USRE35491E - Methods and compositions for detecting human tumors - Google Patents

Methods and compositions for detecting human tumors Download PDF

Info

Publication number
USRE35491E
USRE35491E US07/421,096 US42109689A USRE35491E US RE35491 E USRE35491 E US RE35491E US 42109689 A US42109689 A US 42109689A US RE35491 E USRE35491 E US RE35491E
Authority
US
United States
Prior art keywords
glu
arg
lys
pro
gly
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Lifetime
Application number
US07/421,096
Inventor
Martin J. Cline
Dennis J. Slamon
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of California
Original Assignee
University of California
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
Priority claimed from US06/673,469 external-priority patent/US4699877A/en
Application filed by University of California filed Critical University of California
Priority to US07/421,096 priority Critical patent/USRE35491E/en
Application granted granted Critical
Publication of USRE35491E publication Critical patent/USRE35491E/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/82Translation products from oncogenes
    • 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
    • G01N33/57426Specifically defined cancers leukemia
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/82Translation products from oncogenes

Definitions

  • the src gene is found to be not only active in the malignant cell of chickens, but also in the normal cell. The difference appears to be one of degree, rather than of kind, in that the enzyme expressed by the src gene would appear to be of much higher concentration in the malignant cell as compared to the normal cell.
  • the observed property which is to be diagnostic of the tumor cell must be capable of differentiation from a normal cell or from a physiologic fluid of a normal host, where the fluid rather than cells are assayed. Furthermore, the property should not be specific for the individual, but be common to the malignant nature of the cell.
  • the opportunity or specifically detecting malignant cells is very important. Any technique, in a high percentage of situations where malignancy is suspected, should be able to distinguish malignant cells from normal cells. Furthermore, the diagnostic technique should be useful for a large number of members of the population and not specific for one or a few members of the population.
  • malignancy is a result of a natural process, which in a certain context results in malignancy.
  • malignancy may be based on normal processes, which at the time in question have an abberrant result, it is not surprising that there has been substantial difficulty in demonstrating observable differences between normal cells and cancer cells over a broad spectrum of allogeneic hosts.
  • Methods and compositions are provided for identifying and treating malignant cells of fresh tumors in a human host. From knowledge of DNA sequences capable of transforming cells of a lower vertebrate to malignancy, polynucleotide probes can be made for determining the level of transcription of said DNA in human cells and receptors produced capable of specifically recognizing determinant sites of peptide products of said DNA sequence.
  • the probes and receptors may be labeled with a wide variety of labels for diagnosis and treatment.
  • novel methods and compositions are provided for the diagnosis and treatment of cancer in humans and other primates. It has now been observed that DNA which is capable of transforming cells of lower vertebrates to malignancy is present in human cells and has a much higher level of transcription and expression in malignant cells than in normal cells. Thus, by being able to detect the higher level of messenger RNA or the expression product of such messenger RNA, the presence of malignant cells in a host may be diagnosed. In addition, the production of the higher level of peptides in the malignant cells can be a basis for treatment of the malignancy. Where the polypeptide expression product can be found in physiological fluids, such as blood, and the levels of the expression product are substantially different in the presence and absence of malignancy, the physiological fluid may be screened as diagnostic for the presence of a particular tumor.
  • the subject invention provides methods and compositions for evaluating the probability or presence of malignant cells in a group of cells, particularly human cells in vivo or freshly removed from a human host.
  • the method looks to cellular products such as mRNA or its expression product as diagnostic of the probable presence of malignant cells.
  • the mRNA which is selected from detection will usually be selected as a result of there being RNA present in a retrovirus genome, which retrovirus is capable of transforming mammalian cells to malignancy.
  • the RNA in the retrovirus which is selected to a sequence which does not encode an essential function of the retrovirus and, in fact, may be silent.
  • the method involves as a first step defining a DNA sequence capable of causing malignancy in a mammalian cell.
  • polynucleotide sequences can be provided which may serve as probes for detection of elevated levels of messenger RNA to determine whether a cell is malignant.
  • the sequence can also be used for defining polypeptide sequences which can define complementary receptors having high specificity for the peptide sequence.
  • the receptors may then be used for determining the presence or the concentration of the peptide in cells or physiological fluids and for treatment where the receptors can be directed to malignant cells. Also, knowing the nature of the peptide and its function, other means may be available for controlling the elevated production of the particular peptide.
  • the first step in the subject method is to define the DNA sequence.
  • Various methods can be used for defining the DNA sequence of a retroviral oncogene.
  • retroviruses have been found capable of transforming lower vertebrate cells to malignancy.
  • the retroviruses which have been characterized have been shown to carry DNA sequences comparable to wild type genes present in the host, genes which are now referred to as oncogenes.
  • oncogenes genes which are now referred to as oncogenes.
  • Rous sarcoma virus the expression product of the gene has been isolated and characterized and shown to be a kinase.
  • kinase In the case of this kinase, it has also been shown that the kinase is normally produced by the cell, but at a much lower level than when the src gene from the Rous sarcoma virus is introduced.
  • a number of viral oncogenes have already been detected in a variety of vertebrates, and the following is a list of the oncogenes and their species of origin.
  • Other ways will also become available in time for detecting and defining DNA sequences capable of transforming normal cells.
  • a further analysis can be employed by screening cDNA from fetuses with messenger RNA from malignant cells.
  • the oncogene is a gene which is silent or relatively quiescent in the mature vertebrate, while highly active in the embryo, the screening may further serve to narrow the field of sequences to be screened.
  • a cloned viral oncogene or short polynucleotide sequences can be employed as probes for detection of the level of production of messenger RNA in cells suspected of being malignant.
  • the preparation of both RNA and DNA nucleotide sequences, the labeling of the sequences, and the preferred size of the sequences has received ample description and exemplification in the literature.
  • a sequence should have at least about 14 nucleotides, usually at least about 18 nucleotides, and the polynucleotide probes may be one or more kilobases.
  • Various labels may be employed, most commonly radionuclides, particularly 32 P.
  • biotin modified nucleotides for introduction into a polynucleotide.
  • the biotin then serves as the site of binding to avidin or antibodies, which may be labeled with a wide variety of labels, such as radionuclides, fluorescers, enzymes, or the like.
  • antibodies may be employed which can recognize specific duplexes, including DNA duplexes, RNA duplexes and DNA-RNA hybrid duplexes or DNA-protein duplexes.
  • the antibodies in turn may be labeled and the assay may be carried out where the duplex is bound to a surface, so that upon the formation of duplex on the surface, the presence of antibody bound to the duplex can be detected.
  • the sequence of bases may be determined by known means, e.g., Maxam and Gilbert, PNAS USA (1977) 74:560.
  • the sequence can be used for the determination of the amino acid sequence of the protein expressed by the oncogene.
  • codons for methionine followed by a sequence which does not have stop codons which prevent expression, one can usually find a single sequence in frame with a methionine codon for defining the oncogene.
  • hybrid DNA technology may be employed for obtaining expression.
  • the DNA sequence may be restriction mapped and appropriate sites for cleavage defined. In this way, the sequence may be excised and introduced into a vector having the appropriate regulatory signals.
  • antibodies can be made to the polypeptide.
  • oocytes for expression of the messenger RNA which is then translated to produce the peptide expressed by the oncogene, the protein defined by the messenger may be produced. The identity of the peptide from the oocyte which the peptide produced by the expression of the hybrid DNA may then be determined.
  • Antibodies can be prepared in a variety of ways, depending upon whether monoclonal or polyclonal antibodies are desired.
  • polyclonal antibodies a vertebrate, normally a domestic animal, is hyperimmunized with the antigen and blood collected shortly after repeat immunizations and the gamma globulin isolated.
  • monoclonal antibodies a small animal is hyperimmunized, the spleen removed and the lymphocytes fused with an appropriate fusing partner. The resulting hybridomas are then grown under limiting dilution and clones providing the desired antibodies selected.
  • oligopeptides of at least about eight amino acids, usually at least about 10 and not more than 20, usually not more than 18 amino acids, to define a hapten which can be used to induce antibody formation.
  • the oligopeptide is bound to an appropriate immunogen and introduced into a vertebrate to produce antibodies, either polyclonal or monoclonal antibodies, as described previously.
  • the present invention also provides a series of oligopeptides corresponding to antigenic regions in the peptide expression products of RNA present in retrovirus oncogenes.
  • exemplary species of the antigenic oligopeptides useful in accordance with the subject invention are listed below in groups based on the retroviral oncogene (expression product) which is recognized by antibodies produced from the oligopeptide.
  • the above described techniques may be used for isolating the gene, mRNA or pseudo-gene and obtaining antibodies to the human expression product.
  • the human oncogene would be expected to have substantial complementarity to the related, v-onc, normally differing in fewer than about 5% of the bases, generally differing by fewer than 5% of the amino acids in the expression product.
  • the antibodies may be used in a variety of ways. Particularly, they may be used for diagnosis. In instances where the antigen may be found in a physiological fluid at an elevated concentration only when malignancy exists, the physiological fluid, such as serum, plasma, whole blood or cerebrospinal fluid may be assayed. Antibodies employed in assays may be labeled or unlabeled. Unlabeled antibodies may be employed in agglutination; labeled antibodies may be employed in a wide variety of assays, employing a wide variety of labels, such as radionuclides, enzymes, fluorescers, enzyme substrates or cofactors, or the like. These techniques are amply defined in the literature and exemplary assays may be found in U.S. Pat. Nos. 3,817,834, 3,935,074, 4,233,402 and 4,318,980, as illustrative.
  • kits which have the combination of the labeled antigen or labeled fragment and the antibody in amounts which provide for optimum sensitivity and accuracy.
  • a polyepitopic antigen can serve as a bridge between antibody bound to a support and labeled antibody in the assay medium.
  • the tissue sample may be lysed by conventional methods, e.g. base, detergents, or the like, cellular debris separated by filtration or centrifugation and the filtrate or supernatant isolated and assayed.
  • xenogeneic or allogeneic antibodies may be employed, depending upon the nature of the treatment, and whether the foreign antibodies will induce an immune response.
  • the literature has described a number of ways of making human antibodies, where it is found that mouse or other mammalian antibodies are not satisfactory.
  • the antibodies may be used in a wide variety of ways. By employing the appropriate IgG (other than IgG 1 ), one may induce lysis through the natural complement process. Alternatively, the lysing portion of a toxin may be joined to the antibodies, particularly a Fab fragment.
  • the antibodies may be bound to liposomes for directing the liposomes to the malignant cells to become ingested by the cells by merging of the membranes.
  • labels may also be bound to the antibodies, such as radionuclides, fluorescers, enzymes, and the like.
  • the antibodies By introducing the antibodies in vivo, the antibodies will direct the label to the malignant cells, where the presence of malignancy may be diagnosed or treated.
  • the formation of the antibodies will vary widely, depending on the nature of the label, the purpose of the antibodies, the site to which the antibodies are to be directed, and the like.
  • the antibodies will be formulated in a physiologically acceptable carrier, e.g. saline or phosphate buffered saline, and injected into the host, when possible at the desired site, and when this is not possible, into a circulating system, such as blood.
  • the antibodies obtained in accordance with this invention can also be used to isolate cells expressing the oncogene and to remove cells in vitro from a heterogeneous cell population containing cells expressing the oncogene. Separation can be achieved with a fluorescence activated cell sorter (FACS). This same technique can be used for identifying and isolating cells expressing the oncogene.
  • FACS fluorescence activated cell sorter
  • the subject antibodies may be combined with complement, joined to the lysing fragment (A fragment) of a toxin (see E.P.O. application no. 17,507 and U.K. Patent Application No. 2,034,324) or the cells agglutinated and separated by physical means.
  • Tumors were obtained from fresh surgical specimens at the time of resection and were untreated by chemotherapy or radiotherapy. An effort was made to obtain only viable tumor and to process the tissues as rapidly as possible to avoid messenger RNA (mRNA) degradation. Specimens were quickly frozen and stored in liquid nitrogen until processed for RNA. When the surgical specimens included wide margins of normal tissue, some of this was taken for analysis as an internal control of the level of c-onc gene expression. C-onc gene expression could then be compared in normal and malignant tissue from the same patient.
  • mRNA messenger RNA
  • RNA transcript of 3 kilobases (kb) per cell or approximately 2 micrograms of poly A RNA applied to the filter could be detected by this method (Kafatos et al., Nucleic Acids Res. (1979)7:1541).
  • appropriate controls including unrelated RNA's, poly A-negative fraction RNA, plasmid DNA, and mouse and human DNA, false-negative as well as false-positive results could reasonably be excluded.
  • the dot blots were quantitatively evaluated by means of a soft laser scanning densitometer. Where sufficient material was available, mRNA was further characterized by Northern analysis to confirm the presence of, and to size, specific transcripts (Thomas, PNAS USA (1980)77:5201).
  • RNA sequences homologous to c-erb, c-yes, c-abl, c-mos, c-fms, or c-sis could be detected. This was not the result of lack of homology between the viral gene probe and the human messenger RNA, since it was possible to detect homologues of all these probes in human genomic DNA. RNA from microscopically normal tissue did not contain any detectable transcripts by this analysis.
  • RNA sequences related to c-fes were detected in only 2 of 14 tumors examined; both of these were lung cancers.
  • C-myb expression was detected in only one of 14 tumors; this, too, was a lung cancer.
  • C-src messenger RNA sequences were observed only in circulating tumor cells of a patient with lymphosarcoma.
  • RNA from cells in the areas of the tumor sample and the control sample shows a correlation between the presence or absence of tumor and c-onc gene expression.
  • an adenocarcinoma of the small bowel, c-onc-related sequences were found in histologically normal adjacent tissue.
  • RNA from tumors and control tissues were performed by the Northern technique. Two c-myc-related transcripts of 4.0 and 2.0 kb were found in all tumors examined. In addition to these transcripts, there was obvious degradation of some of the messenger RNA in these hybridization analyses, most likely resulting from degradation occurring during tissue anoxia in the period after surgical removal of the tissue.
  • RNA samples were isolated by affinity chromatography on oligo(dT)-cellulose columns and spotted on nitrocellulose paper (dot blots) (Kafatos et al., Nucl. Acids Res. (1979) 7:1541-1552). The samples were hybridized to [ 32 P]-labeled molecularly cloned oncogene-specific probes. Dot blots were quantitatively evaluated by means of a soft laser scanning densitometer. Transcriptional activity of c-onc's was additionally studied in more detail in various tissues of newborn and 10 day old mice.
  • Poly(A + )-RNA was dissolved in water, boiled, quick-cooled on ice and 3 ⁇ g-(1.5 ⁇ l) were applied to sheets of nitrocellulose paper which had previously been equilibrated with 20 ⁇ SSC (1 ⁇ SSC is 0.15 NaCl, 0.015M sodium citrate) and air dried. After baking overnight at 80° C., the blots were prehybridized for at least 4 h at 45° C.
  • the blots were hybridized for about 20 h at 45° C. with 1 ⁇ 10 6 cpm of nick-translated probe/ml of hybridization buffer (prehybridization buffer with Denhardt's reagent at 1 ⁇ ).
  • the cloned oncogene fragments purified from vector sequences by preparative agarose gel electrophoresis were nick-translated (Rigby et al., J. Mol. Biol. (1977) 113.237-251) in the presence of [ 32 P]-dCTP(3200 Ci/mmol) to specific radioactivities of about 1-2 ⁇ 10 9 cpm/ ⁇ g of DNA.
  • the blots were washed three times in 1 ⁇ SSC at 50° C. for a total of about 2 h and exposed to preflashed X-ray films with intensifying screens at -70° C. for 72 h.
  • Hybridization showed that for c-abl about three-fold higher levels in the embryo proper than in extraembryonal membranes and placenta is observed at the 10th day of gestation, as compared to the concentration observed in the 6, 7, and 9 day conceptuses.
  • Expression of c-abl in the fetus appears to decrease after the 11th day of prenatal development.
  • the oncogene c-abl is transcriptionally active in all postnatal mouse tissue examined with spleen and thymus poly(A) + RNA exhibiting a slightly stronger hybridization than from other tissues.
  • the oncogene c-ras Ha was found to be expressed in considerable, but similar levels at all stages of prenatal development both in the embryo proper as well as in extraembryonal tissues. High levels of c-ras Ha expression were also observed in various tissues of newborn or 10 day old mice, particularly in bone, brain, kidney, skin and spleen.
  • the oncogene c-myc was detectable at days 7 and 8, but much higher levels were observed in late embryonic development (days 17 and 18).
  • the oncogene c-erb had maximum hybridization at 13 days, while no hybridization was observed at day 6.
  • the oncogene c-src was detected at its highest levels in the latter half of mouse embryonic development with an increase beginning at day 14, peaking at day 15 and gradually decreasing thereafter.
  • peak expressions were observed at days 7 and 16, the day-7 peak was 1.5 to 3 times higher than all other days and the day-16 peak was 1.5 to 2 times higher than days 9 to 13 days 17 and 18.
  • the polypeptides were linked to keyhole limpet hemocyanin in accordance with conventional techniques (Dockray, Regulatory Peptides (1980)1:169) and the resulting immunogen was used to immunize rabbits in a first injection with complete Freund's adjuvant, followed by injections with incomplete Freund's adjuvant over periods of three to four weeks to hyperimmunize the rabbits. The rabbits were bled repeatedly over a period of six months. Of the seven oligopeptides which resulted in the production of antibodies, antibodies to two peptides (5 and 7) were selected for detailed analysis.
  • Antibody No. 5 identifies a specific protein of approximately 58,000 daltons, which is present in the virus-infected cell line but not in controls.
  • Antibody against polypeptide 5 was also reacted with the plasma of chickens bearing tumors induced by amv. A band similar to that observed with the above lysates of approximately 48,000 daltons was identified.
  • Antisera to polypeptide No. 5 was also reacted with lysates of a myeloid human leukemia cell line (HL-60) which is known to express messenger RNA transcripts of the c-myb gene (Gallo and Wong-Staal, Blood (1982) 60:545). This antibody reacted with a protein of about 90,000 daltons. In freshly isolated myeloid leukemia cells, the antibody identifies a series of proteins, 14 kd to 70 kd, not present in normal white blood cells.
  • HL-60 myeloid human leukemia cell line
  • the polyconal antibodies to the fragment no. 5 of the myb protein was tested for its ability to kill normal and leukemic cells.
  • the procedure employed is described in Terasaki and McClelland, infra. The data are set forth in the following table.
  • the expression product of the myb gene can react with antibodies to produce lysis with complement.
  • the myb protein appears to be a surface membrane protein which is available for binding to antibodies.
  • proteins to which specific antibodies will bind which proteins have diagnostic value as indicative of malignance
  • the malignant cells can be identified and treated.
  • the subject antibodies can be used to select for antibodies binding to the same or other determinant site.
  • the subject data demonstrate that antibodies can be prepared which do not affect normal B- and T-cells, but are cytotoxic in combination with complement for a variety of malignant cells. Therefore, the antibodies can be used for cancer therapy without the hazard of substantially inactivating the immune system.
  • Antibodies can be produced to the peptides, which antibodies may be labeled and may then be used for diagnosing the presence of a peptide diagnostic of malignancy.
  • the oncogernic proteins are found to be available for binding to antibodies as surface membrane proteins.
  • the antibodies may serve as diagnostic reagents for determining the presence of malignancy and determining the location of malignant cells.
  • the antibodies may also serve in treating tumors in vivo by using radionuclides, toxins, in combination with the host complement system or opsonins, or other antibody dependent lytic system or the like.
  • the antibodies find use in pre- and postoperative systems, in the later determining whether complete removal has occurred, whether metastases exist.
  • the antibodies can be used postoperatively to destroy any remnants of the tumor which may not have been excised.

Abstract

Methods and compositions for detecting the presence of tumors are provided, where a physiological sample is assayed for the expression product of a c-onc gene as diagnostic for the presence of the tumor. The method finds use in both pre-and postoperative situations with a host suspected of having transformed malignant cells.

Description

RELATED APPLICATIONS
This application is a continuation-in-part application of copending application Ser. No. 439,252, filed Nov. 4, 1982, .Iadd.now abandoned, .Iaddend.and copending application Ser. No. 496,027 filed May 19, 1983, now abandoned, incorporating herein by reference.
BACKGROUND OF THE INVENTION
1. Field of the Invention
The mechanism for malignancy of mammalian cells has been and continues to be the subject matter of intense investigation. One of the areas which is considered to be promising in the elucidation of the mechanism is the area of oncogenes. While the occurrence of oncogenes was first detected with retroviruses, it now seems reasonably firm that the viral oncogenes have cellular counterparts. The role of the cellular counterparts is not clear. An excellent review of oncogenes, their properties and particularly the src gene may be found in the article by J. Michael Bishop, Scientific American, Mar., 1982:81-93. The article also provides a list of various viral oncogenes, demonstrating that a number of them are involved with phosphorylation.
The src gene is found to be not only active in the malignant cell of chickens, but also in the normal cell. The difference appears to be one of degree, rather than of kind, in that the enzyme expressed by the src gene would appear to be of much higher concentration in the malignant cell as compared to the normal cell.
In order to be able to determine the presence of a tumor cell, it is necessary to be able to distinguish between normal cells and tumor cells. Therefore, the observed property which is to be diagnostic of the tumor cell must be capable of differentiation from a normal cell or from a physiologic fluid of a normal host, where the fluid rather than cells are assayed. Furthermore, the property should not be specific for the individual, but be common to the malignant nature of the cell.
In both diagnosis and treatment, the opportunity or specifically detecting malignant cells is very important. Any technique, in a high percentage of situations where malignancy is suspected, should be able to distinguish malignant cells from normal cells. Furthermore, the diagnostic technique should be useful for a large number of members of the population and not specific for one or a few members of the population.
Because a cancer cell is derived from a normal cell, most of the properties and components of the malignant cell are the same as the normal cell. Furthermore, there is an increasing view that malignancy is a result of a natural process, which in a certain context results in malignancy. In view of the fact that malignancy may be based on normal processes, which at the time in question have an abberrant result, it is not surprising that there has been substantial difficulty in demonstrating observable differences between normal cells and cancer cells over a broad spectrum of allogeneic hosts.
2. Description of the Prior Art
The following papers provide a general description of oncogenes and the role of retroviruses in tumorigenesis: Bishop, Scientific American, supra; Bishop, New England J. of Med. (1980) 303:675-681; Lancet, Jul. 24, 1982, pages 195-196; Cooper, Science (1982) 218:801-806; Vamus, Science (1982) 216:821-820. Papers concerned with specific oncogenes include Becker et al., PNAS USA (1982) 79:3315-3319; Tsuchida et al., Science (1982) 217:937-938 and Dhar et al., ibid., (1982) 217:934-936.
SUMMARY OF THE INVENTION
Methods and compositions are provided for identifying and treating malignant cells of fresh tumors in a human host. From knowledge of DNA sequences capable of transforming cells of a lower vertebrate to malignancy, polynucleotide probes can be made for determining the level of transcription of said DNA in human cells and receptors produced capable of specifically recognizing determinant sites of peptide products of said DNA sequence. The probes and receptors may be labeled with a wide variety of labels for diagnosis and treatment.
DESCRIPTION OF THE SPECIFIC EMBODIMENTS
In accordance with the subject invention, novel methods and compositions are provided for the diagnosis and treatment of cancer in humans and other primates. It has now been observed that DNA which is capable of transforming cells of lower vertebrates to malignancy is present in human cells and has a much higher level of transcription and expression in malignant cells than in normal cells. Thus, by being able to detect the higher level of messenger RNA or the expression product of such messenger RNA, the presence of malignant cells in a host may be diagnosed. In addition, the production of the higher level of peptides in the malignant cells can be a basis for treatment of the malignancy. Where the polypeptide expression product can be found in physiological fluids, such as blood, and the levels of the expression product are substantially different in the presence and absence of malignancy, the physiological fluid may be screened as diagnostic for the presence of a particular tumor.
The subject invention provides methods and compositions for evaluating the probability or presence of malignant cells in a group of cells, particularly human cells in vivo or freshly removed from a human host. The method looks to cellular products such as mRNA or its expression product as diagnostic of the probable presence of malignant cells. The mRNA which is selected from detection will usually be selected as a result of there being RNA present in a retrovirus genome, which retrovirus is capable of transforming mammalian cells to malignancy. Furthermore, the RNA in the retrovirus which is selected to a sequence which does not encode an essential function of the retrovirus and, in fact, may be silent.
The method involves as a first step defining a DNA sequence capable of causing malignancy in a mammalian cell. Once the DNA sequence is defined, polynucleotide sequences can be provided which may serve as probes for detection of elevated levels of messenger RNA to determine whether a cell is malignant. The sequence can also be used for defining polypeptide sequences which can define complementary receptors having high specificity for the peptide sequence. The receptors may then be used for determining the presence or the concentration of the peptide in cells or physiological fluids and for treatment where the receptors can be directed to malignant cells. Also, knowing the nature of the peptide and its function, other means may be available for controlling the elevated production of the particular peptide.
The first step in the subject method is to define the DNA sequence. Various methods can be used for defining the DNA sequence of a retroviral oncogene. For example, retroviruses have been found capable of transforming lower vertebrate cells to malignancy. The retroviruses which have been characterized have been shown to carry DNA sequences comparable to wild type genes present in the host, genes which are now referred to as oncogenes. Furthermore, in the case of Rous sarcoma virus, the expression product of the gene has been isolated and characterized and shown to be a kinase. In the case of this kinase, it has also been shown that the kinase is normally produced by the cell, but at a much lower level than when the src gene from the Rous sarcoma virus is introduced. A number of viral oncogenes have already been detected in a variety of vertebrates, and the following is a list of the oncogenes and their species of origin.
              TABLE 1                                                     
______________________________________                                    
                     Species of                                           
Oncogene             origin                                               
______________________________________                                    
v-src                chicken                                              
v-fps                "                                                    
v-yes                "                                                    
v-fos                "                                                    
v-myc                "                                                    
v-erb                "                                                    
v-myb                "                                                    
v-rel                turkey                                               
v-mos                mouse                                                
v-bas                "                                                    
v-abl                "                                                    
v-ras                rat                                                  
v-fes                cat                                                  
v-fms                "                                                    
v-sis                monkey                                               
______________________________________                                    
Other sources of DNA sequences capable of including malignant transformations in vertebrate cells may include isolated DNA from a malignant cell or cell line, cloned DNA from a genomic library or cloned DNA from a messenger RNA library, where the total messenger of the malignant cell is reverse transcribed to DNA and cloned. Either of these libraries may be screened for their ability to induce malignancy. A refinement in the technique of screening may be achieved by taking the total messenger from a normal cell and preparing cDNA from the messenger. One can then use the single stranded DNA as a probe to remove messenger RNA associated with the normal cell from the total messenger RNA from a malignant cell. The residual messenger RNA will then include messenger being expressed by genes associated with the malignancy. One may then use the messenger to screen a genomic library and use the cloned DNA which hybridizes with messenger in a bioassay for the determination of the ability to transform to malignancy. Other ways will also become available in time for detecting and defining DNA sequences capable of transforming normal cells.
A further analysis can be employed by screening cDNA from fetuses with messenger RNA from malignant cells. Particularly, where the oncogene is a gene which is silent or relatively quiescent in the mature vertebrate, while highly active in the embryo, the screening may further serve to narrow the field of sequences to be screened.
Once haVing identified a DNA sequence capable of inducing malignancy, a cloned viral oncogene or short polynucleotide sequences can be employed as probes for detection of the level of production of messenger RNA in cells suspected of being malignant. The preparation of both RNA and DNA nucleotide sequences, the labeling of the sequences, and the preferred size of the sequences has received ample description and exemplification in the literature. Normally, a sequence should have at least about 14 nucleotides, usually at least about 18 nucleotides, and the polynucleotide probes may be one or more kilobases. Various labels may be employed, most commonly radionuclides, particularly 32 P. However, other techniques may also be employed, such as using biotin modified nucleotides for introduction into a polynucleotide. The biotin then serves as the site of binding to avidin or antibodies, which may be labeled with a wide variety of labels, such as radionuclides, fluorescers, enzymes, or the like. Alternatively, antibodies may be employed which can recognize specific duplexes, including DNA duplexes, RNA duplexes and DNA-RNA hybrid duplexes or DNA-protein duplexes. The antibodies in turn may be labeled and the assay may be carried out where the duplex is bound to a surface, so that upon the formation of duplex on the surface, the presence of antibody bound to the duplex can be detected.
By isolating the nucleotide sequence for the whole oncogene, the sequence of bases may be determined by known means, e.g., Maxam and Gilbert, PNAS USA (1977) 74:560. The sequence can be used for the determination of the amino acid sequence of the protein expressed by the oncogene. By identifying codons for methionine followed by a sequence which does not have stop codons which prevent expression, one can usually find a single sequence in frame with a methionine codon for defining the oncogene.
Alternatively, hybrid DNA technology may be employed for obtaining expression. The DNA sequence may be restriction mapped and appropriate sites for cleavage defined. In this way, the sequence may be excised and introduced into a vector having the appropriate regulatory signals. After obtaining expression of the DNA sequence, antibodies can be made to the polypeptide. By employing oocytes for expression of the messenger RNA which is then translated to produce the peptide expressed by the oncogene, the protein defined by the messenger may be produced. The identity of the peptide from the oocyte which the peptide produced by the expression of the hybrid DNA may then be determined.
Once the protein has identified and verified, one can then use the protein or subunit peptides as an antigen for the production of antibodies for diagnosis and treatment. Antibodies can be prepared in a variety of ways, depending upon whether monoclonal or polyclonal antibodies are desired. For polyclonal antibodies, a vertebrate, normally a domestic animal, is hyperimmunized with the antigen and blood collected shortly after repeat immunizations and the gamma globulin isolated. For monoclonal antibodies, a small animal is hyperimmunized, the spleen removed and the lymphocytes fused with an appropriate fusing partner. The resulting hybridomas are then grown under limiting dilution and clones providing the desired antibodies selected.
Rather than preparing the entire peptide, one can determine various regions which are likely to be determinant sites and use these oligopeptides of at least about eight amino acids, usually at least about 10 and not more than 20, usually not more than 18 amino acids, to define a hapten which can be used to induce antibody formation. The oligopeptide is bound to an appropriate immunogen and introduced into a vertebrate to produce antibodies, either polyclonal or monoclonal antibodies, as described previously.
Accordingly, the present invention also provides a series of oligopeptides corresponding to antigenic regions in the peptide expression products of RNA present in retrovirus oncogenes. Exemplary species of the antigenic oligopeptides useful in accordance with the subject invention are listed below in groups based on the retroviral oncogene (expression product) which is recognized by antibodies produced from the oligopeptide.
______________________________________                                    
A. Myb                                                                    
met-ala-phe-ala-his-asn-pro-pro-ala-gly-pro-leu-pro-                      
 gly-ala                                                                  
pro-phe-his-lys-asp-gln-thr-phe-thr-glu-tyr-arg-lys-met-                  
 his-gly-gly-ala-val                                                      
pro-phe-his-lys-asp-gln-thr-phe-thr-glu-tyr-arg-lys-met-                  
asp-asn-thr-arg-thr-ser-gly-asp-asn-ala-pro-val-ser-cys-                  
 leu-gly-glu                                                              
B. Src                                                                    
arg-leu-ileu-glu-asp-asn-glu-tyr-thr-ala-arg-gln-gly-ala-                 
 lys-phe-pro                                                              
trp-arg-arg-asp-pro-glu-glu-arg-pro-thr                                   
C. Ras.sup.Ki                                                             
arg-leu-lys-lys-ileu-ser-lys-glu-glu-lys-thr-pro-gly-cys-                 
 val-lys-ileu-lys-lys                                                     
asp-leu-pro-ser-arg-thr-val-asp-thr-asp-thr-lys-gln-ala-gln-glu-          
 leu-ala-arg                                                              
met-thr-glu-tyr-lys-leu-val-val-val-gly-ala-ser-gly-val-                  
 gly-lys-ser-ala                                                          
D. Ras.sup.Ha                                                             
glu-asp-ileu-his-gln-tyr-arg-glu-gln-ileu-lys-arg-val-lys-                
 asp-ser-asp-asp                                                          
val-arg-glu-ileu-arg-gln-his-lys-ser-arg-lys-leu-asn-pro-                 
 pro-asp-glu-ser-gly-pro                                                  
met-thr-glu-tyr-lys-leu-val-val-val-gly-ala-gly-gly-val-                  
 gly-lys-ser-ala                                                          
val-asp-glu-tyr-asp-pro-thr-ileu-glu-asp-ser-tyr-arg-lys-                 
 gln-val                                                                  
E. Fes                                                                    
arg-his-ser-thr-ser-ser-ser-glu-gln-glu-arg-glu-gly-gly-                  
 arg                                                                      
asn-gln-gln-thr-arg-glu-phe-val-glu-lys-gly-gly-arg                       
pro-glu-val-gln-lys-pro-leu-his-glu-gln                                   
ala-ser-pro-tyr-pro-asn-leu-ser-asn-gln-gln-thr-arg                       
F. Myc                                                                    
arg-leu-ileu-ala-glu-lys-glu-gln-leu-arg-arg-arg-arg-glu-                 
 gln                                                                      
asn-asn-glu-lys-ala-pro-lys-val-val                                       
______________________________________                                    
In those situations where the human gene is different from the v-onc, e.g. human c-ras, the above described techniques may be used for isolating the gene, mRNA or pseudo-gene and obtaining antibodies to the human expression product. The human oncogene would be expected to have substantial complementarity to the related, v-onc, normally differing in fewer than about 5% of the bases, generally differing by fewer than 5% of the amino acids in the expression product.
The antibodies may be used in a variety of ways. Particularly, they may be used for diagnosis. In instances where the antigen may be found in a physiological fluid at an elevated concentration only when malignancy exists, the physiological fluid, such as serum, plasma, whole blood or cerebrospinal fluid may be assayed. Antibodies employed in assays may be labeled or unlabeled. Unlabeled antibodies may be employed in agglutination; labeled antibodies may be employed in a wide variety of assays, employing a wide variety of labels, such as radionuclides, enzymes, fluorescers, enzyme substrates or cofactors, or the like. These techniques are amply defined in the literature and exemplary assays may be found in U.S. Pat. Nos. 3,817,834, 3,935,074, 4,233,402 and 4,318,980, as illustrative.
In some techniques, it will be useful to label the antigen or fragment thereof, rather than the antibody and have a competition between labeled antigen and antigen in the sample for antibody. In this situation, it is common to provide kits which have the combination of the labeled antigen or labeled fragment and the antibody in amounts which provide for optimum sensitivity and accuracy.
In other situations, it is desirable to have a solid support, where either antigen or antibody is bound. A polyepitopic antigen can serve as a bridge between antibody bound to a support and labeled antibody in the assay medium. Alternatively, one may have a competition between labeled antigen and any antigen in the sample for a limited amount of antibody.
Where the antigen may not be found in a physiological fluid or if found there is not diagnostic of malignancy, then cells will have to be isolated and the cells assayed for the presence of messenger RNA or the antigen. Methods of detecting messenger RNA have already been described. For detecting the antigen, the tissue sample may be lysed by conventional methods, e.g. base, detergents, or the like, cellular debris separated by filtration or centrifugation and the filtrate or supernatant isolated and assayed.
For purposes of therapy, either xenogeneic or allogeneic antibodies may be employed, depending upon the nature of the treatment, and whether the foreign antibodies will induce an immune response. The literature has described a number of ways of making human antibodies, where it is found that mouse or other mammalian antibodies are not satisfactory. The antibodies may be used in a wide variety of ways. By employing the appropriate IgG (other than IgG1), one may induce lysis through the natural complement process. Alternatively, the lysing portion of a toxin may be joined to the antibodies, particularly a Fab fragment. The antibodies may be bound to liposomes for directing the liposomes to the malignant cells to become ingested by the cells by merging of the membranes. Other labels may also be bound to the antibodies, such as radionuclides, fluorescers, enzymes, and the like. By introducing the antibodies in vivo, the antibodies will direct the label to the malignant cells, where the presence of malignancy may be diagnosed or treated.
The formation of the antibodies will vary widely, depending on the nature of the label, the purpose of the antibodies, the site to which the antibodies are to be directed, and the like. Usually, the antibodies will be formulated in a physiologically acceptable carrier, e.g. saline or phosphate buffered saline, and injected into the host, when possible at the desired site, and when this is not possible, into a circulating system, such as blood.
The antibodies obtained in accordance with this invention can also be used to isolate cells expressing the oncogene and to remove cells in vitro from a heterogeneous cell population containing cells expressing the oncogene. Separation can be achieved with a fluorescence activated cell sorter (FACS). This same technique can be used for identifying and isolating cells expressing the oncogene. For removing cells expressing the oncogene from a mixture of cells, the subject antibodies may be combined with complement, joined to the lysing fragment (A fragment) of a toxin (see E.P.O. application no. 17,507 and U.K. Patent Application No. 2,034,324) or the cells agglutinated and separated by physical means.
The following examples are offered by way of illustration and not by way of limitation.
Tumors were obtained from fresh surgical specimens at the time of resection and were untreated by chemotherapy or radiotherapy. An effort was made to obtain only viable tumor and to process the tissues as rapidly as possible to avoid messenger RNA (mRNA) degradation. Specimens were quickly frozen and stored in liquid nitrogen until processed for RNA. When the surgical specimens included wide margins of normal tissue, some of this was taken for analysis as an internal control of the level of c-onc gene expression. C-onc gene expression could then be compared in normal and malignant tissue from the same patient. As little as 20 pg of Maloney murine sarcoma virus equivalent to approximately one RNA transcript of 3 kilobases (kb) per cell or approximately 2 micrograms of poly A RNA applied to the filter could be detected by this method (Kafatos et al., Nucleic Acids Res. (1979)7:1541). By use of appropriate controls including unrelated RNA's, poly A-negative fraction RNA, plasmid DNA, and mouse and human DNA, false-negative as well as false-positive results could reasonably be excluded. The dot blots were quantitatively evaluated by means of a soft laser scanning densitometer. Where sufficient material was available, mRNA was further characterized by Northern analysis to confirm the presence of, and to size, specific transcripts (Thomas, PNAS USA (1980)77:5201).
Expression of 13 cellular oncogenes in 14 tumors was examined by DNA-RNA hybridization techniques. These data are summarized in Table 2.
                                  TABLE 2                                 
__________________________________________________________________________
HUMAN MALIGNANCY*                                                         
GASTROINESTINAL                                                           
MALIGNANCIES                                       LUNG                   
Adenocarcinoma of                                  ADENO-                 
                                                         LYMPHO-          
             Small                                                        
                 Rec-                                                     
                     RENAL CELL         OVARAIN    CARCI-                 
                                                         SAR-             
V-ONC                                                                     
     Colon   Bowel                                                        
                 tum CARCINOMA          CARCINOMA  NOMA  COMA             
PROBE                                                                     
     1   2   1   1   1    2    3   4    1  2  3    1  2  1                
__________________________________________________________________________
Myc**                                                                     
     +++ +++ +++ +++ ++++ ++++ ++  ++++ ++ ++ ++   ++ ++ -                
Myb  -   -   -   -   -    -    -   -    -  -  -    -  ++ -                
Erb  -   -   -   -   -    -    -   -    -  -  -    -  -  -                
Src  -   -   -   -   -    -    -   -    -  -  -    -  -  +++              
Yes  -   -   -   -   -    -    -   -    -  -  -    -  -  -                
Abi***                                                                    
     -   -   -   -   -    -    -   -    -  -  -    -  -  -                
Fos  +++ +++ +++ +++ ++   +++  +++ ++   +  ++ ++++ +  ++ -                
Mos  -   -   -   -   -    -    -   -    -  -  -    -  -  -                
Ras.sup.Ha                                                                
     ++  ++  ++  ++  ++   ++   ++  ++   ++ ++ ++   ++ ++ -                
Ras.sup.Ki                                                                
     ++  ++  ++  ++  ++   ++   ++  ++   ++ ++ ++   ++ ++ -                
Fes  -   -   -   -   -    -    -   -    -  -  -    +  ++ -                
Fms  -   -   -   -   -    -    -   -    -  -  -    -  -  -                
Sis  -   -   -   -   -    -    -   -    -  -  -    -  -  -                
__________________________________________________________________________
 *Increasing numbers of pluses indicates increasing intensity of          
 hybridization of tissue mRNA to vonc probes.                             
 **Avian                                                                  
 ***Murine                                                                
Three patterns were observed: (1) expression of specific c-onc mRNA sequences in all or nearly all tumor samples (e.g., c-myc); (2) detection of c-onc expression in sporadic tumors(e.g., c-fes); and (3) no detectable expression (e.g., c-mos).
No significant expression of mRNA sequences homologous to c-erb, c-yes, c-abl, c-mos, c-fms, or c-sis could be detected. This was not the result of lack of homology between the viral gene probe and the human messenger RNA, since it was possible to detect homologues of all these probes in human genomic DNA. RNA from microscopically normal tissue did not contain any detectable transcripts by this analysis.
Four cellular oncogenes showed a consistent pattern of expression in a variety of human tumors. These were c-myc, c-fos, c-rasHa, and c-rasKi. A comparison was made of the intensity of hybridization, which was possible since all probes were labeled to approximately the same specific activity. V-myc and v-fos demonstrated the highest intensity of hybridization to human tumor RNA's suggesting a large number of copies of mRNA per cell. Expression of both these genes was observed in all malignancies examined. C-rasHa and c-rasKi sequences were also detected in most of the human tumors but with less intense hybridization.
Messengers RNA sequences related to c-fes were detected in only 2 of 14 tumors examined; both of these were lung cancers.
C-myb expression was detected in only one of 14 tumors; this, too, was a lung cancer.
C-src messenger RNA sequences were observed only in circulating tumor cells of a patient with lymphosarcoma.
In order to test whether expression of cellular oncogene sequences was related to neoplasia, an effort was made to obtain both grossly normal-appearing tissue and obviously malignant tissue from the same site in the same patient at the same time. Hybridization studies were then performed on RNA samples from the tumor and from adjacent noninvolved tissues. In 6 of the 14 patients it was possible to perform this analysis. In 1 of these 6 cases the presumed normal tissue was subsequently shown by histologic analysis to be infiltrated by tumor. In 4 of the remaining 5 cases there was differential expression between the tumor and normal tissue, with low or undetectable levels observed in the normal tissues and elevated levels observed in the malignancy. Three of the renal cell carcinomas and one colon carcinoma demonstrated this phenomenon.
Blot analysis of RNA from cells in the areas of the tumor sample and the control sample shows a correlation between the presence or absence of tumor and c-onc gene expression. In one tumor, an adenocarcinoma of the small bowel, c-onc-related sequences were found in histologically normal adjacent tissue.
Analyses of poly A RNA from tumors and control tissues were performed by the Northern technique. Two c-myc-related transcripts of 4.0 and 2.0 kb were found in all tumors examined. In addition to these transcripts, there was obvious degradation of some of the messenger RNA in these hybridization analyses, most likely resulting from degradation occurring during tissue anoxia in the period after surgical removal of the tissue.
Using the procedures given above, several other tumor types obtained from fresh surgical specimens were examined for c-onc gene expression. In this series of tests, DNA-RNA hybridization was used to look for expression of 10 different cellular oncogenes in 9 tumors. The data obtained are summarized in Table 3 below.
                                  TABLE 3                                 
__________________________________________________________________________
HUMAN MALIGNANCY*                                                         
                                                     CHRONIC              
      BREAST      UTERINE                 NON-HODGKIN'S                   
                                                     MYELOCYTIC           
V-ONC CARCINOMA   CARCINOMA                                               
                          THYMOMA HODGKIN'S                               
                                          LYMPHOMA   LEUKEMIA             
PROBE 1   2   3   1       1       1       1          1    2               
__________________________________________________________________________
Myc**                                                                     
     ++   +++ +++ -       +       ++      +          ++   -               
Myb  +    ++  -   -       -       +       -          +    -               
Src  +    +   -   -       -       ++      +          +++  -               
Rel  -    -   -   -       -       -       -          -    -               
Abi***                                                                    
     -    -   -   -       -       -       -          -    -               
Fos  ++   ++  ++  -       +       ++      +          ++++ ++++            
Ras.sup.Ha                                                                
     +++  +++ +++ -       +       ++      +          ++   +               
Ras.sup.Ki                                                                
     +    +   ++  -       -       +       -          +    +               
Fes  +    ++  +++ -       -       +       +          +++  ++              
Sis  -    -   -   -       -       -       -          -    -               
__________________________________________________________________________
 *Increasing numbers of pulses indicates increasing intensity of          
 hybridization of tissue mRNA to vonc probes.                             
 **Avian                                                                  
 ***Murine                                                                
The results given in Table 3 correlate fairly well with the results previously reported in Table 2 in that the cellular oncogenes c-myc, c-fos, c-rasHa, and c-rasKi show a consistent pattern of expression in the additional tumor types examined. Further, c-myb, c-src and c-fes were also detected in several additional tumor types whereas c-rel, c-abl and c-sis expression was not observed in any of the additional tumor types examined. Interestingly, none of the cellular oncogenes looked for were found to be expressed at any significant level in the single uterine carcinoma evaluated.
To determine whether the messenger RNA shown to be present in malignant cells in elevated amounts were related to genes involved in embryogenesis, experiments were carried out generally as follows. Total RNA was isolated from embryo/fetuses of random-bred Swiss mice at daily intervals starting on the 6th day of gestation (day of coital plug was taken as day 0 of prenatal development). Beginning at day 10 of prenatal development, the embryo proper was separated from the extraembryonal membranes and placenta. The small size prior to day 10 prevented separation and therefore the embryos of days 6-9 represent the entire conceptus as dissected from the uterine wall.
Aliquots of poly(A)-containing RNA (poly(A+) RNA), were isolated by affinity chromatography on oligo(dT)-cellulose columns and spotted on nitrocellulose paper (dot blots) (Kafatos et al., Nucl. Acids Res. (1979) 7:1541-1552). The samples were hybridized to [32 P]-labeled molecularly cloned oncogene-specific probes. Dot blots were quantitatively evaluated by means of a soft laser scanning densitometer. Transcriptional activity of c-onc's was additionally studied in more detail in various tissues of newborn and 10 day old mice. Agarose gel electrophoresis followed by blotting on nitrocellulose paper (Northern blotting), (Thomas, PNAS USA (1980) 77:5201-5205) was used to confirm the results obtained by dot blot analysis and additionally to determine the sizes of the different c-onc-related transcripts.
More specifically, RNA was isolated from Swiss-Webster mouse embryo fetuses at various stages of development using the guanidine thiocyanate method. (Cox, Methods Enzymol. (1967) 12:120-129; Adams et al., PNAS USA (1980) 74:3399-3043).
As indicated above, days 6-9 Swiss-Webster mouse embryos represent the entire conceptuses including all extraembryonal tissues, such as membranes and those cells giving rise to the placenta at later developmental stages. At all later stages, the embryo proper was dissected free of extraembryonal tissues. RNA was selected for poly(A+) -RNA by one cycle of chromatography on oligo(dT)-cellulose columns (Aviv and Leder, PNAS USA (1972) 69:1408-1412). Poly(A+)-RNA was dissolved in water, boiled, quick-cooled on ice and 3 μg-(1.5 μl) were applied to sheets of nitrocellulose paper which had previously been equilibrated with 20×SSC (1×SSC is 0.15 NaCl, 0.015M sodium citrate) and air dried. After baking overnight at 80° C., the blots were prehybridized for at least 4 h at 45° C. in a buffer containing 0.75M NaCl, 0.05M sodium phosphate (pH7.5), 0.005M EDTA, 0.2% SDS, 10 mg of glycine/ml, 5×Denhardt's reagent (1×Denhardt's reagent is 0.02% each of ficoll, bovine serum albumin and polyvinylpyrrolidone), 0.25 mg of denatured herring DNA/ml and 50% formamide.
The blots were hybridized for about 20 h at 45° C. with 1×106 cpm of nick-translated probe/ml of hybridization buffer (prehybridization buffer with Denhardt's reagent at 1×). The cloned oncogene fragments purified from vector sequences by preparative agarose gel electrophoresis were nick-translated (Rigby et al., J. Mol. Biol. (1977) 113.237-251) in the presence of [32 P]-dCTP(3200 Ci/mmol) to specific radioactivities of about 1-2×109 cpm/μg of DNA. After hybridization, the blots were washed three times in 1×SSC at 50° C. for a total of about 2 h and exposed to preflashed X-ray films with intensifying screens at -70° C. for 72 h.
Employing the above procedure, a number of known oncogenes were screened to determine whether they were expressed in the embryos. The following table indicates the individual oncogene and the observations concerning their expression in embryonic cells.
                                  TABLE 4                                 
__________________________________________________________________________
                           mRNA production                                
                           embryos                                        
                           days                                           
Oncogene                                                                  
      Virus      Disease   6-9                                            
                              10-18                                       
                                  fetus                                   
__________________________________________________________________________
fos.sup.1.                                                                
      FBJ-osterosarcoma                                                   
                 ostersarcoma                                             
                           +  -   +                                       
abl.sup.2.                                                                
      Abelson leukemia                                                    
                 lymphoma  +  +   +                                       
ras.sup.Ha3.                                                              
      Harvey sarcoma                                                      
                 erythroleukemia,                                         
                           +  +   +                                       
                 sarcoma                                                  
mos.sup.4.                                                                
      Maloney sarcoma                                                     
                 sarcoma   -  -   -                                       
myc.sup.5.                                                                
      Avian      carcinoma, sarcoma                                       
                           +  +                                           
      myelocytomatosis                                                    
                 leukemia                                                 
erb.sup.6.                                                                
      Avian      leukemia, sarcoma                                        
                           -  +                                           
      erythroblastosis                                                    
                 sarcoma                                                  
src.sup.7.                                                                
      Rous sarcoma virus                                                  
                 sarcoma   +  +                                           
myb.sup.8.                                                                
      Avian      leukemia  -  -                                           
      myeloblastosis                                                      
fes.sup.9.                                                                
      Synder-Theilin                                                      
                 sarcoma   -  -                                           
      feline sarcoma                                                      
sis.sup.10.                                                               
      Simian sarcoma                                                      
                 sarcoma   +  +                                           
__________________________________________________________________________
 .sup.1. Curran et al., J. Virol, (1982)                                  
 .sup.2. Goff et al., Cell (1980) 22:777785                               
 .sup.3. Ellis et al., J. Virol. (1980) 36:408420                         
 .sup.4. Oskarmon et al., Science (1980) 207:12221227; Jones et al., PNAS 
 USA (9180) 77:26512655                                                   
 .sup.5. Eva et al., Nature (1982) 295:116                                
 .sup.6. Gonda et al., Mol. Cell. Biol. (1982) 2:617                      
 .sup.7. Wang et al., (1977) J. Virol. 24:64                              
 .sup.8. Vister et al., PNAS USA (1982) 79:36773681                       
 .sup.9. Fedele et al., PNAS (1982), in press                             
 .sup.10. Devare et al., PNAS (1982) 79:31793182                          
Relatively high levels of c-fos related sequences were detected in poly(A+)-RNA prepared from 6, 7, 8 and 9 day conceptuses containing the embryo proper and extraembryonal tissues. More than 10-fold lower fos expression was observed in embryos of later development stages dissected free of extraembryonal tissues. Data from the placenta and extraembryonal membranes of fetuses from days 10 to 18, showed that expression was primarily in those tissues. In postnatal tissue, c-fos expression could be observed in all tissues investigated with stronger hybridization to the fos-specific probe from bones.
Hybridization showed that for c-abl about three-fold higher levels in the embryo proper than in extraembryonal membranes and placenta is observed at the 10th day of gestation, as compared to the concentration observed in the 6, 7, and 9 day conceptuses. Expression of c-abl in the fetus appears to decrease after the 11th day of prenatal development. The oncogene c-abl is transcriptionally active in all postnatal mouse tissue examined with spleen and thymus poly(A)+ RNA exhibiting a slightly stronger hybridization than from other tissues.
The oncogene c-rasHa was found to be expressed in considerable, but similar levels at all stages of prenatal development both in the embryo proper as well as in extraembryonal tissues. High levels of c-rasHa expression were also observed in various tissues of newborn or 10 day old mice, particularly in bone, brain, kidney, skin and spleen.
The oncogene c-myc was detectable at days 7 and 8, but much higher levels were observed in late embryonic development (days 17 and 18).
The oncogene c-erb had maximum hybridization at 13 days, while no hybridization was observed at day 6.
The oncogene c-src was detected at its highest levels in the latter half of mouse embryonic development with an increase beginning at day 14, peaking at day 15 and gradually decreasing thereafter. For the oncogene c-sis, peak expressions were observed at days 7 and 16, the day-7 peak was 1.5 to 3 times higher than all other days and the day-16 peak was 1.5 to 2 times higher than days 9 to 13 days 17 and 18.
In the next study, the nucleotide sequence of the presumed oncogene region of Avian myeloblastosis virus myb was employed (Vister et al. (1982) supra). Using the published nucleotide sequence, a number of antigenic oligopeptide sequences were derived and seven of the polypeptides so derived were synthesized and evaluated as being potentially antigenic. These seven oligopeptides, which are representative antigenic oligopeptides according to the invention, have the following formulas:
(1) pro-phe-his-lys-asp-gln-thr-phe-glu-tyr-arg-lys-met
(2) pro-ser-pro-pro-val-asp-his-gly-cys-leu-pro-glu-glu-ser-ala-ser-pro-ala-ar
(3) asp-asn-thr-arg-thr-ser-gly-asp-asn-ala-pro-val-ser-cys-leu-gly-glu
(4) pro-gln-glu-ser-ser-lys-ala-gly-pro-pro-ser-gly-thr-thr-gly
(5) met-ala-phe-ala-his-asn-pro-pro-ala-gly-pro-leu-pro-gly-ala
(6) pro-pro-val-asp-his-gly-cys-leu-pro-glu-glu-ser-ala-ser-pro-ala
(7) pro-phe-his-lys-asp-gln-thr-phe-thr-gly-tyr-arg-lys-met-his-gly-gly-ala-val
The polypeptides were linked to keyhole limpet hemocyanin in accordance with conventional techniques (Dockray, Regulatory Peptides (1980)1:169) and the resulting immunogen was used to immunize rabbits in a first injection with complete Freund's adjuvant, followed by injections with incomplete Freund's adjuvant over periods of three to four weeks to hyperimmunize the rabbits. The rabbits were bled repeatedly over a period of six months. Of the seven oligopeptides which resulted in the production of antibodies, antibodies to two peptides (5 and 7) were selected for detailed analysis. The antibodies were reacted with radioactively labeled cell lysates from a cell line containing multiple copies of the Avian myeloblastosis virus and with lysates from appropriate non-infected cell lines. Antibody No. 5 identifies a specific protein of approximately 58,000 daltons, which is present in the virus-infected cell line but not in controls.
Antibody against polypeptide 5 was also reacted with the plasma of chickens bearing tumors induced by amv. A band similar to that observed with the above lysates of approximately 48,000 daltons was identified.
Antisera to polypeptide No. 5 was also reacted with lysates of a myeloid human leukemia cell line (HL-60) which is known to express messenger RNA transcripts of the c-myb gene (Gallo and Wong-Staal, Blood (1982) 60:545). This antibody reacted with a protein of about 90,000 daltons. In freshly isolated myeloid leukemia cells, the antibody identifies a series of proteins, 14 kd to 70 kd, not present in normal white blood cells.
The polyconal antibodies to the fragment no. 5 of the myb protein was tested for its ability to kill normal and leukemic cells. The procedure employed is described in Terasaki and McClelland, infra. The data are set forth in the following table.
                                  TABLE 5                                 
__________________________________________________________________________
CYTOTOXICITY* OF ANTI-Myb2 ANTIBODY                                       
AGAINST HUMAN CELLS                                                       
              Cells.sup.++                                                
                                Common                                    
              Normal                                                      
                   Normal                                                 
                        Molt 4                                            
                            HL-60                                         
                                ALL   AML T-ALL                           
Antiserum                                                                 
         Dilution                                                         
              B    T    (T) (AML)                                         
                                1  2  (5)**                               
                                          1 2                             
__________________________________________________________________________
Medium Alone  1    1    1   1   1  1  1   1 1                             
(-control)                                                                
ALS.sup.+                                                                 
         --   8    8    8   8   8  8  8   8 8                             
(+control)                                                                
Anti-myb --   1    1    8   6   6  1  6   6 6                             
         1:2  1    1    6   1   1  1  6   6 2                             
         1:4  1    1    6   1   1  1  4   4 1                             
Pre-immune                                                                
         --   1    1    4   1   1  1  1   4 2                             
         1:2  1    1    2   1   1  1  1   2 2                             
         1:4  1    1    2   1   1  1  1   2 2                             
__________________________________________________________________________
 *8 = 80-100% killing 6 = 60-80% killing 1 = 0-10% killing                
 The method od Terasaki and McClelland, Nature (1964) 204-998, was employe
 for complement lysis.                                                    
 **3 of 5 are killed; a representative one is shown                       
 .sup.+ ALS  antileukocyte serum                                          
 .sup.++ Molt 4 is a nonmalignant human T lymphoid cell line that is known
 to express myb nRNA.                                                     
 HL60  human myeloid leukemia cell line                                   
 AML  acute myeloid leukemia                                              
 ALL  acute lymphocyte leukemia                                           
 TALL  Tcell active lymphoblastic leukemia                                
The above results demonstrate that the expression product of the myb gene can react with antibodies to produce lysis with complement. Thus, the myb protein appears to be a surface membrane protein which is available for binding to antibodies. By identifying proteins to which specific antibodies will bind, which proteins have diagnostic value as indicative of malignance, the malignant cells can be identified and treated. In the subject work, no determination has been made as to the specificity or cross-reactivity of the subject antibodies. Since only a fragment was used to prepare the antibodies, it would be expected that antibodies of greater binding specificity and avidity could be prepared with the whole protein, particularly with the whole protein in a membrane. The subject antibodies can be used to select for antibodies binding to the same or other determinant site.
The subject data demonstrate that antibodies can be prepared which do not affect normal B- and T-cells, but are cytotoxic in combination with complement for a variety of malignant cells. Therefore, the antibodies can be used for cancer therapy without the hazard of substantially inactivating the immune system.
It is evident from the above results, that one can detect the presence of malignancy in a human host by determining the transcription and/or expression products of the oncogene. One can screen retroviruses or other source of nucleic acids to .Iadd.demonstrate the ability of such nucleic acids to .Iaddend.transform vertebrates to malignancy. One may then use these nucleic acids to deduce peptide composition and screen malignant cells for transcripts or peptides, by hybridization in the former case and with appropriate receptors in the latter case, employing any of a wide variety of diagnostic assays. Antibodies can be produced to the peptides, which antibodies may be labeled and may then be used for diagnosing the presence of a peptide diagnostic of malignancy. The oncogernic proteins are found to be available for binding to antibodies as surface membrane proteins. The antibodies may serve as diagnostic reagents for determining the presence of malignancy and determining the location of malignant cells. The antibodies may also serve in treating tumors in vivo by using radionuclides, toxins, in combination with the host complement system or opsonins, or other antibody dependent lytic system or the like. The antibodies find use in pre- and postoperative systems, in the later determining whether complete removal has occurred, whether metastases exist. The antibodies can be used postoperatively to destroy any remnants of the tumor which may not have been excised.
Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be obvious that certain changes and modifications may be practiced within the scope of the appended claims.

Claims (15)

What is claimed is:
1. A method for evaluating the probability of cellular malignancy in a human host, said method comprising:
bringing into close associate (1) a probe specific for a cellular product, said cellular product being mRNA or its expression product, where said mRNA is complementary to a DNA sequence of a retrovirus capable of transforming a normal cell to malignancy and said probe is a nucleic acid sequence capable of duplexing with said mRNA or antibody capable of binding to said expression product, and (2) a source from said human host suspected of containing cellular product; and
determining the level of binding of said probe to said cellular product, wherein an elevated level is indicative of the presence of cellular malignancy.
2. A method according to claim 1, wherein said source is cells from said human host.
3. A method according to claim 1, wherein said source is a physiological fluid from said human host.
4. A method according to claim 1, wherein said DNA sequence is selected from the group consisting of the oncogenes src, fps, yes, fos, myc, erb, myb, rel, mos, bas, abl, ras, fes, fms, and sis.
5. A method according to any of claims 1, 2, 3, or 4, wherein said probe is an antibody.
6. A method according to claim 5, wherein said antibody is labeled with a label capable of providing a detectible signal.
7. A method according to claims 1, 2, 3, or 4, wherein said probe is a polynucleotide of at least 14 bases complementary to said mRNA.
8. A method for evaluating the probability of leukemia in a human host, said method comprising:
combining antibodies .[.specific.]. .Iadd.diagnostic for the presence of .Iaddend.for the oncogene myb and blood cells from a human host suspected of having leukemia; and
determining the level of binding of said antibodies to said host blood cells as diagnostic of a leukemic host.
9. A method according to claim 8, wherein said antibodies are produced in response to an oligopeptide mimicking a portion of the conformation of the myb protein. .[.10. A method for substantially eliminating human malignant cells from a combination of human malignant and normal cells, which comprises:
combining under cytotoxic conditions said combination of cells with an antibody specific for an expression product of a DNA sequence present in a retrovirus genome or substantially complementary to said DNA sequence, which sequence is expressed in said malignant cells as a surface protein; and
isolating normal cells, substantially free of malignant cells..]..[.11. A method according to claim 10, wherein said separation occurs in the presence of complement as said cytotoxic condition..]..[.12. A method according to claim 10, wherein said antibodies are labeled with a radionuclide as said cytotoxic condition..]..[.13. A method according to claims 10, 11, or 12, wherein said DNA sequence is the myb gene..]..[.14. A method for treating a human host suspected of having malignant cells, which comprises:
administering to said human host under cytotoxic conditions antibodies to the expression product of a gene, which gene is part of a retrovirus genome capable of inducing malignancy in a normal cell or which gene is substantially complementary to said gene of said retrovirus genome..]..[.15. A method according to claim 14, wherein said cytotoxic condition is the presence of complement..]..[.16. Antibodies specific for the expression product of the human oncogenes c-myc, c-fos, c-rasHa, c-rasKi, c-fes, c-myb, and c-src..]..[.17. Antibodies according to claim 16, labeled with a label capable of providing a detectible signal..]..[.18. Antibodies according to claim 16, labeled with a
cytotoxic agent..].19. An antigenic oligopeptide selected from the class consisting of:
(a) met-ala-phe-ala-his-asn-pro-pro-ala-gly-pro-leu-pro-gly-ala
(b) pro-phe-his-lys-asp-gln-thr-phe-thr-glu-tyr-arg-lsy-met-his-gly-gly-ala-va
(c) pro-phe-his-lys-asp-gln-thr-phe-thr-glu-tyr-arg-lys-met
(d) asp-asn-thr-arg-thr-ser-gly-asp-asn-ala-pro-val-ser-cys-leu-gly-glu
(e) arg-leu-ileu-gly-asp-asn-glu-tyr-thr-ala-arg-gln-gly-ala-lys-phe-pro
(f) trp-arg-arg-asp-pro-glu-glu-arg-pro-thr
(g) arg-leu-lys-lys-ileu-ser-lys-glu-glu-lys-thr-pro-gly-cys-val-lys-ileu-lys-lys
(h) asp-leu-pro-ser-arg-thr-val-asp-thr-lys-gln-ala-gln-glu-leu-ala-arg
(i) met-thr-glu-tyr-lys-leu-val-val-val-gly-ala-ser-gly-val-gly-lys-ser-ala
(j) glu-asp-ileu-his-gln-try-arg-glu-gln-ileu-lys-arg-val-lys-asp-ser-asp-asp
(k) val-arg-glu-ileu-arg-gln-his-lys-leu-arg-lys-leu-asn-pro-pro-asp-glu-ser-gly-pro
(l) met-thr-gly-tyr-lys-leu-val-val-val-gly-ala-gly-gly-val-gly-lys-ser-ala
(m) val-asp-glu-tyr-asp-pro-thr-ileu-glu-asp-ser-tyr-arg-lys-gln-val
(n) arg-his-ser-thr-ser-ser-ser-glu-gln-glu-arg-glu-gly-gly-arg
(o) asn-gln-gln-thr-arg-glu-phe-val-glu-lys-gly-gly-arg
(p) pro-glu-val-gln-lys-pro-leu-his-glu-gln
(q) ala-ser-pro-tyr-pro-asn-leu-ser-asn-gln-gln-thr-arg
(r) arg-leu-ileu-ala-glu-lys-glu-gln-leu-arg-arg-arg-arg-glu-gln
(s) asn-asn-glu-lys-ala-pro-lys-val-val. 20. Antibodies raised to an antigenic polypeptide selected from the class consisting of:
(a) met-ala-phe-ala-his-asn-pro-pro-ala-gly-pro-leu-pro-gly-ala;
(b) pro-phe-his-lys-asp-gln-thr-phe-thr-glu-tyr-arg-lys-met-his-gly-gly-ala-val;
(c) pro-phe-his-lys-asp-gln-thr-phe-thr-glu-tyr-arg-lys-met;
(d) asp-asn-thr-arg-thr-ser-gly-asp-asn-ala-pro-val-ser-cys-leu-gly-glu;
(e) arg-leu-ileu-glu-asp-asn-glu-tyr-thr-ala-arg-gln-gly-ala-lys-phe-pro;
(f) trp-arg-arg-asp-pro-glu-glu-arg-pro-thr;
(g) arg-leu-lys-lys-ileu-ser-lys-glu-glu-lys-thr-pro-gly-cys-val-lys-ileu-lys-lys;
(h) asp-leu-pro-ser-arg-thr-val-asp-thr-lys-gln-ala-gln-glu-leu-ala-arg;
(i) met-thr-glu-try-lys-leu-val-val-val-gly-ala-ser-gly-val-gly-lys-ser-ala;
(j) glu-asp-ileu-his-gln-tyr-arg-glu-gln-ileu-lys-arg-val-lys-asp-ser-asp-asp;
(k) val-arg-glu-ileu-arg-gln-his-lys-leu-arg-lys-leu-asn-pro-pro-asp-glu-ser-gly-pro;
(l) met-thr-glu-tyr-lys-leu-val-val-gly-ala-gly-gly-val-gly-lys-ser-ala;
(m) val-asp-glu-tyr-asp-pro-thr-ileu-glu-asp-ser-tyr-arg-lys-gln-val;
(n) arg-his-ser-thr-ser-ser-ser-glu-gln-glu-arg-glu-gly-gly-arg;
(o) asn-gln-gln-thr-arg-glu-phe-val-glu-lys-gly-gly-arg;
(p) pro-glu-val-gln-lys-pro-leu-his-glu-gln;
(q) ala-ser-pro-tyr-pro-asn-leu-ser-asn-gln-gln-thr-arg;
(r) arg-leu-ileu-ala-glu-lys-glu-gln-leu-arg-arg-arg-arg-glu-gln; and
(s) asn-asn-glu-lys-ala-pro-lys-val-val. 21. Antibodies according to claim
20, labeled with a label capable of providing a detectible signal. 22. Antibodies according to claim 20, labeled with a cytotoxic agent.
.Iadd.23. A method for evaluating the probability of cellular malignancy in a human host, said method comprising:
bringing into close association (1) a probe diagnostic for the presence of a cellular product, said cellular product being mRNA or its expression product, where said mRNA is capable of hybridizing to a DNA sequence of a retrovirus capable of transforming a normal cell to malignancy and said probe is a nucleic acid sequence capable of duplexing with said mRNA or antibody capable of binding to said expression product, and (2) a source from said human host suspected of containing cellular product; and
determining the level of said binding of said probe to said cellular product, wherein an elevated level is indicative of the presence of
cellular malignancy. .Iaddend..Iadd.24. An antibody diagnostic for the presence of the expression product of a human oncogene selected from the group consisting of c-myc, c-fos, c-rasK, c-fes, and c-myb. .Iaddend..Iadd.25. An antibody according to claim 24, wherein said human oncogene is c-myc. .Iaddend..Iadd.26. An antibody according to claim 24, wherein said human oncogene is c-fos. .Iaddend..Iadd.27. An antibody according to claim 24, wherein said human oncogene is c-rasK. .Iaddend..Iadd.28. An antibody according to claim 24, wherein said human oncogene is c-fes. .Iaddend..Iadd.29. An antibody according to claim 24, wherein said human oncogene is c-myb. .Iaddend..Iadd.30. An antibody according to claim 24, labeled with a label capable of providing a detectable signal. .Iaddend..Iadd.31. An antibody according to claim 24, labeled with a cytotoxic agent. .Iaddend.
US07/421,096 1982-11-04 1989-10-12 Methods and compositions for detecting human tumors Expired - Lifetime USRE35491E (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US07/421,096 USRE35491E (en) 1982-11-04 1989-10-12 Methods and compositions for detecting human tumors

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US43925282A 1982-11-04 1982-11-04
US49602783A 1983-05-19 1983-05-19
US06/673,469 US4699877A (en) 1982-11-04 1984-11-20 Methods and compositions for detecting human tumors
US07/421,096 USRE35491E (en) 1982-11-04 1989-10-12 Methods and compositions for detecting human tumors

Related Parent Applications (3)

Application Number Title Priority Date Filing Date
US43925282A Continuation-In-Part 1982-11-04 1982-11-04
US49602783A Continuation-In-Part 1982-11-04 1983-05-19
US06/673,469 Reissue US4699877A (en) 1982-11-04 1984-11-20 Methods and compositions for detecting human tumors

Publications (1)

Publication Number Publication Date
USRE35491E true USRE35491E (en) 1997-04-08

Family

ID=27412019

Family Applications (1)

Application Number Title Priority Date Filing Date
US07/421,096 Expired - Lifetime USRE35491E (en) 1982-11-04 1989-10-12 Methods and compositions for detecting human tumors

Country Status (1)

Country Link
US (1) USRE35491E (en)

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6054561A (en) 1984-02-08 2000-04-25 Chiron Corporation Antigen-binding sites of antibody molecules specific for cancer antigens
US6403766B1 (en) 1999-10-15 2002-06-11 Cornell Research Foundation, Inc. Human actin regulatory proteins and methods for detection, diagnosis and treatment of different stages of carcinogenesis
US20040157255A1 (en) * 2003-02-06 2004-08-12 David Agus Gene expression markers for response to EGFR inhibitor drugs
US20040191817A1 (en) * 2003-02-20 2004-09-30 Randy Scott Use of intronic RNA to measure gene expression
US20040209290A1 (en) * 2003-01-15 2004-10-21 Cobleigh Melody A. Gene expression markers for breast cancer prognosis
US20050019785A1 (en) * 2002-11-15 2005-01-27 Baker Joffre B. Gene expression profiling of EGFR positive cancer
US20050064455A1 (en) * 2003-05-28 2005-03-24 Baker Joffre B. Gene expression markers for predicting response to chemotherapy
US20050131006A1 (en) * 2003-10-14 2005-06-16 Ali Mukherjee Receptor tyrosine kinase signaling pathway analysis for diagnosis and therapy
US20050130238A1 (en) * 2003-04-01 2005-06-16 Po-Ying Chan-Hui ErbB surface receptor complexes as biomarkers
US20050164218A1 (en) * 2003-05-30 2005-07-28 David Agus Gene expression markers for response to EGFR inhibitor drugs
US20050196782A1 (en) * 2003-12-23 2005-09-08 Kiefer Michael C. Universal amplification of fragmented RNA
US7034144B2 (en) 1997-05-13 2006-04-25 Erasmus Universiteit Rotterdam Molecular detection of chromosome aberrations
US20060160106A1 (en) * 1998-05-04 2006-07-20 Dako A/S Method and probes for the detection of chromosome aberrations
US20060166230A1 (en) * 2004-11-05 2006-07-27 Baker Joffre B Predicting response to chemotherapy using gene expression markers
US20070059737A1 (en) * 2002-03-13 2007-03-15 Baker Joffre B Gene expression profiling in biopsied tumor tissues
US7402397B2 (en) 2002-05-21 2008-07-22 Monogram Biosciences, Inc. Detecting and profiling molecular complexes
US20080187948A1 (en) * 2002-05-21 2008-08-07 Monogram Biosciences Inc. Erbb heterodimers as biomarkers
US7526387B2 (en) 2003-07-10 2009-04-28 Genomic Health, Inc. Expression profile algorithm and test for cancer prognosis
US7622251B2 (en) 2004-11-05 2009-11-24 Genomic Health, Inc. Molecular indicators of breast cancer prognosis and prediction of treatment response
US7723033B2 (en) 2003-06-24 2010-05-25 Genomic Health, Inc. Prediction of likelihood of cancer recurrence
US7871769B2 (en) 2004-04-09 2011-01-18 Genomic Health, Inc. Gene expression markers for predicting response to chemotherapy

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2034323A (en) * 1978-12-18 1980-06-04 Inst Nat Sante Rech Med Production of DNA Comprising B Hepatitis Viral Genome
US4331647A (en) * 1980-03-03 1982-05-25 Goldenberg Milton David Tumor localization and therapy with labeled antibody fragments specific to tumor-associated markers
US4348376A (en) * 1980-03-03 1982-09-07 Goldenberg Milton David Tumor localization and therapy with labeled anti-CEA antibody
US4358535A (en) * 1980-12-08 1982-11-09 Board Of Regents Of The University Of Washington Specific DNA probes in diagnostic microbiology
US4361544A (en) * 1980-03-03 1982-11-30 Goldenberg Milton David Tumor localization and therapy with labeled antibodies specific to intracellular tumor-associated markers
US4535058A (en) * 1982-10-01 1985-08-13 Massachusetts Institute Of Technology Characterization of oncogenes and assays based thereon

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2034323A (en) * 1978-12-18 1980-06-04 Inst Nat Sante Rech Med Production of DNA Comprising B Hepatitis Viral Genome
US4331647A (en) * 1980-03-03 1982-05-25 Goldenberg Milton David Tumor localization and therapy with labeled antibody fragments specific to tumor-associated markers
US4348376A (en) * 1980-03-03 1982-09-07 Goldenberg Milton David Tumor localization and therapy with labeled anti-CEA antibody
US4361544A (en) * 1980-03-03 1982-11-30 Goldenberg Milton David Tumor localization and therapy with labeled antibodies specific to intracellular tumor-associated markers
US4358535A (en) * 1980-12-08 1982-11-09 Board Of Regents Of The University Of Washington Specific DNA probes in diagnostic microbiology
US4358535B1 (en) * 1980-12-08 1986-05-13
US4535058A (en) * 1982-10-01 1985-08-13 Massachusetts Institute Of Technology Characterization of oncogenes and assays based thereon

Non-Patent Citations (104)

* Cited by examiner, † Cited by third party
Title
Becker et al., Proc. Natl. Acad. Sci. USA (1982) 79:3315 3319. *
Becker et al., Proc. Natl. Acad. Sci. USA (1982) 79:3315-3319.
Bishop, New England J. of Med. (1980) 303:675 681. *
Bishop, New England J. of Med. (1980) 303:675-681.
Bishop, Scientific American, Mar. 1982, 81 93. *
Bishop, Scientific American, Mar. 1982, 81-93.
Chang E.H., et al., Characterization of Four Members of the p21 Gene Family Isolated from Normal Human Genomic DNA and Demonstration of their Oncogenic Potential. RNA Tumor Viruses, Cold Spring Harbor Symposium, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, May 26 30, 1982. *
Chang E.H., et al., Characterization of Four Members of the p21 Gene Family Isolated from Normal Human Genomic DNA and Demonstration of their Oncogenic Potential. RNA Tumor Viruses, Cold Spring Harbor Symposium, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, May 26-30, 1982.
Cooper, Science (1982) 218:801 806. *
Cooper, Science (1982) 218:801-806.
Cronkite et al, Chem. Abstracts, 99(1983) 35338n. *
Cronkite et al, Chem. Abstracts, 99(1983)#35338n.
Dalla Favera, R. et al., Nature 299:61 63 (1982). *
Dalla Favera, R. et al., Nature 299:61-63 (1982).
Dhar, Science (1982) 217:934 936. *
Dhar, Science (1982) 217:934-936.
Essex, J. Natl. Cancer Inst., 69(1982) 981 5. *
Essex, J. Natl. Cancer Inst., 69(1982) 981-5.
Eva, A. et al., Nature 295:116 119 (1982). *
Eva, A. et al., Nature 295:116-119 (1982).
Eva, A., et al. (I), Human Tumor Cells Contain Transcripts Related to the Transforming Genes of Retroviruses. RNA Tumor Viruses, Cold Spring Harbor Symposium, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, May 20 24, 1981. *
Eva, A., et al. (I), Human Tumor Cells Contain Transcripts Related to the Transforming Genes of Retroviruses. RNA Tumor Viruses, Cold Spring Harbor Symposium, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, May 20-24, 1981.
Eva, A., et al. (II), Transcription of Retrovirus onc Gene Analogues in Human Solid Tumor Cells. Advances in Comparative Leukemia Research 1981, pp. 381 382, Proceedings of the Xth International Symposium for Comparative Research on Leukemia and Related Diseases, held at U.C.L.A., Aug. 31 Sep. 4, 1981, published/copyrighted 1982 by Elsevier North Holland, Inc., D.S. Yohn and J.R. Blakeslee, editors, Elsevier Biomedical. *
Eva, A., et al. (II), Transcription of Retrovirus onc Gene Analogues in Human Solid Tumor Cells. Advances in Comparative Leukemia Research 1981, pp. 381-382, Proceedings of the Xth International Symposium for Comparative Research on Leukemia and Related Diseases, held at U.C.L.A., Aug. 31-Sep. 4, 1981, published/copyrighted 1982 by Elsevier North Holland, Inc., D.S. Yohn and J.R. Blakeslee, editors, Elsevier Biomedical.
Furth, M.E., et al., Characterization of the p21 src Protein of Harvey Sarcoma Virus and Related Using Monoclonal Antibodies. RNA Tumor Viruses, Cold Spring Harbor Symposium, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, May 20 24, 1981. *
Furth, M.E., et al., Characterization of the p21 src Protein of Harvey Sarcoma Virus and Related Using Monoclonal Antibodies. RNA Tumor Viruses, Cold Spring Harbor Symposium, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, May 20-24, 1981.
Gallo et al, Proc. Natl. Acad. Sci. USA, 79(1982)5680 3. *
Gallo et al, Proc. Natl. Acad. Sci. USA, 79(1982)5680-3.
Gallo, R.C. et al., Blood 60:545 557 (1982). *
Gallo, R.C. et al., Blood 60:545-557 (1982).
Gowans et al, J. Medvirology, 8(1981)67 78. *
Gowans et al, J. Medvirology, 8(1981)67-78.
Heilmann et al., J. Virology, 24(1977) 498 504. *
Heilmann et al., J. Virology, 24(1977) 498-504.
Herbrink et al, Cancer Res., 40(1980)166 73. *
Herbrink et al, Cancer Res., 40(1980)166-73.
Hopp et al, Proc. Acad. Sci. USA, 78(1981)3824 8. *
Hopp et al, Proc. Acad. Sci. USA, 78(1981)3824-8.
Larsen et al., Proc. Natl. Acad. Sci. USA 72:4900 4904 (1975). *
Larsen et al., Proc. Natl. Acad. Sci. USA 72:4900-4904 (1975).
Larsen, C.J. et al., Search for Nucleic Acid Sequences Complementary to a Murine Oncornaviral Genome in Poly(A) Rich RNA of Human Leukemic Cells. Proc. Natl. Acad. Sci. USA 72(12):4900 4904, 1975. *
Larsen, C.J. et al., Search for Nucleic Acid Sequences Complementary to a Murine Oncornaviral Genome in Poly(A)-Rich RNA of Human Leukemic Cells. Proc. Natl. Acad. Sci. USA 72(12):4900-4904, 1975.
Microbiology, 3rd ed, Harper & Row, Philadelphia, 1980, 539 47, 1232 61. *
Microbiology, 3rd ed, Harper & Row, Philadelphia, 1980, 539-47, 1232-61.
Nevans et al, Adv. Virus Res, 26(1981) 1 35. *
Nevans et al, Adv. Virus Res, 26(1981) 1-35.
Oppermann et al., Proc. Natl. Acad. Scie. USA 76:1804 1808 (1979). *
Oppermann et al., Proc. Natl. Acad. Scie. USA 76:1804-1808 (1979).
Oppermann, H. et al., Uninfected Vertebrate Cells Contain a Protein that Is Closely Related to the Product of the Avian Sarcoma Virus Transforming Gene(src). Proc. Natl. Acad. Sci. USA 76:1804 1808, 1979. *
Oppermann, H. et al., Uninfected Vertebrate Cells Contain a Protein that Is Closely Related to the Product of the Avian Sarcoma Virus Transforming Gene(src). Proc. Natl. Acad. Sci. USA 76:1804-1808, 1979.
Ozanne, B., et al., Arrangement and Expression of Abelson Murine Leukemia Virus Related Nucleic Acid Sequences in a Novel Human pre B Lymphoblastic Leukemia. RNA Tumor Viruses, Cold Spring Harbor Symposium, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, May 26 30, 1982. *
Ozanne, B., et al., Arrangement and Expression of Abelson Murine Leukemia Virus Related Nucleic Acid Sequences in a Novel Human pre-B Lymphoblastic Leukemia. RNA Tumor Viruses, Cold Spring Harbor Symposium, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, May 26-30, 1982.
Petterson et al, J. Mol. Biol., 73(1973)125 30. *
Petterson et al, J. Mol. Biol., 73(1973)125-30.
Poiesz et al, Nature, 294(19 Nov 1981) 268 71. *
Poiesz et al, Nature, 294(19 Nov 1981) 268-71.
Prakash, K., et al., Analysis of Sequences in Human DNA Related to the Transforming Genes of Murine Transforming Viruses. RNA Tumor Viruses, Cold Spring Harbor Symposium, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, May 20 24, 1981. *
Prakash, K., et al., Analysis of Sequences in Human DNA Related to the Transforming Genes of Murine Transforming Viruses. RNA Tumor Viruses, Cold Spring Harbor Symposium, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, May 20-24, 1981.
Reitz, M.F., et al., Characterization and Distribution of Nucleic Acid Sequences of a Novel Type C Retrovirus Isolated from Neoplastic Human T Lymphocytes. Proc. Natl. Acad. Sci. USA 76, 78(3):1887 1891, 1981. *
Reitz, M.F., et al., Characterization and Distribution of Nucleic Acid Sequences of a Novel Type C Retrovirus Isolated from Neoplastic Human T Lymphocytes. Proc. Natl. Acad. Sci. USA 76, 78(3):1887-1891, 1981.
Rohrschneider et al., Proc. Natl. Acad. Sci. USA 76:4479 4483 (1979). *
Rohrschneider et al., Proc. Natl. Acad. Sci. USA 76:4479-4483 (1979).
Rohrschneider, L.R., et al., Identification of a Rous sarcoma Virus Transformation Related Protein in Normal Avian and Mammalian Cells. Proc. Natl. Acad. Sci. USA 76:4479 4483, 1979. *
Rohrschneider, L.R., et al., Identification of a Rous sarcoma Virus Transformation-Related Protein in Normal Avian and Mammalian Cells. Proc. Natl. Acad. Sci. USA 76:4479-4483, 1979.
Rosson, D. and Tereba, A., Transcription of Oncogenes in Human Primary Tumors. RNA Tumor Viruses, Cold Spring Harbor Symposium, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, May 26 30, 1982. *
Rosson, D. and Tereba, A., Transcription of Oncogenes in Human Primary Tumors. RNA Tumor Viruses, Cold Spring Harbor Symposium, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, May 26-30, 1982.
Rovigattim U.G., et al., Oncogene Expression in Human Tumors. RNA Tumor Viruses, Cold Spring Harbor Symposium, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, May 26 30, 1982. *
Rovigattim U.G., et al., Oncogene Expression in Human Tumors. RNA Tumor Viruses, Cold Spring Harbor Symposium, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, May 26-30, 1982.
Sambrook, et al (1989) Molecular Cloning: A Laboratory Manual (CSH Press) pp. 9.56, 9.57, 11.7 and 11.8. *
Santos, E., et al., T24 Human Bladder Carcinoma Oncogene Is an Activated Form of the Norman Human Homologue of Balb and Harvey MSV transforming Genes. Nature 298:343 347, 1982. *
Santos, E., et al., T24 Human Bladder Carcinoma Oncogene Is an Activated Form of the Norman Human Homologue of Balb-and Harvey MSV transforming Genes. Nature 298:343-347, 1982.
Shafrite et al, Hepatology, 1(1981). *
Spector, Current Top. Microbiol. Immunol., 1981, 41 80. *
Spector, Current Top. Microbiol. Immunol., 1981, 41-80.
Stehelin et al, Chemical Abstracts, 94(1981) 27211 X. *
Stehelin et al, Chemical Abstracts, 94(1981)#27211 X.
Stehelin et al, Cold Spring Harbor Sympos. on Quant. Biol. vol. 44, pp. 1215 23(1980). *
Stehelin et al, Cold Spring Harbor Sympos. on Quant. Biol. vol. 44, pp. 1215-23(1980).
Stone et al, Virology, 100 (1980)370 81. *
Stone et al, Virology, 100 (1980)370-81.
Sutcliffe et al, Nature, 287(0ct. 30, 1980) 8D1 5. *
Sutcliffe et al, Nature, 287(0ct. 30, 1980) 8D1-5.
The Lancet, Jul. 24, 1982, 195 196. *
The Lancet, Jul. 24, 1982, 195-196.
Tsuchida, Science (1982) 217:937 938. *
Tsuchida, Science (1982) 217:937-938.
Tsuchida, Science, 217(1982)937 8. *
Tsuchida, Science, 217(1982)937-8.
Varmus, Science (1982) 216:812 820. *
Varmus, Science (1982) 216:812-820.
Westin, E., et al. (Westin II), Expression of the abl, myc, and amy Genes in Human Hematopoietic Cells. Advances in Comparative Leukemia Research 1981, pp. 405 406, Proceedings of the Xth International Symposium for Comparative Research on Leukemia and Related Diseases, held at U.C.L.A., Aug. 31 Sep. 4, 1981, published/copyrighted 1982 by Elsevier North Holland, Inc., D.S. Yohn and J.R. Blakeslee, editors, Elsevier Biomedical. *
Westin, E., et al. (Westin II), Expression of the abl, myc, and amy Genes in Human Hematopoietic Cells. Advances in Comparative Leukemia Research 1981, pp. 405-406, Proceedings of the Xth International Symposium for Comparative Research on Leukemia and Related Diseases, held at U.C.L.A., Aug. 31-Sep. 4, 1981, published/copyrighted 1982 by Elsevier North Holland, Inc., D.S. Yohn and J.R. Blakeslee, editors, Elsevier Biomedical.
Westrin, E.H., et al. (Westin I), Human Hematopoietic Cells Express Cellular Homologs of Retroviral Onc Genes. RNA Tumor Viruses, Cold Spring Harbor Symposium, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, May 26 30, 1982. *
Westrin, E.H., et al. (Westin I), Human Hematopoietic Cells Express Cellular Homologs of Retroviral Onc Genes. RNA Tumor Viruses, Cold Spring Harbor Symposium, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, May 26-30, 1982.
Wong Staal et al, Proc. Natl. Acad. Sci. USA 79(1982)2490 4. *
Wong Staal et al, Science,213(1981)226 8. *
Wong Staal, F. et al., Science 213:226 228 (1981). *
Wong Staal, F., et al., Cloning of the Simian Sarcoma Virus Genome and Its Homologous Human onc Gene and Studies on the Expression of this Gene in Different Human Hematopoietic Cells. Advances in Comparative Leukemia Research 1981, pp. 407 408, Proceedings of Xth International Symposium for Comparative Research on Leukemia and Related Diseases, held at U.C.L.A., Aug. 31 Sep. 4, 1981, published/copyrighted 1982 by Elsevier North Holland, Inc., D.S. Yohn and J.R. Blakeslee, editors, Elsevier Biomedical. *
Wong-Staal et al, Proc. Natl. Acad. Sci. USA 79(1982)2490-4.
Wong-Staal et al, Science,213(1981)226-8.
Wong-Staal, F. et al., "Science" 213:226-228 (1981).
Wong-Staal, F., et al., Cloning of the Simian Sarcoma Virus Genome and Its Homologous Human onc Gene and Studies on the Expression of this Gene in Different Human Hematopoietic Cells. Advances in Comparative Leukemia Research 1981, pp. 407-408, Proceedings of Xth International Symposium for Comparative Research on Leukemia and Related Diseases, held at U.C.L.A., Aug. 31-Sep. 4, 1981, published/copyrighted 1982 by Elsevier North Holland, Inc., D.S. Yohn and J.R. Blakeslee, editors, Elsevier Biomedical.
Yoshida et al, Proc. Natl. Acad. Sci. USA, 79(1982)2031 5. *
Yoshida et al, Proc. Natl. Acad. Sci. USA, 79(1982)2031-5.

Cited By (58)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6054561A (en) 1984-02-08 2000-04-25 Chiron Corporation Antigen-binding sites of antibody molecules specific for cancer antigens
US20110020822A1 (en) * 1997-05-13 2011-01-27 Eramus Universiteit Rotterdam Molecular detection of chromosome aberrations
US7034144B2 (en) 1997-05-13 2006-04-25 Erasmus Universiteit Rotterdam Molecular detection of chromosome aberrations
US7642057B2 (en) 1998-05-04 2010-01-05 Dako Denmark A/S Method and probes for the detection of chromosome aberrations
US20080187934A1 (en) * 1998-05-04 2008-08-07 Dako A/S Method and probes for the detection of chromosome aberrations
US7368245B2 (en) 1998-05-04 2008-05-06 Dako Denmark A/S Method and probes for the detection of chromosome aberrations
US7105294B2 (en) 1998-05-04 2006-09-12 Dako Denmark A/S Method and probes for the detection of chromosome aberrations
US20060160106A1 (en) * 1998-05-04 2006-07-20 Dako A/S Method and probes for the detection of chromosome aberrations
US6403766B1 (en) 1999-10-15 2002-06-11 Cornell Research Foundation, Inc. Human actin regulatory proteins and methods for detection, diagnosis and treatment of different stages of carcinogenesis
US8071286B2 (en) 2002-03-13 2011-12-06 Genomic Health, Inc. Gene expression profiling in biopsied tumor tissues
US20100209920A1 (en) * 2002-03-13 2010-08-19 Baker Joffre B Gene Expression Profiling in Biopsied Tumor Tissues
US7838224B2 (en) 2002-03-13 2010-11-23 Genomic Health, Inc. Gene expression profiling in biopsied tumor tissues
US7858304B2 (en) 2002-03-13 2010-12-28 Genomic Health, Inc. Gene expression profiling in biopsied tumor tissues
US10241114B2 (en) 2002-03-13 2019-03-26 Genomic Health, Inc. Gene expression profiling in biopsied tumor tissues
US20080182255A1 (en) * 2002-03-13 2008-07-31 Baker Joffre B Gene Expression Profiling in Biopsied Tumor Tissues
US20070059737A1 (en) * 2002-03-13 2007-03-15 Baker Joffre B Gene expression profiling in biopsied tumor tissues
US20070065845A1 (en) * 2002-03-13 2007-03-22 Baker Joffre B Gene expression profiling in biopsied tumor tissues
US20070141588A1 (en) * 2002-03-13 2007-06-21 Baker Joffre B Gene expression profiling in biopsied tumor tissues
US7402397B2 (en) 2002-05-21 2008-07-22 Monogram Biosciences, Inc. Detecting and profiling molecular complexes
US20080187948A1 (en) * 2002-05-21 2008-08-07 Monogram Biosciences Inc. Erbb heterodimers as biomarkers
US20050019785A1 (en) * 2002-11-15 2005-01-27 Baker Joffre B. Gene expression profiling of EGFR positive cancer
US8148076B2 (en) 2002-11-15 2012-04-03 Genomic Health, Inc. Gene expression profiling of EGFR positive cancer
US8008003B2 (en) 2002-11-15 2011-08-30 Genomic Health, Inc. Gene expression profiling of EGFR positive cancer
US9944990B2 (en) 2003-01-15 2018-04-17 Genomic Health, Inc. Gene expression markers for breast cancer prognosis
US8034565B2 (en) 2003-01-15 2011-10-11 Genomic Health, Inc. Gene expression markers for breast cancer prognosis
US11220715B2 (en) 2003-01-15 2022-01-11 Genomic Health, Inc. Gene expression markers for breast cancer prognosis
US20040209290A1 (en) * 2003-01-15 2004-10-21 Cobleigh Melody A. Gene expression markers for breast cancer prognosis
US7569345B2 (en) 2003-01-15 2009-08-04 Genomic Health, Inc. Gene expression markers for breast cancer prognosis
US8741605B2 (en) 2003-01-15 2014-06-03 Genomic Health, Inc. Gene expression markers for breast cancer prognosis
US8206919B2 (en) 2003-01-15 2012-06-26 Genomic Health, Inc. Gene expression markers for breast cancer prognosis
US20040157255A1 (en) * 2003-02-06 2004-08-12 David Agus Gene expression markers for response to EGFR inhibitor drugs
US7767391B2 (en) 2003-02-20 2010-08-03 Genomic Health, Inc. Use of intronic RNA to measure gene expression
US20040191817A1 (en) * 2003-02-20 2004-09-30 Randy Scott Use of intronic RNA to measure gene expression
US20050170439A1 (en) * 2003-04-01 2005-08-04 Po-Ying Chan-Hui Methods for Detecting Receptor Complexes Comprising PI3K
US7648828B2 (en) 2003-04-01 2010-01-19 Monogram Biosciences, Inc. Methods for detecting receptor complexes comprising PI3K
USRE44437E1 (en) 2003-04-01 2013-08-13 Monogram Biosciences, Inc. Methods for detecting receptor complexes comprising PI3K
US20050170438A1 (en) * 2003-04-01 2005-08-04 Po-Ying Chan-Hui Methods for Detecting Receptor Complexes Comprising PDGFR
US20050130238A1 (en) * 2003-04-01 2005-06-16 Po-Ying Chan-Hui ErbB surface receptor complexes as biomarkers
US20050064455A1 (en) * 2003-05-28 2005-03-24 Baker Joffre B. Gene expression markers for predicting response to chemotherapy
US20080318230A1 (en) * 2003-05-30 2008-12-25 David Agus Gene expression markers for response to egfr inhibitor drugs
US20050164218A1 (en) * 2003-05-30 2005-07-28 David Agus Gene expression markers for response to EGFR inhibitor drugs
US7723033B2 (en) 2003-06-24 2010-05-25 Genomic Health, Inc. Prediction of likelihood of cancer recurrence
US20100267032A1 (en) * 2003-06-24 2010-10-21 Baker Joffre B Prediction of Likelihood of Cancer Recurrence
US10619215B2 (en) 2003-06-24 2020-04-14 Genomic Health, Inc. Prediction of likelihood of cancer recurrence
US7526387B2 (en) 2003-07-10 2009-04-28 Genomic Health, Inc. Expression profile algorithm and test for cancer prognosis
US20090280490A1 (en) * 2003-07-10 2009-11-12 Baker Joffre B Expression Profile Algorithm and Test for Cancer Prognosis
US7939261B2 (en) 2003-07-10 2011-05-10 Genomic Health, Inc. Expression profile algorithm and test for cancer prognosis
US7402399B2 (en) 2003-10-14 2008-07-22 Monogram Biosciences, Inc. Receptor tyrosine kinase signaling pathway analysis for diagnosis and therapy
US20050131006A1 (en) * 2003-10-14 2005-06-16 Ali Mukherjee Receptor tyrosine kinase signaling pathway analysis for diagnosis and therapy
US8329398B2 (en) 2003-12-23 2012-12-11 Genomic Health, Inc. Universal amplification of fragmented RNA
US20050196782A1 (en) * 2003-12-23 2005-09-08 Kiefer Michael C. Universal amplification of fragmented RNA
US7871769B2 (en) 2004-04-09 2011-01-18 Genomic Health, Inc. Gene expression markers for predicting response to chemotherapy
US9605318B2 (en) 2004-04-09 2017-03-28 Genomic Health, Inc. Gene expression markers for predicting response to chemotherapy
US7622251B2 (en) 2004-11-05 2009-11-24 Genomic Health, Inc. Molecular indicators of breast cancer prognosis and prediction of treatment response
US7930104B2 (en) 2004-11-05 2011-04-19 Genomic Health, Inc. Predicting response to chemotherapy using gene expression markers
US8868352B2 (en) 2004-11-05 2014-10-21 Genomic Health, Inc. Predicting response to chemotherapy using gene expression markers
US20110178374A1 (en) * 2004-11-05 2011-07-21 Baker Joffre B Predicting Response to Chemotherapy Using Gene Expression Markers
US20060166230A1 (en) * 2004-11-05 2006-07-27 Baker Joffre B Predicting response to chemotherapy using gene expression markers

Similar Documents

Publication Publication Date Title
US4699877A (en) Methods and compositions for detecting human tumors
EP0108564B1 (en) Methods for oncogenic detection
USRE35491E (en) Methods and compositions for detecting human tumors
US5506344A (en) Antibodies specific for BCL-2 gene product
EP0453560B1 (en) Localization and characterization of the wilms' tumor gene
US5202429A (en) DNA molecules having human BCL-2 gene sequences
CA2479476C (en) Compositions and methods for the treatment of tumor
AU2003201315C1 (en) WNT-1 Induced secreted polypeptides; WISP-1, -2 and -3
JP2006525026A (en) Use of JAG2 expression in the diagnosis of plasma cell diseases
JP5031581B2 (en) AML, B-ALL and T-ALL diagnostic markers
US5015571A (en) Polypeptide-induced monoclonal receptors to protein ligands
US5753437A (en) Method of diagnosing cancer susceptibility or metastatic potential
US20010014474A1 (en) Development of DNA probes and immunological reagents of human tumor associated antigens
US20070048808A1 (en) Hepatocellular carcinoma screening
JP2726264B2 (en) Assays and antibodies for N-myc proteins
JP4227881B2 (en) Diagnostic agent for adult T-cell leukemia
JPH08500731A (en) Diagnostic method
DK174459B1 (en) Detection and treatment of cancer - using antibodies to oncogenic expression prods.
DK174458B1 (en) Detection and treatment of cancer - using antibodies to oncogenic expression prods.
US6159751A (en) Development of DNA probes and immunological reagents of human tumor associated antigens
KR100861465B1 (en) A gastric carcinoma gene znf312b, a protein translated from the gene and a diagnostic kit using the same
JPH11507014A (en) Immunoassay using antibodies directed to the reaction products of the enzymes used as labels

Legal Events

Date Code Title Description
FEPP Fee payment procedure

Free format text: PAYOR NUMBER ASSIGNED (ORIGINAL EVENT CODE: ASPN); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

FPAY Fee payment

Year of fee payment: 12