercially available and often sold as sets.
  • HEMA 3® (CMS, Houston, Texas) stain set comprises xanthine dye and thiazine dye. Methyleneblue may also be used.
  • Examples of other mo ⁇ hological stains that may be used on the instant method include, but are not limited to, dyes that do not significantly autofluoresce at the same wavelength as fluorescent label(s) used for the subsequent FISH analysis.
  • the tissue section may be analyzed by standard techniques of microscopy.
  • a pathologist or the like assesses the tissue for the presence of abnormal or normal cells or a specific cell type and provides the loci of the cell types of interest.
  • a pathologist or the like would review the slides and identify normal breast cells and abnormal breast cells (See e.g. Example 2).
  • any means of defining the loci of the cells of interest may be used (e.g., coordinates on an X-Y axis).
  • the slides are destained by conventional methodology.
  • the present method provides an advantage over the prior procedures in the art in that no separate destaining procedure is required prior to FISH. Avoidance of a destaining step is actually preferred in order to protect the integrity of the DNA for in situ hybridization.
  • the primary and/or secondary antibody used for immunohistochemistry typically will be labeled with a detectable moiety.
  • Numerous labels are available which can be generally grouped into the following categories:
  • Fluorescent labels including, but are not limited to, rare earth chelates (europium chelates), Texas Red, rhodamine, fluorescein, dansyl, Lissamine, umbelliferone, phycocrytherin, phycocyanin, or commercially available fluorophores such SPECTRUM ORANGE® and SPECTRUM GREEN® and/or derivatives of any one or more of the above.
  • the fluorescent labels can be conjugated to the antibody using the techniques disclosed in Current Protocols in Immunology, supra, for example. Fluorescence can be quantified using a fluorimeter.
  • HRPO Horseradish peroxidase
  • OPD orthophenylene diamine
  • TMB 3,3',5,5'-tetramethyl benzidine hydrochloride
  • the label is indirectly conjugated with the antibody.
  • the antibody can be conjugated with biotin and any of the four broad categories of labels mentioned above can be conjugated with avidin, or vice versa. Biotin binds selectively to avidin and thus, the label can be conjugated with the antibody in this indirect manner.
  • the antibody is conjugated with a small hapten and one of the different types of labels mentioned above is conjugated with an anti-hapten antibody.
  • indirect conjugation of the label with the antibody can be achieved.
  • tissue section prior to, during or following IHC may be desired.
  • epitope retrieval methods such as heating the tissue sample in citrate buffer may be carried out [see, e.g., Leong et al. Appl Immunohistochem. 4(3):201 (1996)].
  • the tissue section is exposed to primary antibody for a sufficient period of time and under suitable conditions such that the primary antibody binds to the target protein antigen in the tissue sample. Appropriate conditions for achieving this can be determined by routine experimentation.
  • the extent of binding of antibody to the sample is determined by using any one of the detectable labels discussed above.
  • the label is an enzymatic label (e.g.
  • HRPO which catalyzes a chemical alteration of the chromogenic substrate such as 3,3'-diaminobenzidinechromogen.
  • the enzymatic label is conjugated to antibody which binds specifically to the primary antibody (e.g. the primary antibody is rabbit polyclonal antibody and secondary antibody is goat anti-rabbit antibody).
  • Specimens thus prepared may be mounted and coverslipped. Slide evaluation is then determined, e.g. using a microscope. Where the antigen is HER2 protein, staining intensity criteria may be evaluated as follows: TABLE 1 HER2 Protein Staining Intensity Criteria
  • tissue samples with 1+ and 2+ scores, and especially, the 2+ subgroup may be subjected to further FISH as explained below.
  • In situ hybridization is generally carried out on cells or tissue sections fixed to slides. In situ hybridization may be performed by several conventional methodologies [See for e.g. Leitch et al. In Situ Hybridization: a practical guide, Oxford BIOS Scientific Publishers, Micropscopy handbooks v. 27 (1994)].
  • fluorescent dyes such as fluorescein isothiocyanate (FITC) which fluoresces green when excited by an Argon ion laser
  • FITC fluorescein isothiocyanate
  • Each cell containing the target nucleotide sequence will bind the labeled probe producing a fluorescent signal upon exposure, of the cells to a light source of a wavelength appropriate for excitation of the specific fluorochrome used.
  • hybridization stringency can be employed. As the hybridization conditions become more stringent, a greater degree of complementarity is required between the probe and target to form and maintain a stable duplex. Stringency is increased by raising temperature, lowering salt concentration, or raising formamide concentration. Adding dextran sulfate or raising its concentration may also increase the effective concentration of labeled probe to increase the rate of hybridization and ultimate signal intensity. After hybridization, slides are washed in a solution generally containing reagents similar to those found in the hybridization solution with washing time varying from minutes to hours depending on required stringency. Longer or more stringent washes typically lower nonspecific background but run the risk of decreasing overall sensitivity. Exemplary in situ hybridization conditions for detecting HERllneu amplification in breast tissue are shown in Example 2.
  • Probes used in the FISH analysis may be either RNA or DNA oligonucleotides or polynucleotides and may contain not only naturally occurring nucleotides but their analogs like digoxygenin dCTP, biotin dcTP 7- azaguanosine, azidothymidine, inosine, or uridine.
  • Other useful probes include peptide probes and analogues thereof, branched gene DNA, peptidometics, peptide nucleic acid (PNA) and/or antibodies. Probes should have sufficient complementarity to the target nucleic acid sequence of interest so that stable and specific binding occurs between the target nucleic acid sequence and the probe.
  • the degree of homology required for stable hybridization varies with the stringency of the hybridization medium and/or wash medium.
  • completely homologous probes are employed in the present invention, but persons of skill in the art will readily appreciate that probes exhibiting lesser but sufficient homology can be used in the present invention [see for e.g. Sambrook, J., Fritsch, E.F., Maniatis, T., Molecular Cloning A Laboratory Manual, Cold Spring Harbor Press, (1989)].
  • Genetic abnormalities that can be detected by this method include, but are not limited to, amplification, translocation, deletion, addition and the like.
  • Examples of amplification include, but are not limited to, HER2lneu in breast and ovarian cancer, ⁇ N-myc in neuroblastoma, C-myc in small cell lung cancer.
  • Examples of abnormal chromosome number include, but are not limited to, trisomy 8 in leukemia, monosomy 7 in myloproliferative disorders, and trisomy 12 in chronic lymphoblastic leukemia.
  • translocations include, but are not limited to, bcr/abl [t (9;22)] translocation in chronic mylogenous leukemia and the t ( 15; 17) translocation FAB-M3 (acute promyelocytic leukemia).
  • deletions include EGFR vIII and p53.
  • a probe spanning a 140 kb region on the long arm of chromosome 17 containing the HER2lneu gene (17q 1 1.2-17ql2) may be used.
  • a probe for the -satellite sequences in the centromeric region of chromosome 17(D1721 ) may be used to evaluate for aneusomy of chromosome 17 as a source or cause for HERllneu amplification.
  • a cocktailed version of these probes may be obtained from Vysis, Inc. where each probe is directly labeled with easily distinguishable fluorophores, such as SPECTRUM ORANGE® and SPECTRUM GREEN®.
  • Probes may also be generated and chosen by several means including, but not limited to, mapping by in situ hybridization, somatic cell hybrid panels, or spot blots of sorted chromosomes; chromosomal linkage analysis; or cloned and isolated from sorted chromosome libraries from human cell lines or somatic cell hybrids with human chromosomes, radiation somatic cell hybrids, microdissection of a chromosome region, or from yeast artificial chromosomes (YACs) identified by PCR primers specific for a unique chromosome locus or other suitable means like an adjacent YAC clone.
  • mapping by in situ hybridization, somatic cell hybrid panels, or spot blots of sorted chromosomes chromosomal linkage analysis
  • cloned and isolated from sorted chromosome libraries from human cell lines or somatic cell hybrids with human chromosomes, radiation somatic cell hybrids, microdissection of a chromosome region, or from yeast artificial chromos
  • Probes may be genomic DNA, cDN A, or RNA cloned in a plasmid, phage, cosmid, YAC, Bacterial Artificial Chromosomes (BACs), viral vector, or any other suitable vector. Probes may be cloned or synthesized chemically by conventional methods. When cloned, the isolated probe nucleic acid fragments are typically inserted into a vector, such as lambda phage, pBR322, M13, or vectors containing the SP6 or T7 promoter and cloned as a library in a bacterial host. [See for e.g. Sambrook, J ., Fritsch, E.F., Maniatis, T., Molecular Cloning A Laboratory Manual, Cold Spring Harbor Press, (1989)].
  • Probes are preferably labeled with a fluorophor.
  • fluorophores include, but are not limited to, rare earth chelates (europium chelates), Texas Red, rhodamine, fluorescein, dansyl, Lissamine, umbelliferone, phycocrytherin, phycocyanin, or commercially available fluorophors such SPECTRUM ORANGE® and SPECTRUM GREEN® and/or derivatives of any one or more of the above.
  • Multiple probes used in the assay may be labeled with more than one distinguishablefluorescent or pigment color. These color differences provide a means to identify the hybridization positions of specific probes.
  • Probes can be labeled directly or indirectly with the fluorophor, utilizing conventional methodology.
  • HERllneu gene is in chromosome 17, and as an internal control a probe for -satellite sequences specific for chromosome 17 (D17Z1 ) may be used (Vysis Inc.) to prove diploidy in areas of non-malignant cells and/or to establish the presence or absence of chromosome 17 aneusomy in areas of HER2/neu amplification.
  • the slides may be analyzed by standard techniques of fluorescence microscopy [see for e.g. Ploem and Tanke Introduction to Fluorescence Microscopy, New York, Oxford University Press ( 1987)]. Briefly, each slide is observed using a microscope equipped with appropriate excitation filters, dichromic, and barrier filters. Filters are chosen based on the excitation and emission spectra of the fluorochromes used. Photographs of the slides may be taken with the length of time of film exposure depending on the fluorescent label used, the signal intensity and the filter chosen. For FISH analysis the physical loci of the cells of interest determined in the mo ⁇ hological analysis are recalled and visually conformed as being the appropriate area for FISH quantification.
  • a computer-driven, motorized stage which stores location of co-ordinates. This may be used to evaluate the same area by two different analytical techniques. For example, color images of the mo ⁇ hologically stained areas may be captured and saved using a computer-assistedcooled CCD camera. The same section may be subsequently taken through the FISH procedure, the stored locations recalled, and the designated areas scored for the presence of fluorescent nuclear signals. A similar procedure for IHC followed by FISH is contemplated.
  • Deviation of the number of spots in a cell from a norm may be indicative of a malignancy or a predisposition to a malignancy, disease, or other abnormality.
  • the relative number of abnormal cells to the total cell sample population may also indicative of the extent of the condition or abnormality.
  • using family health histories and/or genetic screening it is possible to estimate the probability that a particular subject has for developing certain types of cancer.
  • Those subjects that have been identified as being predisposed to developing a particular form of cancer can be monitored or screened to detect early evidence of disease. Upon discovery of such evidence, early treatment can be undertaken to combat the disease. Similarly, those subjects who have already developed a malignancy and who have been treated to remove the cancer or are otherwise in remission are particularly susceptible to relapse and reoccurrence, including the metastasis of tumors. Such subjects can be monitored and screened using the presently disclosed methods to detect evidence of metastasis and upon discovery of such evidence, immediate treatment can be undertaken to combat the disease.
  • this invention provides a kit comprising a fluorescently labeled probe specific for the target nucleic acid sequence of interest and a mo ⁇ hological stain and/or an antibody which specifically binds target antigen.
  • the kit further comprises a set of instructions for applying the stain or antibody and probe to the same section of a tissue sample.
  • the fluorescently labeled probe may be complementary to the HERllneu gene and the mo ⁇ hological stain may be HEMA 3® (CMS, Houston, Texas). Any fluorescent label as described above may be used to label the probe.
  • the IHC/FISH kit may comprise a fluorescently labeled probe complementary to the HER2/neu gene and an antibody (e.g.
  • the kit may include both a primary and secondary antibody, wherein the secondary antibody is conjugated to a label, e.g., an enzymatic label.
  • the invention also provides an IHC kit which has instructions to perform FISH on the same section of tissue sample on which IHC has been previously performed.
  • kits include one or more buffers (e.g., block buffer, wash buffer, substrate buffer, etc), other reagents such as substrate (e.g., chromagen) which is chemically altered by an enzymatic label, epitope retrieval solution, control samples (positive and/or negative controls), control slide(s) etc.
  • buffers e.g., block buffer, wash buffer, substrate buffer, etc
  • substrate e.g., chromagen
  • a sensitive and specific evaluation of breast tumors for the amplification of HER2lneu by FISH requires definitive identification and scoring of invasive ductal carcinoma cells as distinct from other stromal and inflammatory elements found in the biopsy. Thus, it was necessary to identify a stain which would allow for complete mo ⁇ hological evaluation of the tissue, and which also allowed for easy quantification of nuclear signals upon subsequent hybridization with fluorescently labeled nucleic acid probes.
  • Many mo ⁇ hological stains fluoresce when illuminated by light of the same wavelength required to visualize the probe fluorophores of interest. When this autofluorescenceis of a color similarto that of the probe fluorophore, signal quantification is made difficult. Results from the evaluation of six commonly used mo ⁇ hological stains may be found in Table 2.
  • Methyl green pyronin (MGP) and Gill hematoxylin were not analyzed further due to poor mo ⁇ hological definition.
  • Paragon toluidine blue/basic fuchsin
  • Weigert were eliminated from consideration because of an unacceptably high level of red or orange autofluorescence which masked the orange HERl/neu signals in the nucleus.
  • Methylene blue yielded acceptable mo ⁇ hological staining, but demonstrated orange autofluorescence which caused moderate difficulty in visualizing HER2/neu signals. This stain was considered a possible candidate.
  • HEMA 3® was superior in that it provided good mo ⁇ hological detail and showed only modest dim green autofluorescence which did not interfere to any extent with FISH evaluation.
  • HEMA 3® An initial attempt at destaining HEMA 3® prior to FISH was found to damage the integrity of the DNA in the cells and ultimately proved to be unnecessary anyway. It was possible for the stained tissue to be taken directly through the FISH procedure without intermediate processing, yielding high-quality nuclear signals without any negative consequence. HEMA 3®, therefore, met all essential requirements and was the stain of choice for the proposed system. Unless otherwise indicated, the tissues and FISH analysis were conducted as discussed in Example 2.
  • HEMA 3® Solution I CMS, Houston, TX
  • HEMA 3® Solution II HEMA 3® Solution II
  • aqueous-based mounting medium Biomeda Co ⁇ oration, Foster City, CA
  • the coverglass was removed by placing the slide in a water-filled Coplin jar, which had been pre-heated in a 73+2°C waterbath, for approximately 15-30 minutes, or until the coverglass slipped off.
  • the section was dehydrated in 70% EtOH for 3 minutes, 85% EtOH for 3 minutes, and 100% EtOH for 3 minutes.
  • These slides, and any deparaffinized sections that were not stained but were to be processed by FISH, were immersed in a sodium isothiocyanate Pretreatment Solution (Vysis, Inc., Downers Grove, IL) for 30 minutes at 80+l°C.
  • the sections were washed in deionized water for one minute, then in Protease Wash Buffer (2XSSC, pH 7.4, Vysis, Inc.) for 5 minutes at room temperature, x2.
  • the sections were treated in Protease Solution (Vysis, Inc.) for 10 minutes at 37°C. Following protease digestion, the sections were washed in Protease Wash Buffer for 5 minutes, x2, and dried on a 45-50°C slide warmer for 2-5 minutes.
  • Chromosome 17 aneusomy of the cell was evaluated using an -satellite sequence probe (D17Z1) specific for the centromere of chromosome 17 (CEP 17).
  • D17Z1 -satellite sequence probe specific for the centromere of chromosome 17
  • the pre-treated (see above) slide-mounted sections were immersed in Denaturation Solution (70% formamide/2xSSC, pH 7.0) for 5 minutes at 73+l °C. Sections were then dehydrated in 70% EtOH for one minute, 85% EtOH for one minute, and 100% EtOH for one minute. Drained slides were placed on a 45-50°C slide warmer for 2-5 minutes just before application of the hybridization mixture. This mixture was prepared as directed by VYSIS® protocol. The probe solution was denatured by incubation in a 73°C waterbath for 5 minutes. Following brief centrifugation, 10 L of the solution was pipetted onto each section, a coverslip applied, and the edges sealed with rubber cement.
  • Denaturation Solution 70% formamide/2xSSC, pH 7.0
  • Hybridization was carried out in a humidified box overnight (14-18 h) in a 37°C incubator. The rubber cement was carefully removed and the slides immersed in room temperature Wash Buffer I (0.4xSSC/0.3% NP-40) until the coverslip floated off. Slides were drained of excess liquid by blotting the edges on a paper towel, then placed in a Coplin jar of Wash Buffer I, which had been prewarmed to 72+1 °C, for 2 minutes. Sections were washed in room temperature Wash Buffer II (2xSSC/0.1 %NP-40) for one minute, drained vertically on paper towels, and allowed to air-dry in darkness being careful not to overdry.
  • Wash Buffer II 2xSSC/0.1 %NP-40
  • DAPI counterstain a mixture of equal parts DAPI I and DAPI II, Vysis, Inc.
  • a coverglass was added and the edges sealed with clear nail polish.
  • Slides were analyzed immediately or stored in the dark at -20°C.
  • Controls Cultured cell lines SKBR3, MDA175, and MDA231 were harvested, formalin-fixed, and paraffin- embedded for use as highly amplified, barely amplified, and non-amplified controls, respectively. These cell lines were used in the evaluation of the mo ⁇ hological stains. The presence of appropriate nuclear FISH signals served to assure that the reagents and procedure involved in the devised methodology worked properly.
  • Microscopy An Olympus BX60 epifluorescence microscope equipped with a 100 watt mercury-arc lamp, a
  • the filter slider was built by Chroma Technology Co ⁇ oration (Brattleboro, VT) for Olympus and included three single band pass filters optimized for visualization of DAPI, FITC, and Texas Red fluorochromes, a dual-band pass filter optimized for FITC and Texas Red, and a triple-band pass filter optimized for DAPI, FITC, and Texas Red.
  • the microscope was also equipped with 4x, 1 Ox, 20x, and 40x UPlanFl objectives for use in transmittance light microscopy.
  • a BioPoint X, Y (Ludl Electronic Products, Ltd., Hawthorne, NY) computer-driven motorized stage was used for storage and retrieval of physical locations. Images were acquired using a CoolCam 2000 3-chip color cooled CCD camera (Sci-Measure, Atlanta, GA) and printed using a Tektronix Phaser 440 dye sublimation printer (Tektronix, Inc., Wilsonville, OR). FISH scoring criteria
  • tissue sections were scanned using the 40x objective to evaluate for tissue loss and to ensure that hybridization was uniform across the section.
  • Mo ⁇ hologically identified areas of normal or tumor tissue were located on FISH processed slides by recalling stored x and y coordinates. Alternatively, if location coordinates had not been stored, a saved mo ⁇ hological image (HEMA 3® stain) of the area of interest was used as a reference to localize the appropriate area after FISH processing.
  • Signals were enumerated using the lOOx objective, switching between different filters to optimize signal discrimination. Nuclear boundaries were defined by DAPI excitation. Only those nuclei were scored which could be clearly identified as intact and non-overlapping.
  • Sections cut serially from those used for FISH analysis were immunohistochemically stained using a murine monoclonal antibody against HER2 [4D5; Fendly et al Cancer Research 50: 1550-1558 (1990)].
  • Slide- mounted sections were placed into xylenes 3x, 10 minutes each time, then dehydrated in absolute ethanol 2x, 5 minutes each. Endogenous peroxidase activity was quenched by placing the slides in a 0.3% solution of hydrogen peroxide in methanol. The sections were then serially rehydrated in 95% ethanol for 5 minutes, followed by 80% ethanol for 5 minutes. Sections were incubated briefly in the buffer routinely used with the Ventana ES automated immunostaining instrument.
  • the staining program began with a 4 minute protease treatment. Incubation with the primary antibody (lO ⁇ g/ml) proceeded for the maximum allowable time period, 32 minutes.
  • the detection system employed a biotin/avidin reaction using DAB and a hematoxylin counterstain. After staining and detection, the sections were dehydrated in a graded series of ethanol into xylenes in preparation for permanent coverslipping. mmunohistochemistry scoring criteria
  • the immunohistochemical staining was inte ⁇ reted by a breast tumor pathologist and the results assigned to one of four categories defined in the following way: negative/weak, cytoplasmic, 1+, 2+.
  • the second category was reserved for those cells in which staining occurred only in the cytoplasm.
  • the latter two categories applied to cell surface antibody staining. Images of immunostained sections representative of each category were captured using the CCD camera. Attempts were made to relocate areas that had been scored for FISH on the corresponding serial section, and whenever possible that image was captured. Specimens
  • HEMA 3® stain was evaluated in formalin-fixed, paraffin-embeddedcells harvested from each of three cell-lines of known HERllneu amplification status ( Figure 1). Results from analysis by two individuals were comparable and successfully identified the different cell-lines as being of the appropriate, expected amplification status. The stain had no adverse effect on FISH quantification in these cell lines. A total of 10 patient specimens was included in the study comparing stained and hybridized tissue versus tissue which was hybridized without prior staining. The same areas of tumor and normal cells identified on each of two serial sections was analyzed for the presence of HER2lneu and chromosome 17 by FISH either with or without prior mo ⁇ hological staining.
  • a normal range creates a threshold for determining amplification status for HER2lneu.
  • correlation between normal values derived from non-diseased and diseased tissue provides justification for use of a mo ⁇ hologicallynormal cellular area in a tumorbiopsy as an internal biological control for specificity in that specimen.
  • HER2lneu and chromosome 17 fluorescent nuclear signals were scored by two analysts and the mean ratios calculated. Ratios for HER2lneu amplification were assigned to one of two ranges, either moderately amplified borderline or highly amplified. The lower limit of the moderate amplificationrange (1.3) was established statistically by determining at what value the ratio differed significantly from normal.

Abstract

This invention relates to methods of analyzing a tissue sample from a subject. In particular, the invention combines morphological staining and/or immunohistochemistry (IHC) with fluorescence in situ hybridization (FISH) within the same section of a tissue sample. The analysis can be automated or manual. The invention also relates to kits for use in the above methods.

Description

TISSUE ANALYSIS AND KITS THEREFOR FIELD OF THE INVENTION
The present invention relates generally to the field of tissue analysis. Specifically, the invention combines morphological staining and/or immunohistochemistry (IHC) with fluorescence in situ hybridization (FISH) within the same section of a tissue sample thereby allowing for accurate and simplified prognostic, diagnostic, or research applications on a subject's tissue sample. In addition, the invention provides kits for analysis of a tissue sample utilizing the present methods.
BACKGROUND OF THE INVENTION Advancements in the understanding of genetics and developments in technology and epidemiology have allowed for the correlation of genetic abnormalities with certain malignancies as well as risk assessment of an individual for developing certain malignancies. However, most of the methodologies available for evaluation of tissue for the presence of genes associated with or predisposing an individual to a malignancy have well- known drawbacks. For example, methods that require disaggregation of the tissue, such as Southern, Northern, or Western blot analysis, are rendered less accurate by dilution of the malignant cells by the normal or otherwise non-malignant cells that are present in the same tissue. Furthermore, the resulting loss of tissue architecture precludes the ability to correlate malignant cells with the presence of genetic abnormalities in a context that allows morphological specificity. This issue is particularly problematic in tissue types known to be heterogeneous, such as in human breast carcinoma, where a significant percentage of the cells present in any area may be non-malignant. Fluorescence in situ hybridization (FISH) is a recently developed method for directly assessing the presence of genes in intact cells. FISH is an attractive means of evaluating paraffin-embedded tissue for the presence of malignancy because it provides for cell specificity, yet overcomes the cross-linking problems and other protein-altering effects caused by formalin fixation. FISH has historically been combined with classical staining methodologies in an attempt to correlate genetic abnormalities with cellular morphology [see e.g., Anastasi et al., Blood 11 '.2456-2462 (1991); Anastasi et al., Blood 79: 1796-1801 (1992); Anastasi et al., Blood 81 :1580-1585 (1993); van Lorn et al, Blood 82:884-888 (1993); ohnan et al., Diagnostic Molecular Pathology 1(3): 192-199 (1992); Zitzelberger, Journal of Pathology 172:325-335 (1994)]. However, several of these studies address hematological disorders where genetic changes are assessed in freshly fixed smears from bone marrow aspirates or peripheral blood specimens. Of those two studies where paraffin-embedded tissue was analyzed, one involved evaluation of FISH and morphological staining on separate, serial sections. In the other study, both procedures were performed on the same section, but morphological staining was subsequent to evaluation by FISH. Use of serial sections in this type of analysis increases the probability of error, especially in heterogeneous tissue such as breast tissue.
Immunohistochemical staining of tissue sections has been shown to be a reliable method of assessing alteration of proteins in a heterogeneous tissue. Immunohistochemistry (IHC) techniques utilize an antibody to probe and visualize cellular antigens in situ, generally by chromagenic or fluorescent methods. This technique excels because it avoids the unwanted effects of disaggregation and allows for evaluation of individual cells in the context of morphology. In addition, the target protein is not altered by the freezing process. HFR2 HER2
The HER2/neu gene encodes a protein product, often identified as pi 85 . The native pi 85 protein is a membrane receptor-like molecule with homology to the epidermal growth factor receptor (EGFR). Amplification and overexpressionof HER2 in human breast cancer has been correlated with shorter disease-free interval and shorter overall survival in some studies [van de Vijver et al. New Eng. J. Med. 319:1239(1988); Walker et al. Br. J. Cancer 60:426( 1989); Tandon et al. J. Clin. Oncology 7: 1 120 (1989); Wright et al. Cancer Res. 49:2087 (1989); McCann et al. Cancer Res 51 :3296 (1991 ); Paterson et al. Cancer 7? 5. 51 :556 (1991); and Winstanley et al. Br. J. Cancer 63:447 (1991)] but not in others [Zhou et al. Oncogene 4:105 (1989); Heintz et al. Arch Path Lab Med 1 14: 160 (1990); Kury et al. Eur. J. Cancer 26:946 (1990); Clark et al. Cancer Res. 51 :944 (1991); and Ravdin et al. J. Clin. Oncol. 12:467-74 (1994)]. In an initial evaluation of 103 patients with breast cancer, those having more than three tumor cell positive axillary lymph nodes (node positive) were more likely to overexpress HER2 protein than patients with less than three positive nodes [Slamon et al. Science 235: 177 (1987)]. In a subsequent evaluation of 86 node- positive patients with breast cancer, there was a significant correlation among the extent of gene amplification, early relapse, and short survival. HER2 overexpression was determined using Southern and Northern blotting which correlate with the HER2 oncoprotein expression evaluated by Western blotting and IHC [Slamon et al. Science 235:177 (1987); Slamon et al. Science 244:707 (1989)]. The median period of survival was found to be approximately 5-fold shorter in patients with more than five copies of the HER2 gene than in patients without gene amplification. This correlation was present even after correcting for nodal status and other prognostic factors in multivariate analyses. These studies were extended in 187 node-positive patients and indicated that gene amplification, increased amounts of mRNA (determined by Northern blotting), and increased protein expression (determined immunohistochemically) were also correlated with shortened survival time [Slamon et al. Science 244:707 (1989)]. See also US Patent 4,968,603 to Slamon et al
Nelson et al. have compared HER2/neu gene amplification using FISH with immunohistochemically determined overexpression in breast cancer [Nelson et al. Modern Pathology 9 (1) 21A (1996)]. SUMMARY OF THE INVENTION
The present invention combines cellular morphological analysis with fluorescence in situ hybridization (FISH) to provide for a correlation of genetic abnormalities and cellular morphology within the same section of a subject's tissue sample. Accordingly, one may identify and score by FISH cancer cells (e.g. invasive ductal carcinoma cells) as distinct from other normal cells (e.g. stromal and inflammatory elements found in the biopsy). Alternatively, or additionally, the invention combines immunohistochemistry (IHC) with FISH to provide for a correlation of genetic abnormalities with protein expression in the same tissue section.
Morphologic assessment, or evaluation of protein expression, in a tissue prior to quantitative FISH analysis provides for accurate, specific evaluation of that tissue in a timely and cost-efficientmanner. Thus, there is a need in research, prognostic, and diagnostic applications for a method that can allow for morphologic and/or protein expression analyses followed by FISH assessment in a single tissue sample section, particularly when testing a heterogeneous tissue. The invention described in this disclosure offers these features.
Accordingly, in a first aspect the invention provides a method of correlating cellular morphology with the presence of a cellular target nucleic acid sequence in a section of a tissue sample comprising the following steps:
(a) staining the section of tissue sample with a morphological stain;
(b) determining cellular morphology in the section of tissue sample;
(c) hybridizing a first fluorescently labeled nucleic acid probe to the target nucleic acid sequence in the same section of tissue sample;
(d) detecting the presence of the first nucleic acid probe in the section of tissue sample; and
(e) correlating step (b) with step (d).
In an alternative embodiment, the invention pertains to a method of correlating the presenceof a cellular target protein with the presence of a cellular target nucleic acid sequence in a section of a tissue sample comprising the following steps:
(a) contacting the section of sample tissue with an antibody which specifically binds to the target protein;
(b) determining binding of the antibody to the section of tissue sample;
(c) hybridizing a fluorescently labeled nucleic acid probe to the target nucleic acid sequence in the same section of tissue sample;
(d) detecting the presence of the labeled nucleic acid probe in the section of tissue sample; and
(e) correlating step (b) with step (d).
Additionally, the invention provides a kit comprising: (a) a morphological stain; (b) a fluorescently labeled probe complementary to a genetic abnormality; and (c) instructions for applying the stain (a) and probe (b) to the same section of tissue sample.
Moreover, a kit is provided comprising: (a) a primary antibody which specifically binds a cellular target protein; (b) a fluorescently labeled probe complementary to a genetic abnormality; and (c) instructions for applying the antibody (a) and probe (b) to the same section of tissue sample.
DESCRIPTION OF THE FIGURES Figure 1 shows the effect of HEMA 3® staining on the ability to score FISH in formalin fixed, paraffin- embedded cells harvested from each of three cell lines of known HER2/neu amplification status; SKBR3 (highly amplified HER2), MDA175 (barely amplified HER2), and MDA231 (non-amplified HER2).
Figure 2 shows the effect of HEMA 3® moφhologic stain on FISH with respect to tumor biopsy sections. Duplicate sections from 13 tumor specimens were analyzed for HER2 and chromosome 17 (Chr 17) by FISH either with or without prior staining for moφhologic analysis prior to FISH. The same area of tumor was scored for FISH on each section.
Figure 3 shows the mean HER2/neu:Chτ 17 ratio for blinded vs non-blinded assessment of normal tissue.
Figure 4 shows the mean HER2/neu:Chr 17 ratio for blinded vs non-blinded assessment of tumor tissue. Figure 5 shows the relationship between the scoring systems used for HER2lneu amplification by FISH and overexpression by IHC.
DETAILED DESCRIPTION OF THE INVENTION Before the present methods, kits and uses therefor are described, it is to be understood that this invention is not limited to the particular methodology, protocols, cell lines, animal species or genera, constructs, and reagents described as such may, of course, vary. It is also to be understood that the terminology used herein is for the puφose of describing particular embodiments only, and is not intended to limit the scope of the present invention which will be limited only by the appended claims.
It must be noted that as used herein and in the appended claims, the singular forms "a", "and", and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "a genetic alteration" includes a plurality of such alterations and reference to "a probe" includes reference to one or more probes and equivalents thereof known to those skilled in the art, and so forth.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art to which this invention belongs. Although any methods, devices and materials similar or equivalent to those described herein can be used in the practice or testing of the invention, the preferred methods, devices and materials are now described.
All publications mentioned herein are incoφorated herein by reference to disclose and describe the methods and/or materials in connection with which the publications are cited.
Publications cited herein are cited for their disclosure prior to the filing date of the present application. Nothing here is to be construed as an admission that the inventors are not entitled to antedate the publications by virtue of an earlier priority date or prior date of invention. Further the actual publication dates may be different from those shown and require independent verification.
DEFINITIONS By "subject" or "patient" is meant any single subject for which therapy is desired, including humans, cattle, dogs, guinea pigs, rabbits, chickens, insects and so on. Also intended to be included as a subject are any subjects involved in clinical research trials not showing any clinical sign of disease, or subjects involved in epidemiological studies, or subjects used as controls.
By "tissue sample" is meant a collection of similar cells obtained from a tissue of a subject or patient, preferably containing nucleated cells with chromosomal material. The four main human tissues are (1) epithelium; (2) the connective tissues, including blood vessels, bone and cartilage; (3) muscle tissue; and (4) nerve tissue. The source of the tissue sample may be solid tissue as from a fresh, frozen and/or preserved organ or tissue sample or biopsy or aspirate; blood or any blood constituents;bodily fluids such as cerebral spinal fluid, amniotic fluid, peritoneal fluid, or interstitial fluid; cells from any time in gestation or development of the subject. The tissue sample may also be primary or cultured cells or cell lines. The tissue sample may contain compounds which are not naturally intermixed with the tissue in nature such as preservatives, anticoagulants, buffers, fixatives, nutrients, antibiotics, or the like. In one embodiment of the invention, the tissue sample is "non-hemato logic tissue" (i.e. not blood or bone marrow tissue).
For the puφoses herein a "section" of a tissue sample is meant a single part or piece of a tissue sample, e.g. a thin slice of tissue or cells cut from a tissue sample. It is understood that multiple sections of tissue samples may be taken and subjected to analysis according to the present invention, provided that it is understood that the present invention comprises a method whereby the same section of tissue sample is analyzed at both moφhological and molecular levels, or is analyzed with respect to both protein and nucleic acid.
By "correlate" or "correlating" is meant comparing, in any way, the performance and/or results of a first analysis with the performance and/or results of a second analysis. For example, one may use the results of a first analysis in carrying out the second analysis and/or one may use the results of a first analysis to determine whether a second analysis should be performed and/or one may compare the results of a first analysis with the results of a second analysis. With respect to the embodiment of moφhological analysis followed by FISH, one may use the results obtained upon moφhological staining to determine area(s) of a tissue section which are normal and/or area(s) which are cancerous. Thus, histologically normal area(s) in a heterogeneous tumor biopsy may be used as internal normal control(s). In relation to IHC combined with FISH, one may use the results of IHC to determine whether FISH should be performed and/or one may compare the level of protein expression with gene amplification to further characterize a tumor biopsy (e.g. to compare HER2 protein expression with HER2 gene amplification). By "nucleic acid" is meant to include any DNA or RNA. For example, chromosomal, mitochondrial, viral and/or bacterial nucleic acid present in tissue sample. The term "nucleic acid" encompasses either or both strands of a double stranded nucleic acid molecule and includes any fragment or portion of an intact nucleic acid molecule.
By "gene" is meant any nucleic acid sequence or portion thereof with a functional role in encoding or transcribing a protein or regulating other gene expression. The gene may consist of all the nucleic acids responsible for encoding a functional protein or only a portion of the nucleic acids responsible for encoding or expressing a protein. The nucleic acid sequence may contain a genetic abnormality within exons, introns, initiation or termination regions, promoter sequences, other regulatory sequences or unique adjacent regions to the gene. By "genetic abnormality" is meant a deletion, substitution, addition, translocation, amplification and the like relative to the normal native nucleic acid content of a cell of a subject.
By "disease gene" is meant a gene that results in altered protein product (i.e., protein different from native protein in terms of sequence, structure and/or amount expressed) and results in a disease.
By "deletion" is meant the absence of all or part of a gene. By "amplification" is meant the presence of one or more extra gene copies in a chromosome complement.
The word "label" when used herein refers to a compound or composition which is conjugated or fused directly or indirectly to a reagent such as a nucleic acid probe or an antibody and facilitates detection of the reagent to which it is conjugated or fused. The label may itself be detectable (e.g., radioisotope labels or fluorescent labels) or, in the case of an enzymatic label, may catalyze chemical alteration of a substrate compound or composition which is detectable.
The term "fluorescently labeled nucleic acid probe" refers to a probe comprising (1) a nucleic acid sequence capable of hybridizing with a target nucleic acid sequence and (2) a fluorescent label.
By "moφhological stain" is meant a dye that stains different cellular components, in order to facilitate identification of cell type and/or disease status by light microscopy. Preferably, the moφhological stain is readily distinguishable from any label used in the FISH analysis, e.g., a stain which will not autofluoresce at the same wavelength as the fluorochrome used in the FISH analysis.
The term "antibody" is used in the broadest sense and specifically covers monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), and antibody fragments so long as they bind specifically to a target antigen.
The term "monoclonal antibody" as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to conventional (polyclonal) antibody preparations that typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen. The modifier "monoclonal" indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method. For example, the monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler et al, Nature 256:495 ( 1975), or may be made by recombinant DNA methods (see, e.g., U.S. Patent No. 4,816,567). The "monoclonal antibodies" may also be isolated from phage antibody libraries using the techniques described in Clackson et al, Nature 352:624-628 (1991) and Marks et al, J. Mol. Biol. 222:581-597 (1991), for example. The monoclonal antibodies herein specifically include "chimeric" antibodies (immunoglobulins) in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity [U.S. Patent No. 4,816,567; and Morrison et al, Proc. Natl. Acad. Sci. USA 81 :6851-6855 (1984)].
"Humanized" forms of non-human (e.g., murine) antibodies are chimeric antibodies that contain minimal sequence derived from non-human immunoglobulin. For the most part, humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a hypervariable region of the recipient are replaced by residues from a hypervariable region of a non-human species (donor antibody) such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity. In some instances, Fv framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues. Furthermore, humanized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. These modifications are made to further refine antibody performance. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FRs are those of a human immunoglobulin sequence. The humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. For further details, see Jones et al, Nature 321 :522-525 (1986); Riechmann et al, Nature 332:323-329 (1988); and Presta, Curr. Op. Struct. Biol 2:593-596 (1992).
The term "primary antibody" herein refers to an antibody which binds specifically to the target protein antigen in a tissue sample. A primary antibody is generally the first antibody used in an immunohistochemical procedure. In one embodiment, the primary antibody is the only antibody used in an IHC procedure.
The term "secondary antibody" herein refers to an antibody which binds specifically to a primary antibody, thereby forming a bridge between the primary antibody and a subsequent reagent, if any. The secondary antibody is generally the second antibody used in an immunohistochemical procedure.
HER2 Unless indicated otherwise, the terms "HER2", "pi 85 " and "ErbB2" when used herein refer to human HER2 protein or a portion thereof and "HER2 " , "HER2/neu " and "c-er/>B2" refer to the human HER2 gene or a portion thereof. The human HER2 gene and HER2 protein are, for example, described in Semba et al, PNAS (USA) 82:6497-6501 (1985) and Yamamoto et al. Nature 319:230-234 (1986) (Genebank accession number X03363).
DUAL ANALYSIS METHOD The present invention relates to a method which combines moφhological staining and/or immunohistochemical analysis with fluorescence in situ hybridization (FISH) within the same section of a tissue sample. The present methods allow for a highly accurate and simplified method of correlating moφhological criteria or protein expression with genetic abnormalities. Because many tissue types, such as breast tissue, are characterized by significant cellular heterogeneity, inaccurate results may be obtained when serial sections from a tissue block are analyzed by two different methods. According to the present invention, both types of analysis are carried out on the same tissue section, thereby reducing error when analyzing such heterogeneous tissue. As evidenced by the following, the method of this application is useful in a variety of prognostic, diagnostic and research applications. Also disclosed are kits for use in the disclosed methods.
SAMPLE PREPARATION Any tissue sample from a subject may be used. Examples of tissue samples that may be used include, but are not limited to, breast, prostate, ovary, colon, lung, endometrium, stomach, salivary gland or pancreas. The tissue sample can be obtained by a variety of procedures including, but not limited to surgical excision, aspiration or biopsy. The tissue may be fresh or frozen. In one embodiment, the tissue sample is fixed and embedded in paraffin or the like.
The tissue sample may be fixed (i.e. preserved) by conventional methodology [See e.g., "Manual of Histological Staining Method of the Armed Forces Institute of Pathology," 3rd edition (1960) Lee G. Luna, HT (ASCP) Editor, The Blakston Division McGraw-Hill Book Company, New York; The Armed Forces Institute of Pathology Advanced Laboratory Methods in Histology and Pathology (1994) Ulreka V. Mikel, Editor, Armed Forces Institute of Pathology, American Registry of Pathology, Washington, D.C.]. One of skill in the art will appreciate that the choice of a fixative is determined by the puφose for which the tissue is to be histologically stained or otherwise analyzed. One of skill in the art will also appreciate that the length of fixation depends upon the size of the tissue sample and the fixative used. By way of example, neutral buffered formalin, Bouin's or paraformaldehyde, may be used to fix a tissue sample.
Generally, the tissue sample is first fixed and is then dehydrated through an ascending series of alcohols, infiltrated and embedded with paraffin or other sectioning media so that the tissue sample may be sectioned. Alternatively, one may section the tissue and fix the sections obtained. By way of example, the tissue sample may be embedded and processed in paraffin by conventional methodology (See e.g., "Manual of Histological Staining Method of the Armed Forces Institute of Pathology", supra). Examples of paraffin that may be used include, but are not limited to, Paraplast, Broloid, and Tissuemay. Once the tissue sample is embedded, the sample may be sectioned by a microtome or the like (See e.g., "Manual of Histological Staining Method of the Armed Forces Institute of Pathology", supra). By way of example for this procedure, sections may range from about three microns to about five microns in thickness. Once sectioned, the sections may be attached to slides by several standard methods. Examples of slide adhesives include, but are not limited to, silane, gelatin, poly-L-lysine and the like. By way of example, the paraffin embedded sections may be attached to positively charged slides and/or slides coated with poly-L-lysine.
If paraffin has been used as the embedding material, the tissue sections are generally deparaffinizedand rehydrated to water. The tissue sections may be deparaffinized by several conventional standard methodologies. For example, xylenes and a gradually descending series of alcohols may be used (See e.g., "Manual of Histological Staining Method of the Armed Forces Institute of Pathology", supra). Alternatively, commercially available deparaffinizing non-organic agents such as Hemo-De® (CMS, Houston, Texas) may be used.
MORPHOLOGICAL STAINING After deparaffinization, the sections mounted on slides may be stained with a moφhological stain for evaluation. The moφhological stain to be used in the instant method provides for accurate moφhological evaluation of a tissue section and also allows for accurate quantification of fluorescently labeled (e.g., SPECTRUM ORANGE® and SPECTRUM GREEN®) nucleic acid probes when the sections are subsequently processed for FISH. Many moφhological stains fluoresce when illuminated by light of the same wavelength required to visualize the probe fluorophore of interest. The disclosed method solves this problem. Generally, the section is stained with one or more dyes each of which distinctly stains different cellular components. In a preferred embodiment xanthine dye or the functional equivalent thereof and/or a thiazine dye or the functional equivalent thereof are used to enhance and make distinguishable the nucleus, cytoplasm, and "granular" structures within each. Such dyes are commercially available and often sold as sets. By way of example, HEMA 3® (CMS, Houston, Texas) stain set comprises xanthine dye and thiazine dye. Methyleneblue may also be used. Examples of other moφhological stains that may be used on the instant method include, but are not limited to, dyes that do not significantly autofluoresce at the same wavelength as fluorescent label(s) used for the subsequent FISH analysis. For example, where the fluorescent labels used for the FISH are SPECTRUM ORANGE® and SPECTRUM GREEN®, the moφhological stain preferably does not significantly fluoresce at about 509/538 (peak excitation/emission) and/or about 559/588 (peak excitation/emission). One of skill in the art will appreciate that staining may be optimized for a given tissue by increasing or decreasing the length of time the slides remain in the dye.
After staining, the tissue section may be analyzed by standard techniques of microscopy. Generally, a pathologist or the like assesses the tissue for the presence of abnormal or normal cells or a specific cell type and provides the loci of the cell types of interest. Thus, for example, in a study correlating HER2/neu amplification in breast cancer, a pathologist or the like would review the slides and identify normal breast cells and abnormal breast cells (See e.g. Example 2).
Any means of defining the loci of the cells of interest may be used (e.g., coordinates on an X-Y axis). Generally, after light microscopy and prior to the FISH procedure, the slides are destained by conventional methodology. The present method provides an advantage over the prior procedures in the art in that no separate destaining procedure is required prior to FISH. Avoidance of a destaining step is actually preferred in order to protect the integrity of the DNA for in situ hybridization.
IMMUNOHISTOCHEMISTRY (IHC) Prior to FISH, the tissue section may be subjected to IHC. IHC may be performed in combination with moφhological staining as discussed in the preceding section (either prior to, but preferably thereafter).
Two general methods of IHC are available; direct and indirect assays. According to the first assay, binding of antibody to the target antigen is determined directly. This direct assay uses a labeled reagent, such as a fluorescent tag or an enzyme-labeled primary antibody, which can be visualized without further antibody interaction. In a typical indirect assay, unconjugated primary antibody binds to the antigen and then a labeled secondary antibody binds to the primary antibody. Where the secondary antibody is conjugated to an enzymatic label, a chromagenic or fluorogenic substrate is added to provide visualization of the antigen. Signal amplification occurs because several secondary antibodies may react with different epitopes on the primary antibody.
The primary and/or secondary antibody used for immunohistochemistry typically will be labeled with a detectable moiety. Numerous labels are available which can be generally grouped into the following categories:
35 14 125 3 131
(a) Radioisotopes, such as S, C, I, H, and I. The antibody can be labeled with the radioisotope using the techniques described in Current Protocols in Immunology, Volumes 1 and 2, Coligen et al, Ed. Wiley-Interscience, New York, New York, Pubs. (1991) for example and radioactivity can be measured using scintillation counting.
(b) Colloidal gold particles.
(c) Fluorescent labels including, but are not limited to, rare earth chelates (europium chelates), Texas Red, rhodamine, fluorescein, dansyl, Lissamine, umbelliferone, phycocrytherin, phycocyanin, or commercially available fluorophores such SPECTRUM ORANGE® and SPECTRUM GREEN® and/or derivatives of any one or more of the above. The fluorescent labels can be conjugated to the antibody using the techniques disclosed in Current Protocols in Immunology, supra, for example. Fluorescence can be quantified using a fluorimeter.
(d) Various enzyme-substrate labels are available and U.S. Patent No. 4,275, 149 provides a review of some of these. The enzyme generally catalyzes a chemical alteration of the chromogenic substrate that can be measured using various techniques. For example, the enzyme may catalyze a color change in a substrate, which can be measured spectrophotometrically. Alternatively, the enzyme may alter the fluorescence or chemiluminescence of the substrate. Techniques for quantifying a change in fluorescence are described above. The chemiluminescent substrate becomes electronically excited by a chemical reaction and may then emit light which can be measured (using a chemiluminometer, for example) or donates energy to a fluorescent acceptor. Examples of enzymatic labels include luciferases (e.g., firefly luciferase and bacterial luciferase; U.S. Patent No. 4,737,456), luciferin, 2,3-dihydrophthalazinediones, malate dehydrogenase, urease, peroxidase such as horseradish peroxidase (HRPO), alkaline phosphatase, β-galactosidase, glucoamylase, lysozyme, saccharide oxidases (e.g., glucose oxidase, galactose oxidase, and glucose-6-phosphate dehydrogenase), heterocyclic oxidases (such as uricase and xanthine oxidase), lactoperoxidase, microperoxidase, and the like. Techniques for conjugating enzymes to antibodies are described in O'Sullivan et al, Methods for the Preparation of Enzyme- Antibody Conjugates for use in Enzyme Immunoassay, in Methods in Enzym. (ed J. Langone & H. Van Vunakis), Academic press, New York, 73 : 147- 166 ( 1981 ). Examples of enzyme-substrate combinations include, for example:
(i) Horseradish peroxidase (HRPO) with hydrogen peroxidase as a substrate, wherein the hydrogen peroxidase oxidizes a dye precursor [e.g.,orthophenylene diamine (OPD) or 3,3',5,5'-tetramethyl benzidine hydrochloride (TMB)];
(ii) alkaline phosphatase (AP) with para-Nitrophenyl phosphate as chromogenic substrate; and (iii) β-D-galactosidase (β-D-Gal) with a chromogenic substrate (e.g., p-nitrophenyl-β-D- galactosidase) or fluorogenic substrate (e.g., 4-methylumbelliferyl-β-D-galactosidase).
Numerous other enzyme-substrate combinations are available to those skilled in the art. For a general review of these, see U.S. Patent Nos. 4,275,149 and 4,318,980.
Sometimes, the label is indirectly conjugated with the antibody. The skilled artisan will be aware of various techniques for achieving this. For example, the antibody can be conjugated with biotin and any of the four broad categories of labels mentioned above can be conjugated with avidin, or vice versa. Biotin binds selectively to avidin and thus, the label can be conjugated with the antibody in this indirect manner. Alternatively, to achieve indirect conjugation of the label with the antibody, the antibody is conjugated with a small hapten and one of the different types of labels mentioned above is conjugated with an anti-hapten antibody. Thus, indirect conjugation of the label with the antibody can be achieved.
Aside from the sample preparation procedures discussed above, further treatment of the tissue section prior to, during or following IHC may be desired, For example, epitope retrieval methods, such as heating the tissue sample in citrate buffer may be carried out [see, e.g., Leong et al. Appl Immunohistochem. 4(3):201 (1996)]. Following an optional blocking step, the tissue section is exposed to primary antibody for a sufficient period of time and under suitable conditions such that the primary antibody binds to the target protein antigen in the tissue sample. Appropriate conditions for achieving this can be determined by routine experimentation. The extent of binding of antibody to the sample is determined by using any one of the detectable labels discussed above. Preferably, the label is an enzymatic label (e.g. HRPO) which catalyzes a chemical alteration of the chromogenic substrate such as 3,3'-diaminobenzidinechromogen. Preferably the enzymatic label is conjugated to antibody which binds specifically to the primary antibody (e.g. the primary antibody is rabbit polyclonal antibody and secondary antibody is goat anti-rabbit antibody).
Specimens thus prepared may be mounted and coverslipped. Slide evaluation is then determined, e.g. using a microscope. Where the antigen is HER2 protein, staining intensity criteria may be evaluated as follows: TABLE 1 HER2 Protein Staining Intensity Criteria
Figure imgf000013_0001
In this embodiment of the invention, it may be desirable to select a subgrouping of the tissue samples subjected to IHC for further analysis by FISH. For example, tissue samples with 1+ and 2+ scores, and especially, the 2+ subgroup may be subjected to further FISH as explained below.
FLUORESCENCE IN SITU HYBRIDIZATION (FISH)
In situ hybridization is generally carried out on cells or tissue sections fixed to slides. In situ hybridization may be performed by several conventional methodologies [See for e.g. Leitch et al. In Situ Hybridization: a practical guide, Oxford BIOS Scientific Publishers, Micropscopy handbooks v. 27 (1994)].
In one in situ procedure, fluorescent dyes [such as fluorescein isothiocyanate (FITC) which fluoresces green when excited by an Argon ion laser] are used to label a nucleic acid sequence probe which is complementary to a target nucleotide sequence in the cell. Each cell containing the target nucleotide sequence will bind the labeled probe producing a fluorescent signal upon exposure, of the cells to a light source of a wavelength appropriate for excitation of the specific fluorochrome used.
Various degrees of hybridization stringency can be employed. As the hybridization conditions become more stringent, a greater degree of complementarity is required between the probe and target to form and maintain a stable duplex. Stringency is increased by raising temperature, lowering salt concentration, or raising formamide concentration. Adding dextran sulfate or raising its concentration may also increase the effective concentration of labeled probe to increase the rate of hybridization and ultimate signal intensity. After hybridization, slides are washed in a solution generally containing reagents similar to those found in the hybridization solution with washing time varying from minutes to hours depending on required stringency. Longer or more stringent washes typically lower nonspecific background but run the risk of decreasing overall sensitivity. Exemplary in situ hybridization conditions for detecting HERllneu amplification in breast tissue are shown in Example 2.
Probes used in the FISH analysis may be either RNA or DNA oligonucleotides or polynucleotides and may contain not only naturally occurring nucleotides but their analogs like digoxygenin dCTP, biotin dcTP 7- azaguanosine, azidothymidine, inosine, or uridine. Other useful probes include peptide probes and analogues thereof, branched gene DNA, peptidometics, peptide nucleic acid (PNA) and/or antibodies. Probes should have sufficient complementarity to the target nucleic acid sequence of interest so that stable and specific binding occurs between the target nucleic acid sequence and the probe. The degree of homology required for stable hybridization varies with the stringency of the hybridization medium and/or wash medium. Preferably, completely homologous probes are employed in the present invention, but persons of skill in the art will readily appreciate that probes exhibiting lesser but sufficient homology can be used in the present invention [see for e.g. Sambrook, J., Fritsch, E.F., Maniatis, T., Molecular Cloning A Laboratory Manual, Cold Spring Harbor Press, (1989)].
One of skill in the art will appreciate that the choice of probe will depend on the genetic abnormality of interest. Genetic abnormalities that can be detected by this method include, but are not limited to, amplification, translocation, deletion, addition and the like. Examples of amplification include, but are not limited to, HER2lneu in breast and ovarian cancer, ~N-myc in neuroblastoma, C-myc in small cell lung cancer. Examples of abnormal chromosome number include, but are not limited to, trisomy 8 in leukemia, monosomy 7 in myloproliferative disorders, and trisomy 12 in chronic lymphoblastic leukemia. Examples of translocations include, but are not limited to, bcr/abl [t (9;22)] translocation in chronic mylogenous leukemia and the t ( 15; 17) translocation FAB-M3 (acute promyelocytic leukemia). Examples of deletions include EGFR vIII and p53. By way of example for evaluating HER2/neu amplification a probe spanning a 140 kb region on the long arm of chromosome 17 containing the HER2lneu gene (17q 1 1.2-17ql2) may be used. A probe for the -satellite sequences in the centromeric region of chromosome 17(D1721 ) may be used to evaluate for aneusomy of chromosome 17 as a source or cause for HERllneu amplification. For example, a cocktailed version of these probes may be obtained from Vysis, Inc. where each probe is directly labeled with easily distinguishable fluorophores, such as SPECTRUM ORANGE® and SPECTRUM GREEN®.
Probes may also be generated and chosen by several means including, but not limited to, mapping by in situ hybridization, somatic cell hybrid panels, or spot blots of sorted chromosomes; chromosomal linkage analysis; or cloned and isolated from sorted chromosome libraries from human cell lines or somatic cell hybrids with human chromosomes, radiation somatic cell hybrids, microdissection of a chromosome region, or from yeast artificial chromosomes (YACs) identified by PCR primers specific for a unique chromosome locus or other suitable means like an adjacent YAC clone. Probes may be genomic DNA, cDN A, or RNA cloned in a plasmid, phage, cosmid, YAC, Bacterial Artificial Chromosomes (BACs), viral vector, or any other suitable vector. Probes may be cloned or synthesized chemically by conventional methods. When cloned, the isolated probe nucleic acid fragments are typically inserted into a vector, such as lambda phage, pBR322, M13, or vectors containing the SP6 or T7 promoter and cloned as a library in a bacterial host. [See for e.g. Sambrook, J ., Fritsch, E.F., Maniatis, T., Molecular Cloning A Laboratory Manual, Cold Spring Harbor Press, (1989)].
Probes are preferably labeled with a fluorophor. Examples of fluorophores include, but are not limited to, rare earth chelates (europium chelates), Texas Red, rhodamine, fluorescein, dansyl, Lissamine, umbelliferone, phycocrytherin, phycocyanin, or commercially available fluorophors such SPECTRUM ORANGE® and SPECTRUM GREEN® and/or derivatives of any one or more of the above. Multiple probes used in the assay may be labeled with more than one distinguishablefluorescent or pigment color. These color differences provide a means to identify the hybridization positions of specific probes. Moreover, probes that are not separated spatially can be identified by a different color light or pigment resulting from mixing two other colors (e.g., light red+green=yellow)pigment (e.g., blue+yellow=green)or by using a filter set that passes only one color at a time.
Probes can be labeled directly or indirectly with the fluorophor, utilizing conventional methodology.
Additional probes and colors may be added to refine and extend this general procedure to include more genetic abnormalities or serve as internal controls. By way of example the HERllneu gene is in chromosome 17, and as an internal control a probe for -satellite sequences specific for chromosome 17 (D17Z1 ) may be used (Vysis Inc.) to prove diploidy in areas of non-malignant cells and/or to establish the presence or absence of chromosome 17 aneusomy in areas of HER2/neu amplification.
After processing for FISH, the slides may be analyzed by standard techniques of fluorescence microscopy [see for e.g. Ploem and Tanke Introduction to Fluorescence Microscopy, New York, Oxford University Press ( 1987)]. Briefly, each slide is observed using a microscope equipped with appropriate excitation filters, dichromic, and barrier filters. Filters are chosen based on the excitation and emission spectra of the fluorochromes used. Photographs of the slides may be taken with the length of time of film exposure depending on the fluorescent label used, the signal intensity and the filter chosen. For FISH analysis the physical loci of the cells of interest determined in the moφhological analysis are recalled and visually conformed as being the appropriate area for FISH quantification.
In order to correlate cellular moφhology and/or IHC with FISH, one may use a computer-driven, motorized stage which stores location of co-ordinates. This may be used to evaluate the same area by two different analytical techniques. For example, color images of the moφhologically stained areas may be captured and saved using a computer-assistedcooled CCD camera. The same section may be subsequently taken through the FISH procedure, the stored locations recalled, and the designated areas scored for the presence of fluorescent nuclear signals. A similar procedure for IHC followed by FISH is contemplated.
Typically, hundreds of cells are scanned in a tissue sample and quantification of the specific target nucleic acid sequence is determined in the form of fluorescent spots, which are counted relative to the number of cells. Deviation of the number of spots in a cell from a norm (e.g., such as probing for the HER2/neu gene in a normal cell will produce two copies, abnormal greater than two) may be indicative of a malignancy or a predisposition to a malignancy, disease, or other abnormality. The relative number of abnormal cells to the total cell sample population may also indicative of the extent of the condition or abnormality. In addition, using family health histories and/or genetic screening, it is possible to estimate the probability that a particular subject has for developing certain types of cancer. Those subjects that have been identified as being predisposed to developing a particular form of cancer can be monitored or screened to detect early evidence of disease. Upon discovery of such evidence, early treatment can be undertaken to combat the disease. Similarly, those subjects who have already developed a malignancy and who have been treated to remove the cancer or are otherwise in remission are particularly susceptible to relapse and reoccurrence, including the metastasis of tumors. Such subjects can be monitored and screened using the presently disclosed methods to detect evidence of metastasis and upon discovery of such evidence, immediate treatment can be undertaken to combat the disease.
KITS In yet another embodiment, this invention provides a kit comprising a fluorescently labeled probe specific for the target nucleic acid sequence of interest and a moφhological stain and/or an antibody which specifically binds target antigen. The kit further comprises a set of instructions for applying the stain or antibody and probe to the same section of a tissue sample. By way of example, the fluorescently labeled probe may be complementary to the HERllneu gene and the moφhological stain may be HEMA 3® (CMS, Houston, Texas). Any fluorescent label as described above may be used to label the probe. The IHC/FISH kit may comprise a fluorescently labeled probe complementary to the HER2/neu gene and an antibody (e.g. polyclonal antibody) which binds to the HER2 protein. The kit may include both a primary and secondary antibody, wherein the secondary antibody is conjugated to a label, e.g., an enzymatic label. The invention also provides an IHC kit which has instructions to perform FISH on the same section of tissue sample on which IHC has been previously performed.
Other optional components in the kit include one or more buffers (e.g., block buffer, wash buffer, substrate buffer, etc), other reagents such as substrate (e.g., chromagen) which is chemically altered by an enzymatic label, epitope retrieval solution, control samples (positive and/or negative controls), control slide(s) etc.
EXAMPLES The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how to carry out the invention and is not intended to limit the scope of what the inventors regard as their invention. Efforts have been made to ensure accuracy with respect to numbers used (e.g., amounts, temperatures, etc.), but some experimental error and deviation should be accounted for. Unless indicated otherwise, parts are parts by weight, molecular weight is weight average molecular weight, temperature is in degrees Centigrade, and pressure is at or near atmospheric. EXAMPLE 1
Evaluation of Morphological Stains on F SH Analysis
A sensitive and specific evaluation of breast tumors for the amplification of HER2lneu by FISH requires definitive identification and scoring of invasive ductal carcinoma cells as distinct from other stromal and inflammatory elements found in the biopsy. Thus, it was necessary to identify a stain which would allow for complete moφhological evaluation of the tissue, and which also allowed for easy quantification of nuclear signals upon subsequent hybridization with fluorescently labeled nucleic acid probes. Many moφhological stains fluoresce when illuminated by light of the same wavelength required to visualize the probe fluorophores of interest. When this autofluorescenceis of a color similarto that of the probe fluorophore, signal quantification is made difficult. Results from the evaluation of six commonly used moφhological stains may be found in Table 2.
TABLE 2 Evaluation of Morphological Stains
Figure imgf000017_0001
Methyl green pyronin (MGP) and Gill hematoxylin were not analyzed further due to poor moφhological definition. Paragon (toluidine blue/basic fuchsin) and Weigert were eliminated from consideration because of an unacceptably high level of red or orange autofluorescence which masked the orange HERl/neu signals in the nucleus. Methylene blue yielded acceptable moφhological staining, but demonstrated orange autofluorescence which caused moderate difficulty in visualizing HER2/neu signals. This stain was considered a possible candidate. HEMA 3®, however, was superior in that it provided good moφhological detail and showed only modest dim green autofluorescence which did not interfere to any extent with FISH evaluation. An initial attempt at destaining HEMA 3® prior to FISH was found to damage the integrity of the DNA in the cells and ultimately proved to be unnecessary anyway. It was possible for the stained tissue to be taken directly through the FISH procedure without intermediate processing, yielding high-quality nuclear signals without any negative consequence. HEMA 3®, therefore, met all essential requirements and was the stain of choice for the proposed system. Unless otherwise indicated, the tissues and FISH analysis were conducted as discussed in Example 2.
EXAMPLE 2 Detection and Quantification of HER2/neu Amplification in Breast Tissue Materials and Methods Tissue specimens and preparation for F SH
Five micron sections were cut from breast tissue which had been fixed in buffered formalin and embedded in paraffin. Sections were placed on positively charged Superfrost Plus slides (CMS, Houston, TX) which had also been treated with poly-L-lysine or were mounted on positively charged slides which had no additional treatment prior to the mounting of the slides. Some material had been archived for up to fourteen years. Slide mounted tissue sections were heated on a 65°C slide warmer for approximately 3 seconds, placed on the bench top for 2 seconds, and deparaffinized in Hemo-De (CMS, Houston, TX) for 10 minutes, x3, followed by immersion in 100% ethanol (EtOH) for 5 minutes, x2. Slides were air dried in a vertical position. Those slides to be stained for moφhologic evaluation were dipped in HEMA 3® Solution I (CMS, Houston, TX) for one second. x3, then immediately dipped in HEMA 3® Solution II for one second, x3. Each slide was rinsed in deionized, distilled water and allowed to air dry in a vertical position. The slides were mounted and coverslipped using an aqueous-based mounting medium (Biomeda Coφoration, Foster City, CA) for evaluation at the microscope. Areas of interest identified by the pathologist were marked on the coverglass and subsequently circled on the underside of the slide using a diamond-tipped pencil. The coverglass was removed by placing the slide in a water-filled Coplin jar, which had been pre-heated in a 73+2°C waterbath, for approximately 15-30 minutes, or until the coverglass slipped off. The section was dehydrated in 70% EtOH for 3 minutes, 85% EtOH for 3 minutes, and 100% EtOH for 3 minutes. These slides, and any deparaffinized sections that were not stained but were to be processed by FISH, were immersed in a sodium isothiocyanate Pretreatment Solution (Vysis, Inc., Downers Grove, IL) for 30 minutes at 80+l°C. The sections were washed in deionized water for one minute, then in Protease Wash Buffer (2XSSC, pH 7.4, Vysis, Inc.) for 5 minutes at room temperature, x2. The sections were treated in Protease Solution (Vysis, Inc.) for 10 minutes at 37°C. Following protease digestion, the sections were washed in Protease Wash Buffer for 5 minutes, x2, and dried on a 45-50°C slide warmer for 2-5 minutes. Probes
A probe spanning approximately 140 kb of the region on the long arm of chromosome 17 containing the HER2/neu proto-oncogene (17ql 1.2 - 17ql2) was utilized to detect the presence or absence of HERllneu amplification. Chromosome 17 aneusomy of the cell was evaluated using an -satellite sequence probe (D17Z1) specific for the centromere of chromosome 17 (CEP 17). The probes were provided by Vysis, Inc. in an optimized cocktail mixture where the HER2lneu probe is directly labeled with the fluorophore SPECTRUM ORANGE® (peak excitation/emission=559/588)and the CEP 17 probe is directly labeled with the fluorophore SPECTRUM GREEN® (peak excitation/emission=509/538)(Vysis, Inc.). In situ hybridization
The pre-treated (see above) slide-mounted sections were immersed in Denaturation Solution (70% formamide/2xSSC, pH 7.0) for 5 minutes at 73+l °C. Sections were then dehydrated in 70% EtOH for one minute, 85% EtOH for one minute, and 100% EtOH for one minute. Drained slides were placed on a 45-50°C slide warmer for 2-5 minutes just before application of the hybridization mixture. This mixture was prepared as directed by VYSIS® protocol. The probe solution was denatured by incubation in a 73°C waterbath for 5 minutes. Following brief centrifugation, 10 L of the solution was pipetted onto each section, a coverslip applied, and the edges sealed with rubber cement. Hybridization was carried out in a humidified box overnight (14-18 h) in a 37°C incubator. The rubber cement was carefully removed and the slides immersed in room temperature Wash Buffer I (0.4xSSC/0.3% NP-40) until the coverslip floated off. Slides were drained of excess liquid by blotting the edges on a paper towel, then placed in a Coplin jar of Wash Buffer I, which had been prewarmed to 72+1 °C, for 2 minutes. Sections were washed in room temperature Wash Buffer II (2xSSC/0.1 %NP-40) for one minute, drained vertically on paper towels, and allowed to air-dry in darkness being careful not to overdry. Once slides were dry, 15 L of DAPI counterstain (a mixture of equal parts DAPI I and DAPI II, Vysis, Inc.) was pipetted onto the hybridization area. A coverglass was added and the edges sealed with clear nail polish. Slides were analyzed immediately or stored in the dark at -20°C. Controls Cultured cell lines SKBR3, MDA175, and MDA231 were harvested, formalin-fixed, and paraffin- embedded for use as highly amplified, barely amplified, and non-amplified controls, respectively. These cell lines were used in the evaluation of the moφhological stains. The presence of appropriate nuclear FISH signals served to assure that the reagents and procedure involved in the devised methodology worked properly. Microscopy An Olympus BX60 epifluorescence microscope equipped with a 100 watt mercury-arc lamp, a
40xUPlanApo objective, and a lOOxUPlanFl objective was used. The filter slider was built by Chroma Technology Coφoration (Brattleboro, VT) for Olympus and included three single band pass filters optimized for visualization of DAPI, FITC, and Texas Red fluorochromes, a dual-band pass filter optimized for FITC and Texas Red, and a triple-band pass filter optimized for DAPI, FITC, and Texas Red. The microscope was also equipped with 4x, 1 Ox, 20x, and 40x UPlanFl objectives for use in transmittance light microscopy. A BioPoint X, Y (Ludl Electronic Products, Ltd., Hawthorne, NY) computer-driven motorized stage was used for storage and retrieval of physical locations. Images were acquired using a CoolCam 2000 3-chip color cooled CCD camera (Sci-Measure, Atlanta, GA) and printed using a Tektronix Phaser 440 dye sublimation printer (Tektronix, Inc., Wilsonville, OR). FISH scoring criteria
After FISH processing, tissue sections were scanned using the 40x objective to evaluate for tissue loss and to ensure that hybridization was uniform across the section. Moφhologically identified areas of normal or tumor tissue were located on FISH processed slides by recalling stored x and y coordinates. Alternatively, if location coordinates had not been stored, a saved moφhological image (HEMA 3® stain) of the area of interest was used as a reference to localize the appropriate area after FISH processing. Signals were enumerated using the lOOx objective, switching between different filters to optimize signal discrimination. Nuclear boundaries were defined by DAPI excitation. Only those nuclei were scored which could be clearly identified as intact and non-overlapping. The presence of nuclei having no CEP 17 or no HER2lneu signaled the possibility of a hybridization failure or the existence of truncated nuclei, suggesting an unacceptably thin section. The hybridization quality of the entire section was assessed completely before making a decision regarding acceptability. In cases of high HER2/neu gene amplification (> 10-20 signals per nucleus), signals were often coalesced into clusters and could not be precisely enumerated. If, as in some instances, the CEP 17 signals appeared fragmented, a broken signal was scored as two separate signals if the distance between them was of sufficient size that a third comparable signal could be passed through it. A minimum scoring goal of 100 nuclei per area was attempted; however, in certain circumstances, it was not possible to meet this goal. For example, areas of normal tissue tended to be minimally represented and relatively non-cellular; in these situations, all clearly discernible cells were scored. In areas of highly amplified HER2lneu, a total of 30-40 cells was considered adequate to establish amplification status. Immunohistochemical staining
Sections cut serially from those used for FISH analysis were immunohistochemically stained using a murine monoclonal antibody against HER2 [4D5; Fendly et al Cancer Research 50: 1550-1558 (1990)]. Slide- mounted sections were placed into xylenes 3x, 10 minutes each time, then dehydrated in absolute ethanol 2x, 5 minutes each. Endogenous peroxidase activity was quenched by placing the slides in a 0.3% solution of hydrogen peroxide in methanol. The sections were then serially rehydrated in 95% ethanol for 5 minutes, followed by 80% ethanol for 5 minutes. Sections were incubated briefly in the buffer routinely used with the Ventana ES automated immunostaining instrument. The staining program began with a 4 minute protease treatment. Incubation with the primary antibody (lOμg/ml) proceeded for the maximum allowable time period, 32 minutes. The detection system employed a biotin/avidin reaction using DAB and a hematoxylin counterstain. After staining and detection, the sections were dehydrated in a graded series of ethanol into xylenes in preparation for permanent coverslipping. mmunohistochemistry scoring criteria
The immunohistochemical staining was inteφreted by a breast tumor pathologist and the results assigned to one of four categories defined in the following way: negative/weak, cytoplasmic, 1+, 2+. The second category was reserved for those cells in which staining occurred only in the cytoplasm. The latter two categories applied to cell surface antibody staining. Images of immunostained sections representative of each category were captured using the CCD camera. Attempts were made to relocate areas that had been scored for FISH on the corresponding serial section, and whenever possible that image was captured. Specimens
Formalin-fixed, paraffin-embedded human breast tissue was obtained through clinical submission of specimens to Cytometry Associates and Vanderbilt University Medical Center (VUMC), Department of Pathology. Some materials may have been archived at VUMC for up to fourteen years. In some cases, the specimens were identified for the study based on the likelihood of HER2 amplification as predicted by IHC results or hormone receptor status. Other cases were known to be disease-free based on moφhological evaluation at VUMC. Stain plus F SH vs. F SH only
Initially, the effect of HEMA 3® stain on the ability to score FISH was evaluated in formalin-fixed, paraffin-embeddedcells harvested from each of three cell-lines of known HERllneu amplification status (Figure 1). Results from analysis by two individuals were comparable and successfully identified the different cell-lines as being of the appropriate, expected amplification status. The stain had no adverse effect on FISH quantification in these cell lines. A total of 10 patient specimens was included in the study comparing stained and hybridized tissue versus tissue which was hybridized without prior staining. The same areas of tumor and normal cells identified on each of two serial sections was analyzed for the presence of HER2lneu and chromosome 17 by FISH either with or without prior moφhological staining. Two analysts performed FISH quantification of the designated areas. Mean HER2lneu:C r\ l ratios for each of the areas of tumor in the ten patient specimens scored, plus the three cell lines, are shown in Figure 2. There was no statistically significant quantitative difference as a result of the two different treatments of the tissue sections in either the tumor areas (P = 0.196) or the normal areas (P = 0.597) scored. The differences in the mean values between the two analysts performing FISH quantification were not statistically significant in areas of either normal cells (P = 0.065) or tumor cells (P = 0.459). In addition, the inter-observer effect did not depend on which treatment the tissue section had undergone, whether in areas of normal moφhology (P = 0.513) or in areas of invasive carcinoma (P = 0.971). Blinded vs Non-blinded Study A study was designed to determine whether prior knowledge of moφhological cell type biased the scoring of FISH signals. Ten patient specimens included in the study were stained with HEMA 3® for moφhological evaluation by a pathologist. Areas of normal cellulartissue and areas of invasive carcinoma were identified, their x/y coordinates stored, and images captured and saved. The files were named and stored in an anonymous and random manner so that the two analysts who scored the areas for FISH signals were unaware of their identity or cell type. After the blinded evaluation was completed, the analysts were given a second list of file names of the same ten images and locations of normal and tumor areas, but in this case, the identity and cell type was provided. To score FISH signals, the analyst used the x/y coordinate provided to relocate the area of interest and used the saved image to confirm accurate relocation. The mean HER2/neu:Chv 17 ratio was determined for each area scored by the analysts, and the mean ratio of the two analysts calculated. Mean ratios for blinded vs non-blinded assessment of both normal tissue and tumor tissue are shown in Figures 3 and 4, respectively. Non-diseased Normal vs Patient Normal
Normal range data were generated from analysis of histologically normal breast tissue obtained from mammoplasty surgical procedures. Ten biopsies were evaluated by two analysts for quantitative detection of HER2lneu and chromosome 17 by FISH. The mean HER2lneu:C τ 17 ratio for the two analysts was 1.07. In order to establish a normal range, areas of normal cellular tissue were identified in 38 biopsies of breast tissue from patients diagnosed with invasive carcinoma. The mean ratio for these specimens was also 1.07. Statistical treatment of the data with a two way repeated measures ANOVA found no significant difference between the two tissue types (P = 0.821). Normal range values (>0.9 and <1.2) were established based on the diseased normal data, setting upper and lower limits by calculating two standard deviations from the mean (Table 3).
TABLE 3 Normal Range Determinations
Figure imgf000021_0001
Definition of a normal range creates a threshold for determining amplification status for HER2lneu. In addition, correlation between normal values derived from non-diseased and diseased tissue provides justification for use of a moφhologicallynormal cellular area in a tumorbiopsy as an internal biological control for specificity in that specimen. Patient Cohort
Using the described system combining moφhological staining and FISH, a total of 46 cases of known invasive breast carcinoma were evaluated. Serial sections from each were stained immunohistochemically with the 4D5 antibody to HERllneu for comparative puφoses. HER2lneu and chromosome 17 fluorescent nuclear signals were scored by two analysts and the mean ratios calculated. Ratios for HER2lneu amplification were assigned to one of two ranges, either moderately amplified borderline or highly amplified. The lower limit of the moderate amplificationrange (1.3) was established statistically by determining at what value the ratio differed significantly from normal. The upper limit for moderately amplified status (2.0) was taken from previously published data which defined true amplification as having a ratio of >2.0 [Pauletti et al Oncogene 13:63-72 (1996)]. The relationship between the scoring systems used for HERllneu amplification by FISH and overexpression by IHC is depicted in Table 4.
TABLE 4 Scoring System for HER2 Amplification/Expression
Figure imgf000022_0001
Immunohistochemistry scores were based on membrane staining only; specimens which exhibited cytoplasmic staining (n=4) were excluded from the study data. Correlations between FISH mean ratios and irnmunohistochemistryscores showed a positive correlation coefficient (0.760) and a low P value (O.OO 1 ). Both techniques identified unambiguous amplification or overexpression of HER2lneu equally well with two exceptions (Figure 5). One case which produced an IHC score of 2+ fell into the moderately amplified range by FISH (ratio=l .7), and one case which showed high amplification by FISH (ratio=9.8) had an IHC score of 1+. FISH quantification of HER2lneu placed ten specimens in the moderate amplification range. Immunohistochemical evaluation identified only four specimens showing moderate positivity (score of 1+), suggesting a less than optimal sensitivity associated with the hum4D5-8 antibody.
Although the present invention has been described in some detail by way of illustration for the puφoses of clarity of understanding, it will be apparent that certain changes and modifications may be practiced within the scope of the appended claims. Such modifications which may be apparent to a person skilled in the art are intended to be within the scope of the invention.

Claims

What is claimed is:
1. A method of correlating cellular moφhology with the presence of a cellulartarget nucleic acid sequence in a section of a tissue sample comprising the following steps:
(a) staining the section of tissue sample with a moφhological stain;
(b) determining cellular moφhology in the section of tissue sample;
(c) hybridizing a first fluorescently labeled nucleic acid probe to the target nucleic acid sequence in the same section of tissue sample;
(d) detecting the presence of the first nucleic acid probe in the section of tissue sample; and
(e) correlating step (b) with step (d).
2. The method of claim 1, wherein the first nucleic acid probe is constructed to hybridize to the target nucleic acid sequence indicating a genetic abnormality selected from the group consisting of amplification, addition, substitution, translocation and deletion.
3. The method of claim 1, wherein amplification of the target nucleic acid sequence is determined in step (d).
4. The method of claim 3, wherein the target nucleic acid sequence is HER2lneu gene.
5. The method of claim 1 , wherein the moφhological stain used in step (a) does not significantly autofluoresce at the same wavelength as a fluorescent label of the first nucleic acid probe.
6. The method of claim 1 , further comprising hybridizing a second fluorescently labeled nucleic acid probe to a nucleic acid sequence in the section of tissue sample, wherein the second nucleic acid probe comprises a fluorescent label distinguishable from a fluorescent label of the first nucleic acid probe.
7. The method of claim 6, wherein the second nucleic acid probe determines chromosome copy number.
8. The method of claim 1, wherein the moφhological stain used in step (a) comprises xanthine dye and thiazine dye.
9. The method of claim 8, wherein the moφhological stain is HEMA 3®.
10. The method of claim 1 , wherein the tissue sample is selected from the group consisting of breast, prostate, ovary, colon, lung, endometrium, stomach, salivary gland and pancreas tissue sample.
1 1. The method of claim 1 , wherein the target nucleic acid sequence is selected from the group consisting of HER21 neu gene and the centromere of chromosome 17.
12. The method of claim 1 , wherein the first nucleic acid probe is labeled with a fluorescent label selected from the group consisting of Texas Red, fluorescein, phycocrytherin, rhodamine, phycocyanin, dansyl, umbelliferone, SPECTRUM GREEN®, SPECTRUM ORANGE® and derivatives of any of the above labels.
13. The method of claim 1, wherein the section of tissue sample is not destained prior to step (c).
14. The method of claim 1 , wherein the section of tissue sample is obtained from fixed, paraffin- embedded tissue sample.
15. A method of correlating the presence of a cellular target protein with the presence of a cellular target nucleic acid sequence in a section of a tissue sample comprising the following steps:
(a) contacting the section of sample tissue with an antibody which specifically binds to the target protein; (b) determining binding of the antibody to the section of tissue sample;
(c) hybridizing a fluorescently labeled nucleic acid probe to the target nucleic acid sequence in the same section of tissue sample;
(d) detecting the presence of the labeled nucleic acid probe in the section of tissue sample; and
(e) correlating step (b) with step (d).
16. A kit comprising: (a) a moφhological stain; (b) a fluorescently labeled probe complementary to a genetic abnormality; and (c) instructions for applyingthe stain (a) and probe (b) to the same section of tissue sample.
17. The kit of claim 16, wherein the probe hybridizes to HER2/neu nucleic acid.
18. The kit of claim 16, wherein the moφhological stain is HEMA 3®.
19. A kit comprising: (a) a primary antibody which specifically binds a cellulartarget protein; (b) a fluorescently labeled probe complementary to a genetic abnormality; and (c) instructions for applying the antibody (a) and probe (b) to the same section of tissue sample.
20. The kit of claim 19, further comprising a secondary antibody which specifically binds to the primary antibody.
21. The kit of claim 20, wherein the secondary antibody is labeled with an enzymatic label which catalyzes chemical alteration of a substrate compound.
22. The kit of claim 19, wherein the probe hybridizes to HER2/neu nucleic acid and the primary antibody specifically binds to HER2 protein.
23. The kit of claim 22, further comprising instructions for applying the probe to a section of tissue sample having a score of 1+ or 2+ for HER2 Protein Staining Intensity.
PCT/US1999/022909 1998-10-07 1999-10-01 Tissue analysis and kits therefor WO2000020641A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU64097/99A AU6409799A (en) 1998-10-07 1999-10-01 Tissue analysis and kits therefor

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US09/167,691 US6573043B1 (en) 1998-10-07 1998-10-07 Tissue analysis and kits therefor
US09/167,691 1998-10-07

Publications (1)

Publication Number Publication Date
WO2000020641A1 true WO2000020641A1 (en) 2000-04-13

Family

ID=22608409

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1999/022909 WO2000020641A1 (en) 1998-10-07 1999-10-01 Tissue analysis and kits therefor

Country Status (3)

Country Link
US (7) US6573043B1 (en)
AU (1) AU6409799A (en)
WO (1) WO2000020641A1 (en)

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008064067A2 (en) * 2006-11-16 2008-05-29 General Electric Company Method for the sequential analysis of different components in a single biological sample and reagents therefor
EP2116262A2 (en) 2000-05-19 2009-11-11 Genentech, Inc. Gene detection assay for improving the likelihood of an effective response to an ErbB antagonist cancer therapy
US7629125B2 (en) 2006-11-16 2009-12-08 General Electric Company Sequential analysis of biological samples
WO2010022332A1 (en) 2008-08-22 2010-02-25 Ventana Medical Systems, Inc. Method for chromogenic detection of two or more target molecules in a single sample
EP2450457A1 (en) * 2010-11-09 2012-05-09 Olympus Corporation Method of analyzing genetically abnormals cells
CN102607930A (en) * 2012-03-13 2012-07-25 中国水产科学研究院黑龙江水产研究所 Fish germ cell orientated embedding technology
US8305579B2 (en) 2006-11-16 2012-11-06 Thomas Pirrie Treynor Sequential analysis of biological samples
US8609354B2 (en) 2010-03-04 2013-12-17 Olli CARPEN Method for selecting patients for treatment with an EGFR inhibitor
US9201063B2 (en) 2006-11-16 2015-12-01 General Electric Company Sequential analysis of biological samples
US9677125B2 (en) 2009-10-21 2017-06-13 General Electric Company Detection of plurality of targets in biological samples
US9944972B2 (en) 2014-03-11 2018-04-17 President And Fellows Of Harvard College High-throughput and highly multiplexed imaging with programmable nucleic acid probes
CN107917832A (en) * 2017-12-04 2018-04-17 山西农业大学 The production method of fish ovary tissue paraffin section
US10024796B2 (en) 2010-10-29 2018-07-17 President And Fellows Of Harvard College Nucleic acid nanostructure barcode probes
US11092606B2 (en) 2015-08-07 2021-08-17 President And Fellows Of Harvard College Super resolution imaging of protein-protein interactions
US11413296B2 (en) 2005-11-12 2022-08-16 The Regents Of The University Of California Viscous budesonide for the treatment of inflammatory diseases of the gastrointestinal tract
US11536715B2 (en) 2013-07-30 2022-12-27 President And Fellows Of Harvard College Quantitative DNA-based imaging and super-resolution imaging
US11754562B2 (en) 2016-12-09 2023-09-12 Ultivue, Inc. Methods for multiplex imaging using labeled nucleic acid imaging agents

Families Citing this family (146)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0878552A1 (en) * 1997-05-13 1998-11-18 Erasmus Universiteit Rotterdam Molecular detection of chromosome aberrations
CA2327542C (en) * 1998-05-04 2011-11-22 Dako A/S Method and probes for the detection of chromosome aberrations
US20030086924A1 (en) * 1999-06-25 2003-05-08 Genentech, Inc. Treatment with anti-ErbB2 antibodies
US20040013667A1 (en) * 1999-06-25 2004-01-22 Genentech, Inc. Treatment with anti-ErbB2 antibodies
US6949245B1 (en) * 1999-06-25 2005-09-27 Genentech, Inc. Humanized anti-ErbB2 antibodies and treatment with anti-ErbB2 antibodies
AU2001259271A1 (en) * 2000-04-28 2001-11-12 Millennium Pharmaceuticals, Inc. 14094, a novel human trypsin family member and uses thereof
US7128903B2 (en) * 2001-10-03 2006-10-31 Innovative Pharmaceutical Concepts (Ipc) Inc. Pharmaceutical preparations useful for treating tumors and lesions of the skin and the mucous membranes and methods and kits using same
EP1451304A4 (en) * 2001-11-07 2006-09-20 Bioview Ltd Kits and methods for preparing cell samples optimized for dual staining
US20040229294A1 (en) * 2002-05-21 2004-11-18 Po-Ying Chan-Hui ErbB surface receptor complexes as biomarkers
US7402397B2 (en) 2002-05-21 2008-07-22 Monogram Biosciences, Inc. Detecting and profiling molecular complexes
US20040229380A1 (en) * 2002-05-21 2004-11-18 Po-Ying Chan-Hui ErbB heterodimers as biomarkers
US20050037406A1 (en) * 2002-06-12 2005-02-17 De La Torre-Bueno Jose Methods and apparatus for analysis of a biological specimen
US7272252B2 (en) * 2002-06-12 2007-09-18 Clarient, Inc. Automated system for combining bright field and fluorescent microscopy
CA2518569C (en) 2003-03-10 2011-11-15 Expression Pathology, Inc. Liquid tissue preparation from histopathologically processed biological samples, tissues and cells
US8062897B2 (en) 2003-07-21 2011-11-22 Aureon Laboratories, Inc. Diagnostic histopathology using multiplex gene expression FISH
EP1681983A4 (en) * 2003-10-14 2008-12-10 Monogram Biosciences Inc Receptor tyrosine kinase signaling pathway analysis for diagnosis and therapy
JO3000B1 (en) 2004-10-20 2016-09-05 Genentech Inc Antibody Formulations.
US20060115862A1 (en) * 2004-11-17 2006-06-01 Duke University Anti-tenascin monoclonal antibody immunoassays and diagnostic kits
BRPI0518086A (en) * 2004-12-07 2008-10-28 Genentech Inc methods for cancer treatment, methods for evaluating phosphorylation or her activation and method of identification
EP1846030B1 (en) 2005-01-21 2018-11-21 Genentech, Inc. Fixed dosing of her antibodies
MX2007009566A (en) * 2005-02-09 2009-02-16 Genentech Inc Inhibiting her2 shedding with matrix metalloprotease antagonists.
NZ590431A (en) 2005-02-23 2012-08-31 Genentech Inc Extending time to disease progression or survival in cancer patients using a HER dimerization inhibitor
US20060204505A1 (en) * 2005-03-08 2006-09-14 Sliwkowski Mark X Methods for identifying tumors responsive to treatment with HER dimerization inhibitors (HDIs)
JP2006316040A (en) 2005-05-13 2006-11-24 Genentech Inc Herceptin(r) adjuvant treatment
PE20070207A1 (en) * 2005-07-22 2007-03-09 Genentech Inc COMBINED TREATMENT OF TUMORS THAT EXPRESS HER
US9957569B2 (en) * 2005-09-12 2018-05-01 The Regents Of The University Of Michigan Recurrent gene fusions in prostate cancer
US7718369B2 (en) 2005-09-12 2010-05-18 The Regents Of The University Of Michigan Recurrent gene fusions in prostate cancer
US20120208824A1 (en) 2006-01-20 2012-08-16 Cell Signaling Technology, Inc. ROS Kinase in Lung Cancer
US8131476B2 (en) * 2006-08-07 2012-03-06 General Electric Company System and method for co-registering multi-channel images of a tissue micro array
US8060348B2 (en) * 2006-08-07 2011-11-15 General Electric Company Systems for analyzing tissue samples
US20080032321A1 (en) * 2006-08-07 2008-02-07 General Electric Company System and methods for analyzing images of tissue samples
PE20090681A1 (en) 2007-03-02 2009-06-10 Genentech Inc PREDICTION OF RESPONSE TO A HER INHIBITOR
US9551033B2 (en) 2007-06-08 2017-01-24 Genentech, Inc. Gene expression markers of tumor resistance to HER2 inhibitor treatment
US20100298156A1 (en) * 2007-06-08 2010-11-25 Si Tuen Lee-Hoeflich Gene expression markers of tumor resistance to her2 inhibitor treatment
WO2009009432A2 (en) 2007-07-06 2009-01-15 The Regents Of The University Of Michigan Recurrent gene fusions in prostate cancer
JP5433572B2 (en) * 2007-07-06 2014-03-05 ザ リージェンツ オブ ザ ユニバーシティ オブ ミシガン MIPOL1-ETV1 gene rearrangement
TWI472339B (en) 2008-01-30 2015-02-11 Genentech Inc Composition comprising antibody that binds to domain ii of her2 and acidic variants thereof
JP2011511949A (en) * 2008-02-12 2011-04-14 ザ・ブリガーム・アンド・ウーメンズ・ホスピタル・インコーポレーテッド FISH assay for fusion of EML4 and ALK in lung cancer
WO2009117138A2 (en) * 2008-03-21 2009-09-24 Spring Bioscience Corporation Counterstaining method for dual-stain in situ hybridization
US20090304714A1 (en) * 2008-03-25 2009-12-10 The Regents Of The University Of Michigan IKKi Inhibitor Therapies and Screening Methods, and Related IKKi Diagnostics
EP2806054A1 (en) 2008-05-28 2014-11-26 Genomedx Biosciences Inc. Systems and methods for expression-based discrimination of distinct clinical disease states in prostate cancer
BRPI0812682A2 (en) * 2008-06-16 2010-06-22 Genentech Inc metastatic breast cancer treatment
CA2749113A1 (en) 2009-01-09 2010-07-15 The Regents Of The University Of Michigan Recurrent gene fusions in cancer
EP2881402B1 (en) 2009-02-12 2017-05-10 Cell Signaling Technology, Inc. Mutant ROS expression in human liver cancer
KR20130080871A (en) 2009-03-20 2013-07-15 제넨테크, 인크. Bispecific anti-her antibodies
WO2010125566A2 (en) 2009-04-27 2010-11-04 Technion Research And Development Foundation Ltd. Markers for cancer detection
JP5705836B2 (en) 2009-05-29 2015-04-22 エフ・ホフマン−ラ・ロシュ・アクチェンゲゼルシャフト Modulators for HER2 signaling in gastric cancer patients expressing HER2
KR20120108967A (en) 2009-09-16 2012-10-05 제넨테크, 인크. Coiled coil and/or tether containing protein complexes and uses thereof
WO2011034906A2 (en) 2009-09-17 2011-03-24 The Regents Of The University Of Michigan Recurrent gene fusions in prostate cancer
FR2950360B1 (en) * 2009-09-22 2011-11-25 Centre Nat Rech Scient CELL IMAGING METHOD FOR VISUALIZATION OF MICROARN BIOGENESIS IN CELLS
SG183333A1 (en) 2010-02-18 2012-09-27 Genentech Inc Neuregulin antagonists and use thereof in treating cancer
TW201138821A (en) 2010-03-26 2011-11-16 Roche Glycart Ag Bispecific antibodies
US10787701B2 (en) 2010-04-05 2020-09-29 Prognosys Biosciences, Inc. Spatially encoded biological assays
PL2556171T3 (en) 2010-04-05 2016-04-29 Prognosys Biosciences Inc Spatially encoded biological assays
US20190300945A1 (en) 2010-04-05 2019-10-03 Prognosys Biosciences, Inc. Spatially Encoded Biological Assays
US8383793B2 (en) 2010-04-15 2013-02-26 St. Jude Children's Research Hospital Methods and compositions for the diagnosis and treatment of cancer resistant to anaplastic lymphoma kinase (ALK) kinase inhibitors
WO2011146568A1 (en) 2010-05-19 2011-11-24 Genentech, Inc. Predicting response to a her inhibitor
RU2550925C2 (en) 2010-07-07 2015-05-20 Зе Реджентс Оф Зе Юниверсити Оф Мичиган Diagnosing and treating breast cancer
US20120101108A1 (en) 2010-08-06 2012-04-26 Cell Signaling Technology, Inc. Anaplastic Lymphoma Kinase In Kidney Cancer
US9005907B2 (en) 2010-10-01 2015-04-14 St. Jude Children's Research Hospital Methods and compositions for typing molecular subgroups of medulloblastoma
US8945556B2 (en) 2010-11-19 2015-02-03 The Regents Of The University Of Michigan RAF gene fusions
EP2640854B1 (en) 2010-11-19 2018-02-21 The Regents Of The University Of Michigan PCAT1 ncRNA AND USES THEREOF
US20130245233A1 (en) 2010-11-24 2013-09-19 Ming Lei Multispecific Molecules
EP2655413B1 (en) 2010-12-23 2019-01-16 F.Hoffmann-La Roche Ag Polypeptide-polynucleotide-complex and its use in targeted effector moiety delivery
ES2659763T3 (en) 2011-02-14 2018-03-19 The Regents Of The University Of Michigan Compositions and procedures for the treatment of obesity and related disorders
GB201106254D0 (en) 2011-04-13 2011-05-25 Frisen Jonas Method and product
CA2842493A1 (en) 2011-08-02 2013-02-07 James Gail Christensen Crizotinib for use in the treatment of cancer
CN103890007A (en) 2011-08-17 2014-06-25 霍夫曼-拉罗奇有限公司 Neuregulin antibodies and uses thereof
EP2755019B1 (en) * 2011-09-09 2018-01-03 Konica Minolta, Inc. Biological substance detection method
IN2014CN02703A (en) 2011-10-14 2015-07-03 Hoffmann La Roche
US9327023B2 (en) 2011-10-25 2016-05-03 The Regents Of The University Of Michigan HER2 targeting agent treatment in non-HER2-amplified cancers having HER2 expressing cancer stem cells
CA2853760A1 (en) 2011-11-04 2013-05-10 Oslo Universitetssykehus Hf Methods and biomarkers for analysis of colorectal cancer
EP2785864A2 (en) 2011-11-30 2014-10-08 F. Hoffmann-La Roche AG Erbb3 mutations in cancer
WO2013083810A1 (en) 2011-12-09 2013-06-13 F. Hoffmann-La Roche Ag Identification of non-responders to her2 inhibitors
US8568991B2 (en) 2011-12-23 2013-10-29 General Electric Company Photoactivated chemical bleaching of dyes
US9176032B2 (en) 2011-12-23 2015-11-03 General Electric Company Methods of analyzing an H and E stained biological sample
JP2015503923A (en) 2012-01-09 2015-02-05 オスロ ウニヴェルスィテーツスィーケフース ハーエフOslo Universitetssykehus Hf Methods and biomarkers for the analysis of colorectal cancer
WO2013119966A2 (en) 2012-02-10 2013-08-15 Genentech, Inc. Single-chain antibodies and other heteromultimers
CN104220876A (en) 2012-02-21 2014-12-17 奥斯陆大学医院 Methods and biomarkers for detection and prognosis of cervical cancer
US20150111758A1 (en) 2012-03-06 2015-04-23 Oslo Universitetssykehus Hf Gene signatures associated with efficacy of postmastectomy radiotherapy in breast cancer
MX2014011500A (en) 2012-03-27 2014-12-05 Genentech Inc Diagnosis and treatments relating to her3 inhibitors.
WO2013158859A1 (en) 2012-04-18 2013-10-24 Cell Signaling Technology, Inc. Egfr and ros1 in cancer
BR112014028368A2 (en) 2012-06-27 2017-11-14 Hoffmann La Roche method of producing antibody fc region conjugate, antibody fc region conjugate and pharmaceutical formulation
ES2597228T3 (en) 2012-06-27 2017-01-17 F. Hoffmann-La Roche Ag Procedure for the selection and production of objectification entities, such as targets, highly selective and multi-specific, customized, which contain at least two different binding entities, and use of these
WO2014001326A1 (en) 2012-06-27 2014-01-03 F. Hoffmann-La Roche Ag Method for the selection and production of tailor-made, selective and multi-specific therapeutic molecules comprising at least two different targeting entities and uses thereof
CA2881627A1 (en) 2012-08-16 2014-02-20 Genomedx Biosciences Inc. Cancer diagnostics using biomarkers
CA2883264A1 (en) 2012-10-15 2014-04-24 Universitat Zurich Prorektorat Mnw Bispecific her2 ligands for cancer therapy
EP2719706A1 (en) 2012-10-15 2014-04-16 Universität Zürich Bispecific HER2 ligands for cancer therapy
EP2909341A2 (en) 2012-10-18 2015-08-26 Oslo Universitetssykehus HF Biomarkers for cervical cancer
CA2889298C (en) 2012-11-30 2024-01-02 Anton Belousov Identification of patients in need of pd-l1 inhibitor cotherapy
US9135694B2 (en) 2012-12-04 2015-09-15 General Electric Company Systems and methods for using an immunostaining mask to selectively refine ISH analysis results
JP2016513794A (en) 2013-03-06 2016-05-16 ゼネラル・エレクトリック・カンパニイ Method for analyzing H & E stained biological sample
US9815904B2 (en) 2013-04-16 2017-11-14 Genetech, Inc. Pertuzumab variants and evaluation thereof
WO2014179448A2 (en) 2013-05-01 2014-11-06 Five Prime Therapeutics, Inc. Methods of treating cancer
US8946424B2 (en) 2013-05-02 2015-02-03 The Regents Of The University Of Michigan Deuterated amlexanox
WO2014210225A1 (en) 2013-06-25 2014-12-31 Prognosys Biosciences, Inc. Methods and systems for determining spatial patterns of biological targets in a sample
WO2015124702A1 (en) * 2014-02-21 2015-08-27 Ventana Medical Systems, Inc. Single-stranded oligonucleotide probes for chromosome or gene copy enumeration
MX2016014007A (en) 2014-04-25 2017-01-11 Genentech Inc Methods of treating early breast cancer with trastuzumab-mcc-dm1 and pertuzumab.
EP3134084B1 (en) 2014-04-25 2021-03-17 Exelixis, Inc. Method of treating lung adenocarcinoma
ES2764111T3 (en) 2014-12-03 2020-06-02 Hoffmann La Roche Multispecific antibodies
EP3901282B1 (en) 2015-04-10 2023-06-28 Spatial Transcriptomics AB Spatially distinguished, multiplex nucleic acid analysis of biological specimens
US11406715B2 (en) 2015-05-30 2022-08-09 Genentech, Inc. Methods of treating HER2-positive metastatic breast cancer
WO2016196478A1 (en) 2015-06-01 2016-12-08 St. Jude Children's Research Hospital Methods and compositions for prognostications and/or clinical management of graft-versus-host disease and transplant rejection
CN106338423B (en) 2015-07-10 2020-07-14 三斯坎公司 Spatial multiplexing of histological staining
WO2017087280A1 (en) 2015-11-16 2017-05-26 Genentech, Inc. Methods of treating her2-positive cancer
WO2017132538A1 (en) 2016-01-29 2017-08-03 The Regents Of The University Of Michigan Amlexanox analogs
EP3454863A1 (en) 2016-05-10 2019-03-20 INSERM (Institut National de la Santé et de la Recherche Médicale) Combinations therapies for the treatment of cancer
US11414708B2 (en) 2016-08-24 2022-08-16 Decipher Biosciences, Inc. Use of genomic signatures to predict responsiveness of patients with prostate cancer to post-operative radiation therapy
US20180134803A1 (en) 2016-11-04 2018-05-17 Genentech, Inc. Treatment of her2-positive breast cancer
EP3562844A1 (en) 2016-12-28 2019-11-06 Genentech, Inc. Treatment of advanced her2 expressing cancer
WO2018127786A1 (en) 2017-01-06 2018-07-12 Oslo Universitetssykehus Hf Compositions and methods for determining a treatment course of action
UA123292C2 (en) 2017-01-17 2021-03-10 Дженентек, Інк. Subcutaneous her2 antibody formulations
US11208697B2 (en) 2017-01-20 2021-12-28 Decipher Biosciences, Inc. Molecular subtyping, prognosis, and treatment of bladder cancer
LT3589661T (en) 2017-03-02 2024-02-12 Genentech, Inc. Adjuvant treatment of her2-positive breast cancer
WO2018165600A1 (en) 2017-03-09 2018-09-13 Genomedx Biosciences, Inc. Subtyping prostate cancer to predict response to hormone therapy
CA3059241A1 (en) 2017-04-24 2018-11-01 Genentech, Inc. Erbb2/her2 mutations in the transmbrane or juxtamembrane domain
AU2018266733A1 (en) 2017-05-12 2020-01-16 Veracyte, Inc. Genetic signatures to predict prostate cancer metastasis and identify tumor aggressiveness
WO2019202536A1 (en) 2018-04-18 2019-10-24 St. Jude Children's Research Hospital Genotyping assays to identify mutations in xaf1
US11519033B2 (en) 2018-08-28 2022-12-06 10X Genomics, Inc. Method for transposase-mediated spatial tagging and analyzing genomic DNA in a biological sample
KR20210075080A (en) 2018-10-08 2021-06-22 유니베르시태트 취리히 HER2-binding tetrameric polypeptide
US11926867B2 (en) 2019-01-06 2024-03-12 10X Genomics, Inc. Generating capture probes for spatial analysis
US11649485B2 (en) 2019-01-06 2023-05-16 10X Genomics, Inc. Generating capture probes for spatial analysis
WO2021092433A2 (en) 2019-11-08 2021-05-14 10X Genomics, Inc. Enhancing specificity of analyte binding
AU2020412766A1 (en) 2019-12-23 2022-06-09 10X Genomics, Inc. Methods for spatial analysis using RNA-templated ligation
US11732299B2 (en) 2020-01-21 2023-08-22 10X Genomics, Inc. Spatial assays with perturbed cells
US11702693B2 (en) 2020-01-21 2023-07-18 10X Genomics, Inc. Methods for printing cells and generating arrays of barcoded cells
US11898205B2 (en) 2020-02-03 2024-02-13 10X Genomics, Inc. Increasing capture efficiency of spatial assays
US11732300B2 (en) 2020-02-05 2023-08-22 10X Genomics, Inc. Increasing efficiency of spatial analysis in a biological sample
US11891654B2 (en) 2020-02-24 2024-02-06 10X Genomics, Inc. Methods of making gene expression libraries
CN115916999A (en) 2020-04-22 2023-04-04 10X基因组学有限公司 Methods for spatial analysis using targeted RNA depletion
WO2021237087A1 (en) 2020-05-22 2021-11-25 10X Genomics, Inc. Spatial analysis to detect sequence variants
EP4153775A1 (en) 2020-05-22 2023-03-29 10X Genomics, Inc. Simultaneous spatio-temporal measurement of gene expression and cellular activity
WO2021242834A1 (en) 2020-05-26 2021-12-02 10X Genomics, Inc. Method for resetting an array
EP4162074A1 (en) 2020-06-08 2023-04-12 10X Genomics, Inc. Methods of determining a surgical margin and methods of use thereof
WO2021252591A1 (en) 2020-06-10 2021-12-16 10X Genomics, Inc. Methods for determining a location of an analyte in a biological sample
WO2021263111A1 (en) 2020-06-25 2021-12-30 10X Genomics, Inc. Spatial analysis of dna methylation
US11761038B1 (en) 2020-07-06 2023-09-19 10X Genomics, Inc. Methods for identifying a location of an RNA in a biological sample
WO2022011276A1 (en) * 2020-07-09 2022-01-13 Axon Imaging, Llc Advanced nervous tissue imaging system
CN112014187A (en) * 2020-09-07 2020-12-01 四川大学华西医院 Preparation method of section of non-decalcified osteochondral tissue and cell tracing and RNAscope combined testing method thereof
US11926822B1 (en) 2020-09-23 2024-03-12 10X Genomics, Inc. Three-dimensional spatial analysis
US11827935B1 (en) 2020-11-19 2023-11-28 10X Genomics, Inc. Methods for spatial analysis using rolling circle amplification and detection probes
WO2022140028A1 (en) 2020-12-21 2022-06-30 10X Genomics, Inc. Methods, compositions, and systems for capturing probes and/or barcodes
EP4323442A1 (en) * 2021-04-16 2024-02-21 Advanced Cell Diagnostics, Inc. Methods and compositions for reducing autofluorescence
WO2023021330A1 (en) 2021-08-16 2023-02-23 University Of Oslo Compositions and methods for determining a treatment course of action
WO2023034489A1 (en) 2021-09-01 2023-03-09 10X Genomics, Inc. Methods, compositions, and kits for blocking a capture probe on a spatial array
WO2023152568A2 (en) 2022-02-10 2023-08-17 Oslo Universitetssykehus Hf Compositions and methods for characterizing lung cancer

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994009022A1 (en) * 1992-10-09 1994-04-28 Oncor, Inc. Methods for the detection of chromosome structural abnormalities by in situ hybridization to fixed tissue
WO1995003428A1 (en) * 1993-07-20 1995-02-02 University Of Massachusetts Medical Center In vivo nucleic acid hybridization method
FR2711671A1 (en) * 1993-10-22 1995-05-05 Centre Nat Rech Scient Process for the detection and quantification of a nucleotide sequence in a cell population
WO1997008343A1 (en) * 1995-08-30 1997-03-06 Northwestern University Method of sumultaneously detecting amplified nucleic acid sequences and cellular antigens in cells
EP0801306A1 (en) * 1996-04-10 1997-10-15 Becton, Dickinson and Company Method for immunohistochemically quantitating co-localized molecules
WO1998002577A1 (en) * 1996-07-15 1998-01-22 The Children's Medical Center Corporation Method to co-detect introduced genes and their products

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4968603A (en) 1986-12-31 1990-11-06 The Regents Of The University Of California Determination of status in neoplastic disease
WO1994000764A1 (en) 1992-06-26 1994-01-06 THE UNITED STATES OF AMERICA, represented by THE SECRETARY OF HEALTH AND HUMAN SERVICES The met proto-oncogene and a method for predicting breast cancer progression
US5801005A (en) * 1993-03-17 1998-09-01 University Of Washington Immune reactivity to HER-2/neu protein for diagnosis of malignancies in which the HER-2/neu oncogene is associated
US5660982A (en) * 1994-10-04 1997-08-26 Tryggvason; Karl Laminin chains: diagnostic uses
US5846749A (en) 1994-10-12 1998-12-08 The Regents Of The University Of California Quantitative measurement of tissue protein identified by immunohistochemistry and standardized protein determination
US5750340A (en) 1995-04-07 1998-05-12 University Of New Mexico In situ hybridization solution and process
US6165734A (en) 1995-12-12 2000-12-26 Applied Spectral Imaging Ltd. In-situ method of analyzing cells
US6027890A (en) * 1996-01-23 2000-02-22 Rapigene, Inc. Methods and compositions for enhancing sensitivity in the analysis of biological-based assays
WO2000031543A1 (en) 1998-11-25 2000-06-02 Bayer Corporation Measurement of hydride using chemiluminescent acridinium compounds

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994009022A1 (en) * 1992-10-09 1994-04-28 Oncor, Inc. Methods for the detection of chromosome structural abnormalities by in situ hybridization to fixed tissue
WO1995003428A1 (en) * 1993-07-20 1995-02-02 University Of Massachusetts Medical Center In vivo nucleic acid hybridization method
FR2711671A1 (en) * 1993-10-22 1995-05-05 Centre Nat Rech Scient Process for the detection and quantification of a nucleotide sequence in a cell population
WO1997008343A1 (en) * 1995-08-30 1997-03-06 Northwestern University Method of sumultaneously detecting amplified nucleic acid sequences and cellular antigens in cells
EP0801306A1 (en) * 1996-04-10 1997-10-15 Becton, Dickinson and Company Method for immunohistochemically quantitating co-localized molecules
WO1998002577A1 (en) * 1996-07-15 1998-01-22 The Children's Medical Center Corporation Method to co-detect introduced genes and their products

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
GARDINER M ET AL: "Development of a technique that allows simultaneous assessment of the morphology and gene amplification by FISH: application to HER-2/Neu amplification in breast cancer", MODERN PATHOLOGY, vol. 12, no. 1, January 1999 (1999-01-01), pages 191A, XP000878899 *
PRESS M ET AL: "Amplification and overexpression of HER-2/neu in carcinomas of the salivary gland: correlation with poor prognosis", CANCER RESEARCH, vol. 54, 1 November 1994 (1994-11-01), pages 5675 - 82, XP000877239 *
RATCLIFFE N ET AL: "The combination of In Situ Hybridization and Immunohistochemical analysis", MODERN PATHOLOGY, vol. 10, no. 12, December 1997 (1997-12-01), pages 1247 - 52, XP000884734 *

Cited By (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104383535A (en) * 2000-05-19 2015-03-04 杰南技术公司 Gene detection assay for improving the likelihood of an effective response to an erbb antagonist cancer therapy
EP2116262A2 (en) 2000-05-19 2009-11-11 Genentech, Inc. Gene detection assay for improving the likelihood of an effective response to an ErbB antagonist cancer therapy
US11413296B2 (en) 2005-11-12 2022-08-16 The Regents Of The University Of California Viscous budesonide for the treatment of inflammatory diseases of the gastrointestinal tract
JP2010510493A (en) * 2006-11-16 2010-04-02 ゼネラル・エレクトリック・カンパニイ Continuous analysis of biological samples
US8305579B2 (en) 2006-11-16 2012-11-06 Thomas Pirrie Treynor Sequential analysis of biological samples
US9518982B2 (en) 2006-11-16 2016-12-13 General Electric Company Sequential analysis of biological samples
US7741045B2 (en) 2006-11-16 2010-06-22 General Electric Company Sequential analysis of biological samples
US7741046B2 (en) 2006-11-16 2010-06-22 General Electric Company Sequential analysis of biological samples
EP2381257A1 (en) * 2006-11-16 2011-10-26 General Electric Company Sequential analysis of biological samples
EP2383574A3 (en) * 2006-11-16 2011-11-30 General Electric Company Sequential analysis of biological samples
US7629125B2 (en) 2006-11-16 2009-12-08 General Electric Company Sequential analysis of biological samples
US9201063B2 (en) 2006-11-16 2015-12-01 General Electric Company Sequential analysis of biological samples
WO2008064067A3 (en) * 2006-11-16 2008-07-10 Gen Electric Method for the sequential analysis of different components in a single biological sample and reagents therefor
US8822147B2 (en) 2006-11-16 2014-09-02 General Electric Company Sequential analysis of biological samples
WO2008064067A2 (en) * 2006-11-16 2008-05-29 General Electric Company Method for the sequential analysis of different components in a single biological sample and reagents therefor
AU2007323794B2 (en) * 2006-11-16 2013-08-22 Leica Microsystems Cms Gmbh Method for the sequential analysis of different components in a single biological sample and reagents therefor
AU2009282760B2 (en) * 2008-08-22 2013-08-15 Ventana Medical Systems, Inc. Method for chromogenic detection of two or more target molecules in a single sample
US8481270B2 (en) 2008-08-22 2013-07-09 Ventana Medical Systems, Inc. Method for chromogenic detection of two or more target molecules in a single sample
WO2010022332A1 (en) 2008-08-22 2010-02-25 Ventana Medical Systems, Inc. Method for chromogenic detection of two or more target molecules in a single sample
US9677125B2 (en) 2009-10-21 2017-06-13 General Electric Company Detection of plurality of targets in biological samples
US8609354B2 (en) 2010-03-04 2013-12-17 Olli CARPEN Method for selecting patients for treatment with an EGFR inhibitor
US10024796B2 (en) 2010-10-29 2018-07-17 President And Fellows Of Harvard College Nucleic acid nanostructure barcode probes
US10876971B2 (en) 2010-10-29 2020-12-29 President And Fellows Of Harvard College Nucleic acid nanostructure barcode probes
EP2450457A1 (en) * 2010-11-09 2012-05-09 Olympus Corporation Method of analyzing genetically abnormals cells
CN102607930A (en) * 2012-03-13 2012-07-25 中国水产科学研究院黑龙江水产研究所 Fish germ cell orientated embedding technology
US11536715B2 (en) 2013-07-30 2022-12-27 President And Fellows Of Harvard College Quantitative DNA-based imaging and super-resolution imaging
US9944972B2 (en) 2014-03-11 2018-04-17 President And Fellows Of Harvard College High-throughput and highly multiplexed imaging with programmable nucleic acid probes
US10190151B2 (en) 2014-03-11 2019-01-29 President And Fellows Of Harvard College High-throughput and highly multiplexed imaging with programmable nucleic acid probes
US10294510B2 (en) 2014-03-11 2019-05-21 President And Fellows Of Harvard College High-throughput and highly multiplexed imaging with programmable nucleic acid probes
US11092606B2 (en) 2015-08-07 2021-08-17 President And Fellows Of Harvard College Super resolution imaging of protein-protein interactions
US11754562B2 (en) 2016-12-09 2023-09-12 Ultivue, Inc. Methods for multiplex imaging using labeled nucleic acid imaging agents
CN107917832A (en) * 2017-12-04 2018-04-17 山西农业大学 The production method of fish ovary tissue paraffin section

Also Published As

Publication number Publication date
US7129051B2 (en) 2006-10-31
US7919254B2 (en) 2011-04-05
US20100120053A1 (en) 2010-05-13
US7468252B2 (en) 2008-12-23
US20080050748A1 (en) 2008-02-28
US20030152987A1 (en) 2003-08-14
US7674589B2 (en) 2010-03-09
US20050100944A1 (en) 2005-05-12
US7344840B2 (en) 2008-03-18
US6573043B1 (en) 2003-06-03
US20090155803A1 (en) 2009-06-18
US20060183150A1 (en) 2006-08-17
US6905830B2 (en) 2005-06-14
AU6409799A (en) 2000-04-26

Similar Documents

Publication Publication Date Title
US7674589B2 (en) Methods for tissue analysis
Press et al. Evaluation of HER-2/neu gene amplification and overexpression: comparison of frequently used assay methods in a molecularly characterized cohort of breast cancer specimens
Jimenez et al. Determination of Her-2/Neu status in breast carcinoma: comparative analysis of immunohistochemistry and fluorescent in situ hybridization
US7326575B2 (en) Methods and compositions for the preparation and use of fixed-treated cell-lines and tissue in fluorescence in situ hybridization
Hicks et al. Assessment of the HER2 status in breast cancer by fluorescence in situ hybridization: a technical review with interpretive guidelines
US6358682B1 (en) Method and kit for the prognostication of breast cancer
US10302645B2 (en) Materials and methods for diagnosis, prognosis and assessment of therapeutic/prophylactic treatment of prostate cancer
US6942970B2 (en) Identifying subjects suitable for topoisomerase II inhibitor treatment
US8062897B2 (en) Diagnostic histopathology using multiplex gene expression FISH
US20030017491A1 (en) Chromogenic in situ hybridization methods, kits, and compositions
Bozzetti et al. HER-2/neu amplification detected by fluorescence in situ hybridization in fine needle aspirates from primary breast cancer
US20150167102A1 (en) Diagnostic methods for determining prognosis of non-small cell lung cancer
Rummukainen et al. Amplification of c-myc oncogene by chromogenic and fluorescence in situ hybridization in archival breast cancer tissue array samples
CA2651419A1 (en) Diagnostic methods for determining treatment
Powell et al. Metallographic in situ hybridization
JP6626097B2 (en) Significance of intratumoral HER2 heterogeneity in breast cancer and uses therefor
McManus et al. Fluorescence in situ hybridisation detection of erbB2 amplification in breast cancer fine needle aspirates.
US20100297618A1 (en) Methods for determining a prognosis of colorectal cancer
Kang et al. Comparison of silver-enhanced in situ hybridization and fluorescence in situ hybridization for HER2 gene status in breast carcinomas

Legal Events

Date Code Title Description
ENP Entry into the national phase

Ref country code: AU

Ref document number: 1999 64097

Kind code of ref document: A

Format of ref document f/p: F

AK Designated states

Kind code of ref document: A1

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK DM EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase