US20060292113A1 - Prophylactic and therapeutic treatment of the ductal epithelium of a mammary gland for cancer - Google Patents

Prophylactic and therapeutic treatment of the ductal epithelium of a mammary gland for cancer Download PDF

Info

Publication number
US20060292113A1
US20060292113A1 US11/467,838 US46783806A US2006292113A1 US 20060292113 A1 US20060292113 A1 US 20060292113A1 US 46783806 A US46783806 A US 46783806A US 2006292113 A1 US2006292113 A1 US 2006292113A1
Authority
US
United States
Prior art keywords
epithelium
ductal
cells
ductal epithelium
cancer
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/467,838
Inventor
Saraswati Sukumar
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.)
School of Medicine of Johns Hopkins University
Original Assignee
School of Medicine of Johns Hopkins University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by School of Medicine of Johns Hopkins University filed Critical School of Medicine of Johns Hopkins University
Priority to US11/467,838 priority Critical patent/US20060292113A1/en
Publication of US20060292113A1 publication Critical patent/US20060292113A1/en
Assigned to NATIONAL INSTITUTES OF HEALTH - DIRECTOR DEITR reassignment NATIONAL INSTITUTES OF HEALTH - DIRECTOR DEITR CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: THE JOHNS HOPKINS UNIVERSITY
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/193Colony stimulating factors [CSF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/513Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim having oxo groups directly attached to the heterocyclic ring, e.g. cytosine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/52Purines, e.g. adenine
    • A61K31/522Purines, e.g. adenine having oxo groups directly attached to the heterocyclic ring, e.g. hypoxanthine, guanine, acyclovir
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/525Isoalloxazines, e.g. riboflavins, vitamin B2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/662Phosphorus acids or esters thereof having P—C bonds, e.g. foscarnet, trichlorfon
    • A61K31/663Compounds having two or more phosphorus acid groups or esters thereof, e.g. clodronic acid, pamidronic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • A61K31/7072Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid having two oxo groups directly attached to the pyrimidine ring, e.g. uridine, uridylic acid, thymidine, zidovudine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/76Viruses; Subviral particles; Bacteriophages
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/45Transferases (2)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10332Use of virus as therapeutic agent, other than vaccine, e.g. as cytolytic agent
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/24011Poxviridae
    • C12N2710/24111Orthopoxvirus, e.g. vaccinia virus, variola
    • C12N2710/24132Use of virus as therapeutic agent, other than vaccine, e.g. as cytolytic agent

Definitions

  • the present invention relates to the use of an epithelium-destroying agent to destroy the epithelium of an exocrine gland, particularly the mammary gland, in the prophylactic and therapeutic treatment of disease, in particular cancer.
  • Exocrine glands are glands that release a secretion external to or at the surface of an organ by means of a canal or duct.
  • exocrine glands include, among others, the mammary glands, prostate, liver, gall bladder, pancreas, kidneys, sweat glands, and salivary glands.
  • cancers of the breast and prostate are among the leading causes of death among women and men, respectively.
  • the mature human breast comprises from six to nine major ducts, which emanate from the nipple, serially branch into ducts and terminate in lobuloalveolar structures (Russo et al., Lab. Invest. 62(3):244-278 (1990)).
  • This branching network of ducts is composed of epithelial cells in a supporting matrix of connective tissue and endothelial cells.
  • Tissues removed from the human female breast during surgery and autopsy have been examined in numerous studies directed to the nature and site of origin of neoplastic growth. Subgross sampling and histological confirmation have enabled pathological characterization of entire breasts, leading to the postulation of the existence of four major possible sites of origin of mammary carcinomas, namely ducts, terminal ducts, ductules, and acini (Russo et al., supra). Ductal origin is supported by the presence of more extensive epithelial proliferations, which are presumed to be preneoplastic, in surgically removed cancerous breasts as compared to nonmalignant breasts removed during autopsies (Russo et al., supra).
  • an object of the present invention to provide a method of locally treating an exocrine gland prophylactically for disease. It is another object of the present invention to provide a method of locally treating an exocrine gland, in particular the mammary gland, prophylactically for cancer. Another object of the present invention is to provide a method of locally treating an exocrine gland therapeutically for disease. Yet another object of the present invention is to provide a method of locally treating a mammary gland therapeutically for cancer. Still yet another object of the present invention is to provide a method of locally treating a mammary gland both therapeutically and prophylactically for cancer.
  • the present invention provides prophylactic and therapeutic methods of treating the ductal epithelium of an exocrine gland, in particular the mammary gland, for disease, in particular cancer.
  • the method comprises contacting the ductal epithelium of the exocrine gland, in particular the mammary gland, with an epithelial-destroying agent.
  • the ductal epithelium is preferably contacted with the agent by ductal cannulation.
  • the epithelium-destroying agent is preferably a vector comprising a thymidine kinase gene, which is used in combination with ganciclovir (GCV), which can be systemically administered.
  • GCV ganciclovir
  • Another preferred epithelium-destroying agent is a vector comprising a hypoxanthine phosphoribosyl transferase (HPRT) gene, which is used in combination with hypoxanthine aminopterin thymidine (HAT) nucleotide, which can be systemically administered.
  • HPRT hypoxanthine phosphoribosyl transferase
  • HAT hypoxanthine aminopterin thymidine
  • a vector comprising a gene, which, upon transformation of a cell of the ductal epithelium and expression therein, induces apoptosis or death of the transformed cell.
  • a preferred apoptosis-inducing gene is bclxs.
  • Other preferred epithelial-destroying agents include a cytolytic virus, such as Vaccinia virus, and ethanol.
  • the preceding methods can additionally comprise contacting the ductal epithelium with a cytokine or hematopoietic growth factor, such as GM-CSF.
  • a method of treating the ductal epithelium of a mammary gland both therapeutically and prophylactically for cancer comprises treating the mammary gland therapeutically by surgery, radiation and/or chemotherapy and contacting the ductal epithelium of the mammary gland, either concomitantly or subsequently, with an epithelium-destroying agent, which does not specifically target cancerous cells.
  • the combined therapeutic/prophylactic method can additionally comprise contacting the ductal epithelium with a cytokine or hematopoietic growth factor, such as GM-CSF.
  • FIG. 1 is a graph of the ratio of cell no. in the presence of GCV over cell no. in the absence of GCV (Cell No.+GCV/Cell No. ⁇ GCV) versus viral multiplicity of infection (viral MOI), which shows the effect of the addition of 10 ⁇ g/ml GCV on NMU68 and RBA rat tumor cell lines 6 hrs after transduction with adenoviral-Herpes simplex thymidine kinase (AdHS-tk) at titers of 0, 100, 500 and 1,000 moi. After the cells were maintained in the presence of GCV for 3 days, they were counted using trypan blue exclusion as a measure of cell viability and cell numbers were normalized to the growth of cells in the absence of ganciclovir (GCV).
  • GCV ganciclovir
  • the present invention is based on the observation that the overwhelming majority of breast cancers arise from epithelial cells, particularly those epithelial cells which line the ducts of the mammary gland and are collectively referred to as the ductal epithelium.
  • the present invention is also based on the exocrine nature of the mammary gland. Given that the mammary gland is an exocrine gland, it was further observed that the central canal or duct could provide a means of directly accessing the ductal epithelium for localized prophylactic and therapeutic treatment of cancer. Based on these observations, the prophylactic and therapeutic methods of the present invention were developed.
  • the prophylactic method of the present invention is a method of treating the ductal epithelium of an exocrine gland prophylactically for a disease that affects the ductal epithelium of the exocrine gland.
  • the method comprises contacting, preferably by ductal cannulation, the ductal epithelium of the exocrine gland with an epithelium-destroying agent so as to destroy cells of the ductal epithelium affected by the disease.
  • the ductal epithelium of a mammary gland is treated prophylactically for cancer so as to inhibit the formation of cancer of ductal epithelial origin.
  • the method comprises contacting, preferably by ductal cannulation, the ductal epithelium of the mammary gland with an epithelium-destroying agent.
  • the agent preferably is a vector comprising a thymidine kinase gene, such as a Herpes simplex thymidine kinase gene, and ganciclovir, a vector comprising a HPRT gene and HAT nucleotide, a cytolytic virus, such as a Vaccinia virus, or ethanol.
  • the method can additionally comprise contacting the ductal epithelium with a cytokine or hematopoietic growth factor, such as GM-CSF.
  • the ductal epithelium of an exocrine gland such as a mammary gland
  • the method comprises contacting, preferably by ductal cannulation, the ductal epithelium of the exocrine gland with an epithelium-destroying agent to destroy less than all of the ductal epithelium so as to inhibit the formation of cancer of ductal epithelial origin.
  • an epithelium-destroying agent to destroy less than all of the ductal epithelium so as to inhibit the formation of cancer of ductal epithelial origin.
  • up to about 70%, 80%, 85%, 90% or 95% of the ductal epithelium is destroyed.
  • the epithelium-destroying agent is preferably a vector comprising a thymidine kinase gene, such as that from Herpes simplex, and ganciclovir, which can be brought into contact with the ductal epithelium by any suitable means, preferably by ductal-cannulation or by systemic administration, a vector comprising a HPRT gene and HAT nucleotide, which can be brought into contact with the ductal epithelium by any suitable means, preferably by ductal cannulation or by systemic administration, a vector comprising a gene, which upon transformation of a cell of the ductal epithelium and expression therein, induces apoptosis or death of the transformed cell, such as bclxs, ethanol, or a cytolytic virus, such as Vaccinia virus.
  • a thymidine kinase gene such as that from Herpes simplex, and ganciclovir
  • the above method can additionally comprise the administration of a cytokine or hematopoietic growth factor, such as GM-CSF.
  • GM-CSF can be brought into contact with the ductal epithelium of the mammary gland by any suitable means, such as by ductal cannulation of GM-CSF or a vector comprising a gene encoding GM-CSF, in which case the vector can be the same vector as the one encoding the thymidine kinase, HPRT or apoptosis-inducing gene, or it can be systemically administered.
  • This embodiment of the prophylactic method can be used to treat any exocrine gland. However, it is particularly useful in the treatment of the mammary gland.
  • the above-described prophylactic method of treating a mammary gland is particularly useful in treating a mammary gland in a mammal at risk for developing breast cancer.
  • the mammary gland can be characterized as one that has never had a tumor, one that had a tumor previously but the tumor is no longer detectable due to other prior therapeutic treatment, or one that has an incipient or occult tumor, preneoplasia or ductal hyperplasia. Normally, hyperplasias and incipient and occult tumors are not detectable by means of physical examination or radiography.
  • the prophylactic method will find use in cases where there is reason to take some prophylactic measures, such as when there are known inherited factors predisposing to cancers, where there are suspicious lesions present in a breast with the potential for developing into a malignancy, where there has been exposure to carcinogenic agents in the environment, where age predisposes to a cancer, where cancer of another gland, e.g., the mammary gland of the contralateral breast, suggests a propensity for developing cancer, or where there is a fear or suspicion of metastasis.
  • some prophylactic measures such as when there are known inherited factors predisposing to cancers, where there are suspicious lesions present in a breast with the potential for developing into a malignancy, where there has been exposure to carcinogenic agents in the environment, where age predisposes to a cancer, where cancer of another gland, e.g., the mammary gland of the contralateral breast, suggests a propensity for developing cancer, or where there is a fear or suspicion of metasta
  • the therapeutic method of the present invention is a method of treating the ductal epithelium of an exocrine gland therapeutically for a disease that affects the ductal epithelium of the exocrine gland.
  • the method comprises contacting, preferably by ductal cannulation, the ductal epithelium of the exocrine gland with an epithelium-destroying agent so as to destroy cells of the ductal epithelium affected by the disease.
  • the ductal epithelium of a mammary gland is locally treated therapeutically for cancer so as to destroy cancerous and noncancerous cells of the ductal epithelium and inhibit the spread of cancer.
  • the method comprises contacting, preferably by ductal cannulation, the ductal epithelium of the mammary gland with an epithelium-destroying agent, which need not, and preferably does not, specifically target cancerous cells.
  • the agent preferably is a vector comprising a thymidine kinase gene, such as a Herpes simplex thymidine kinase gene, and ganciclovir, a vector comprising a HPRT gene and HAT nucleotide, a cytolytic virus, such as a Vaccinia virus, or ethanol.
  • the method can additionally comprise contacting the ductal epithelium with a cytokine or hematopoietic growth factor, such as GM-CSF.
  • the epithelial-destroying agent should destroy all of the diseased or malignant epithelium.
  • the ductal epithelium immediately surrounding the diseased/malignant epithelium also preferably should be destroyed.
  • the present invention also provides a method of treating the ductal epithelium of a mammary gland both therapeutically and prophylactically for cancer.
  • the method comprises treating the mammary gland therapeutically with any given therapeutic method, such as those currently known and used in the art. Examples of such methods include surgical removal of the cancerous tissue, radiation therapy and chemotherapy.
  • the method further comprises contacting, either concomitantly with or subsequently to the therapeutic treatment, the ductal epithelium of the mammary gland, e.g., by ductal cannulation, with an epithelium-destroying agent, which preferably does not specifically target cancerous cells, so as to destroy any remaining cancerous cells and noncancerous cells and inhibit the spread of cancer.
  • the epithelium-destroying agent is preferably a vector comprising a thymidine kinase gene, such as a Herpes simplex thymidine kinase gene, combined with ganciclovir, a vector comprising a HPRT gene combined with HAT nucleotide, a cytolytic virus, such as a Vaccinia virus, or ethanol.
  • the method can additionally comprise contacting the ductal epithelium with a cytokine or hematopoietic growth factor, such as GM-CSF.
  • the methods of the present invention can be used to treat the ductal epithelium of any exocrine gland.
  • exocrine glands other than the mammary gland, which can be treated with the present inventive methods include, among others, the prostate, liver, gall bladder, pancreas, kidneys, sweat glands, and salivary glands. The methods are especially useful in the prophylactic and therapeutic treatment of the ductal epithelium of mammary glands.
  • the methods can be used to treat an exocrine gland for any disease that affects the exocrine ductal epithelium.
  • the methods are particularly useful in the treatment of cancer, including the stages of hyperplasia, adenoma, carcinoma in situ, and carcinoma, of ductal epithelial origin.
  • Any method can be used to destroy the ductal epithelium. It is preferred, however, that the destruction is limited to the ductal epithelium or a part thereof.
  • ductal cannulation is any method of contacting the ductal epithelium.
  • ductal cannulation is used.
  • any duct or lobule can be cannulated, it is preferred that the central canal or duct be cannulated.
  • Ductal cannulation also enables direct injection of a tumor mass, if desired.
  • Any epithelium-destroying agent can be used to destroy the ductal epithelium of an exocrine gland.
  • the agent preferably should not destroy cells other than cells of the ductal epithelium and preferably should not result in side effects, the adversity of which outweigh the benefits of destruction of the ductal epithelium.
  • the methods be used to destroy completely the ductal epithelium of an exocrine gland in the prophylactic/therapeutic treatment of a given disease, wherein the complete destruction of the ductal epithelium, in and of itself, would result in death of the mammal so treated.
  • cytotoxic agents and their prodrugs include genistein, okadaic acid, 1- ⁇ -D-arabinofuranosyl-cytosine, arabinofuranosyl-5-azacytosine, cisplatin, carboplatin, actinomycin D, asparaginase, bis-chloro-ethyl-nitroso-urea, bleomycin, chlorambucil, cyclohexyl-chloro-ethyl-nitroso-urea, cytosine arabinoside, daunomycin, etoposide, hydroxyurea, melphalan, mercaptopurine, mitomycin C, nitrogen mustard, procarbazine, teniposide, thioguanine, thiotepa, vincristine, 5-fluorouracil, 5-fluorocytosine, adriamycin, cyclophosphamide, methot
  • a cytolytic virus also can be used as an agent. Any cytolytic virus can be used as long as the organism mounts a rapid immunological response to it such that the virus cannot cause disease if it escapes the ductal epithelium.
  • cytolytic viruses include Vaccinia viruses and Sindbis viruses, which can also be used as vectors.
  • a Vaccinia virus is used. Due to lack of mucosal immunity, Vaccinia infectious particles enter and lyse the breast epithelial cells, yet stromal immunity destroys the particles as soon as they leave the ductile tree of the exocrine gland, thereby preventing cytolysis beyond the ductal epithelium.
  • the advantages of Vaccinia administration are that it eliminates the need for high titer virus, the need to induce cell division in the breast, and the need to administer a drug to effect cell death.
  • a vector comprising a suicide gene also can be used as an agent, in conjunction with an agent that destroys the ductal epithelium of an exocrine gland.
  • the vector comprising a suicide gene upon transformation of a cell of the ductal epithelium and expression therein, renders the transformed cell sensitive to the epithelium-destroying agent, increases the sensitivity of the transformed cell to the agent, converts the agent from a prodrug to an active drug, activates the conversion of the agent from a prodrug to an active drug, enhances the effect of the agent or, itself, produces a protein that is cytotoxic.
  • a preferred suicide gene for use in the present inventive methods is the one described above, i.e., a thymidine kinase, such as the one from Herpes simplex, which phosphorylates GCV, which, in turn, inhibits DNA replication.
  • a thymidine kinase such as the one from Herpes simplex
  • Another example of a suicide gene is cytosine deaminase, which is used in conjunction with 5-fluorocytosine. If the vector comprising the suicide gene is administered locally to the ducts, the cytotoxic agent or precursor can be administered systemically, since only transfected cells will be affected.
  • the bystander effect i.e., the death of neighboring uninfected cells, presumably due to transfer of toxic byproducts through gap junctions between cells in the same compartment, obviates the need for every cell in the ductal epithelium, which is to be destroyed, to be infected.
  • sufficient time must be allowed between contacting the ductal epithelium with the suicide gene and the prodrug, for example, to achieve efficient killing of the breast epithelial cells.
  • a vector comprising an apoptosis-inducing gene also can be used as an agent that destroys the ductal epithelium of an exocrine gland (Vaux, Cell 76:777-779 (1994)).
  • apoptosis-inducing genes include ced genes, myc genes (overexpressed), the bclxs gene, the bax gene, and the bak gene.
  • the apoptosis-inducing gene causes death of transfected cells, i.e., by inducing programmed cell death.
  • the bclxs gene, bax gene, or bak gene can be used to inhibit bcl-2 or bcl-x L , leading to apoptosis.
  • a vector comprising an apoptosis-inducing gene can be used in combination with an agent that inactivates apoptosis inhibitors such as bcl-z, p35, IAP, NAIP, DAD1 and A20 proteins.
  • Suicide and apopotosis genes can be administered by way of a viral vector, such as an adenoviral or retroviral vector.
  • a viral vector such as an adenoviral or retroviral vector.
  • Adenoviral vectors enable the generation of high titer recombinant viruses (10 11 /ml) and the efficient transduction of postmitotic cells because adenoviral DNA exists as an episome in the nucleus (Verma, Molecular Medicine 1:2-3 (1994)).
  • the gene can be under the transcriptional regulation of a Rous sarcoma viral promoter. Alternatively, it can be under the control of an epithelial tissue-specific or cell-specific promoter.
  • Uptake of recombinant virus can be facilitated by pretreatment or simultaneous treatment with polybrene or, for example, in the case of a retrovirus, attachment of the functional fragment of an antibody to the viral particle.
  • the apoptosis gene or suicide gene can be present in a recombinant microorganism, which will express the gene.
  • a recombinant microorganism which will express the gene.
  • One particularly preferred microorganism for this purpose is the bacterium Listeria monocytogenes.
  • liposomes complexes between polypeptide ligands for receptors on mammary ductal epithelial cells, including complexes of antibodies and functional fragments thereof, and plasmids can be used (Mulligan, Science 260:926-931 (1993)).
  • Epithelial cell-specific promoters such as whey acidic protein (wap)
  • wap whey acidic protein
  • Use can also be made of wild-type tumor suppressor genes, such as p53 or Mcs-1 (rat), homeobox genes expressed in normal cells but not in cancerous cells, and the maspin gene.
  • the ductal epithelium can be contacted with an agent to effect the scavenging of epithelial cells destroyed in accordance with the present invention, e.g., a cytokine/growth factor.
  • a cytokine/growth factor include GM-CSF, G-CSF, IL-2, IL-4, IL-6, IL-7, hCG, TNF- ⁇ , INF- ⁇ and INF- ⁇ .
  • Such factors can be contacted with the ductal epithelium directly or by expression of a vector comprising a gene encoding the factor, in which case the vector can be the same one that comprises a suicide gene, for example.
  • the factors stimulate a potent, long-lasting, and specific cell immunity, requiring both CD4 and CD8 cells.
  • the immune response is designed to scavenge destroyed ductal epithelial cells by generating autoimmunity towards epithelial cell antigens.
  • the ductal epithelium is preferably contacted with the agent by introduction of the agent through the central canal or duct of the exocrine ductal epithelium, such as by ductal cannulation.
  • the agent such as by ductal cannulation.
  • ductal cannulation enables intratumoral injection.
  • the methods of the present invention can be combined with other methods of prophylactic and therapeutic treatment in addition to those cited above, such as methods that target destruction of cancer cells, e.g., by targeting of cell-surface markers, receptor ligands, e.g., ligands to gastrin-releasing peptide-like receptors, tumor-associated antigens, e.g., the 57 kD cytokeratin or the antigen recognized by the monoclonal antibody GB24, the extracellular matrix glycoprotein tamascin, antisense oncogenes such as c-fos, homeobox genes that are expressed in cancer cells but not normal cells, tumor-infiltrating lymphocytes that express cytokines, RGD-containing peptides and proteins, which are administered following surgery, lipophilic drug-containing liposomes to which are covalently conjugated monoclonal antibodies for targeting to cancer cells, low fat diet, moderate physical exercise and hormonal modulation.
  • anti-testosterone agents can be used as well
  • This example demonstrates the successful delivery of virus and other agents into the mammary ductile tree by a single injection through the teat.
  • ADV/CMV- ⁇ -gal (from Dr. William Burns, Johns Hopkins University) is an adenoviral 5 vector constructed with a 13-galactosidase gene controlled by a cytomegaloviral promoter. It was delivered into the mammary gland by injection of a viral suspension in 20 ⁇ l of 0.2% trypan blue in Tris buffer through the teat of a rat. The nipple was extruded, and the sphincter removed by excising the nipple. In the rat, the muscle prevents fluid from regurgitating into the breast and had to be excised in order to visualize the ductal opening and administer the agent. Trypan blue was used as a tracking dye to ensure correct delivery to the ductile tree. Injection about 30 days postpartum resulted in the mammary epithelial tree being clearly visible. Ethyl alcohol (70%) was also successfully delivered by a single injection through the teat.
  • This example demonstrates that adenovirus can efficiently transduce human mammary epithelial cells in vitro.
  • ADV/CMV- ⁇ -gal was used to transduce HBLIOO mammary epithelial cells in vitro.
  • the mammary glands (6 on each side) of six virgin 50 day old Sprague Dawley rats were injected with AdHS-tk on the left side and trypan blue on the right side or left untreated.
  • One rat remained completely untreated with the virus or GCV and served as a positive control for NMU-induced tumorigenesis.
  • rats were given an intramuscular injection of 5 ⁇ g estradiol valerate and were anesthetized with an isofluorane/O 2 mixture.
  • the nipples were cannulated with a 33 gauge needle.
  • AdHS-tk diluted in trypan blue carrier (1 mM MgCl 2 , 20 ⁇ g/ml polybrene in 10% glycerol, 0.4% trypan blue in saline) at a concentration of 5 ⁇ 10 7 particles/ ⁇ l were injected into the duct.
  • Carrier control mammary glands received 20 ⁇ l of trypan blue carrier alone.
  • An animal was considered treated when at least three glands were successfully injected with trypan blue and another three glands were successfully injected with AdHS-tk. The remaining glands were left untreated. Twelve hours later, the rats were injected with 125 mg/kg body weight GCV twice daily for three days.
  • the rats were then given a second intramuscular injection of 5 ⁇ g estradiol valerate and intraperitoneal injections of 100 ⁇ g/kg body weight GCV once daily for three days.
  • the rats were given an intravenous injection of NMU dissolved in 0.05% acetic acid (Ash Stevens, Colo.; 50 mg NMU/kg body weight) and were subsequently monitored for general health and the appearance of tumors at weekly intervals for 8 months.
  • the results were as follows: No. of Total % of Glands Treatment No.
  • This example demonstrates the efficient transfection of mammary epithelial cells in vivo.
  • Lytic Vaccinia virus (10 6 Vaccinia-HA, which also carries lacZ, in 20 ⁇ l 0.2% trypan blue) was injected into the mammary glands through the teat of 45 day old virgin rats. Contralateral control glands were injected with 0.2% trypan blue. The glands were excised after 3 days. Frozen mammary gland sections were stained with X-gal and counterstained with eosin. When Vaccinia-HA was injected via the rat teat, it was able to infect the epithelial cells. At 3 days post-infection, the X-gal staining was confined primarily to the epithelial cells.
  • This example demonstrates the cytotoxicity of 20 Vaccinia/HA on HBL100 cells in vitro.
  • HBL100 cells (from ATCC, Rockville, Md.) were plated in DME:F12 medium (50% Dulbecco's Modified Eagles Medium: 50%Ham's F12 Supplement) containing 10% fetal bovine serum and 10 ⁇ g/ml insulin at a density of 5 ⁇ 10 4 cells/well and incubated at 37° C. overnight.
  • Vaccinia/HA at concentrations of 0 moi, 0.1 moi, or 1.0 moi, was added to the culture medium, and the cells were incubated at 37° C. for at least 3 days. More than 90% of the cells were dead within 72 hrs of infection at 0.1 moi.
  • This example shows the death of rat mammary tumor cells in culture by infection with Vaccinia/HA.
  • Vaccinia virus engineered to express ⁇ -galactosidase and hemagglutinin genes was added to the culture medium at concentrations of 0 moi, 0.1 moi, or 1.0 moi and incubated at 37° C. for at least 3 days. Up to 90% of the cells were lysed within 72 hours of injection at 1.0 moi.
  • This example demonstrates the destruction of mammary epithelium by transfection with Vaccinia/HA in vivo.
  • the mammary glands of 45-day old virgin rats were injected through the teat with 1 ⁇ 10 7 particles of Vaccinia/HA in 20 ⁇ l 0.2% trypan blue (tracking dye). Contralateral control glands were injected with 0.2% trypan blue.
  • the glands were excised after 3 days, fixed in chloroform:methanol:acetic acid and stained in iron-hematoxylin. Branching structures of a whole-mounted mammary gland injected with tracking dye alone were visible up to the end buds and alveoli. Also visible as brown bodies were the mammary lymph nodes. Examination of a whole-mounted mammary gland of the same rat receiving Vaccinia/HA in trypan blue on the contralateral side revealed that only about 30% of the ducts remained. In addition, the lymph nodes were considerably enlarged, denoting the mounting of an immune response to clear the Vaccinia from the vicinity.

Abstract

The present invention provides prophylactic and therapeutic methods of treating the ductal epithelium of an exocrine gland, in particular a mammary gland, for disease, in particular cancer. The methods comprise contacting the ductal epithelium of the exocrine gland with an epithelium destroying agent, preferably by ductal cannulation, so as to realize a prophylactic or therapeutic effect.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application is a Continuation of application Ser. No. 11/179,322, filed Jul. 12, 2005, which is a Divisional of application Ser. No. 09/971,901, filed Oct. 5, 2001 (now U.S. Pat. No. 6,932,777), which is a Divisional of application Ser. No. 09/240,206, filed Jan. 29, 1999 (now U.S. Pat. No. 6,330,472), which is a Continuation of application Ser. No. 09/093,145, filed Jun. 8, 1998 (now U.S. Pat. No. 6,153,184), which is a Divisional of application Ser. No. 08/692,001, filed Aug. 2, 1996 (now U.S. Pat. No. 5,763,415), which claims benefit of application No. 60/028,929, filed Aug. 3, 1995, the entire contents of which are incorporated herein by reference.
  • STATEMENT OF GOVERNMENT SUPPORT
  • The invention disclosed in this application was made with government support under NIH planning grant P20 CA/ES66205, “Gene-mediated prevention of Cancer,” and grant NIH 1RO1 CA 57993, “Genetic and hormonal factors in mammary carcinogenesis,” awarded by the National Institutes of Health, and with support of the American Cancer Society under grant RD 388, “Targeted disruption of breast cells: a novel strategy for cancer prevention.” Therefore, the government has certain rights in this invention.
  • TECHNICAL FIELD OF THE INVENTION
  • The present invention relates to the use of an epithelium-destroying agent to destroy the epithelium of an exocrine gland, particularly the mammary gland, in the prophylactic and therapeutic treatment of disease, in particular cancer.
  • BACKGROUND OF THE INVENTION
  • Exocrine glands are glands that release a secretion external to or at the surface of an organ by means of a canal or duct. Examples of exocrine glands include, among others, the mammary glands, prostate, liver, gall bladder, pancreas, kidneys, sweat glands, and salivary glands. Cancers of exocrine glands pose a major health problem, frequently resulting in death. Currently, cancers of the breast and prostate are among the leading causes of death among women and men, respectively.
  • The mature human breast comprises from six to nine major ducts, which emanate from the nipple, serially branch into ducts and terminate in lobuloalveolar structures (Russo et al., Lab. Invest. 62(3):244-278 (1990)). This branching network of ducts is composed of epithelial cells in a supporting matrix of connective tissue and endothelial cells.
  • Tissues removed from the human female breast during surgery and autopsy have been examined in numerous studies directed to the nature and site of origin of neoplastic growth. Subgross sampling and histological confirmation have enabled pathological characterization of entire breasts, leading to the postulation of the existence of four major possible sites of origin of mammary carcinomas, namely ducts, terminal ducts, ductules, and acini (Russo et al., supra). Ductal origin is supported by the presence of more extensive epithelial proliferations, which are presumed to be preneoplastic, in surgically removed cancerous breasts as compared to nonmalignant breasts removed during autopsies (Russo et al., supra).
  • With a cumulative lifetime risk of a woman developing breast cancer estimated to be 1 in 9, there is an urgent need to develop therapeutic methods of treatment that are more effective, less invasive and accompanied by fewer side effects and prophylactic methods of treatment that are more effective than increased and intensified physical monitoring and far less extreme than radical mastectomy. In spite of the recent discovery of the heritable breast cancer susceptibility loci, BRCA1 (Miki et al., Science 266:66-71 (1994)) and BRCA2, and other cancer susceptibility loci, and the increasing ability of physicians to identify women with elevated breast cancer risk, prophylactic methods are still currently limited to physical monitoring and prophylactic mastectomy.
  • In view of the above, it is an object of the present invention to provide a method of locally treating an exocrine gland prophylactically for disease. It is another object of the present invention to provide a method of locally treating an exocrine gland, in particular the mammary gland, prophylactically for cancer. Another object of the present invention is to provide a method of locally treating an exocrine gland therapeutically for disease. Yet another object of the present invention is to provide a method of locally treating a mammary gland therapeutically for cancer. Still yet another object of the present invention is to provide a method of locally treating a mammary gland both therapeutically and prophylactically for cancer. These and other objects and advantages, as well as additional inventive features, will become apparent from the detailed description provided herein.
  • SUMMARY OF THE INVENTION
  • The present invention provides prophylactic and therapeutic methods of treating the ductal epithelium of an exocrine gland, in particular the mammary gland, for disease, in particular cancer. The method comprises contacting the ductal epithelium of the exocrine gland, in particular the mammary gland, with an epithelial-destroying agent. The ductal epithelium is preferably contacted with the agent by ductal cannulation. The epithelium-destroying agent is preferably a vector comprising a thymidine kinase gene, which is used in combination with ganciclovir (GCV), which can be systemically administered. Another preferred epithelium-destroying agent is a vector comprising a hypoxanthine phosphoribosyl transferase (HPRT) gene, which is used in combination with hypoxanthine aminopterin thymidine (HAT) nucleotide, which can be systemically administered. Also preferred as an epithelium-destroying agent is a vector comprising a gene, which, upon transformation of a cell of the ductal epithelium and expression therein, induces apoptosis or death of the transformed cell. A preferred apoptosis-inducing gene is bclxs. Other preferred epithelial-destroying agents include a cytolytic virus, such as Vaccinia virus, and ethanol. The preceding methods can additionally comprise contacting the ductal epithelium with a cytokine or hematopoietic growth factor, such as GM-CSF. Also provided by the present invention is a method of treating the ductal epithelium of a mammary gland both therapeutically and prophylactically for cancer. The combined therapeutic/prophylactic method comprises treating the mammary gland therapeutically by surgery, radiation and/or chemotherapy and contacting the ductal epithelium of the mammary gland, either concomitantly or subsequently, with an epithelium-destroying agent, which does not specifically target cancerous cells. The combined therapeutic/prophylactic method can additionally comprise contacting the ductal epithelium with a cytokine or hematopoietic growth factor, such as GM-CSF.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 is a graph of the ratio of cell no. in the presence of GCV over cell no. in the absence of GCV (Cell No.+GCV/Cell No.−GCV) versus viral multiplicity of infection (viral MOI), which shows the effect of the addition of 10 μg/ml GCV on NMU68 and RBA rat tumor cell lines 6 hrs after transduction with adenoviral-Herpes simplex thymidine kinase (AdHS-tk) at titers of 0, 100, 500 and 1,000 moi. After the cells were maintained in the presence of GCV for 3 days, they were counted using trypan blue exclusion as a measure of cell viability and cell numbers were normalized to the growth of cells in the absence of ganciclovir (GCV).
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention is based on the observation that the overwhelming majority of breast cancers arise from epithelial cells, particularly those epithelial cells which line the ducts of the mammary gland and are collectively referred to as the ductal epithelium. The present invention is also based on the exocrine nature of the mammary gland. Given that the mammary gland is an exocrine gland, it was further observed that the central canal or duct could provide a means of directly accessing the ductal epithelium for localized prophylactic and therapeutic treatment of cancer. Based on these observations, the prophylactic and therapeutic methods of the present invention were developed.
  • Prophylactic Method
  • The prophylactic method of the present invention is a method of treating the ductal epithelium of an exocrine gland prophylactically for a disease that affects the ductal epithelium of the exocrine gland. The method comprises contacting, preferably by ductal cannulation, the ductal epithelium of the exocrine gland with an epithelium-destroying agent so as to destroy cells of the ductal epithelium affected by the disease.
  • In one embodiment of the prophylactic method of the present invention, the ductal epithelium of a mammary gland is treated prophylactically for cancer so as to inhibit the formation of cancer of ductal epithelial origin. The method comprises contacting, preferably by ductal cannulation, the ductal epithelium of the mammary gland with an epithelium-destroying agent. The agent preferably is a vector comprising a thymidine kinase gene, such as a Herpes simplex thymidine kinase gene, and ganciclovir, a vector comprising a HPRT gene and HAT nucleotide, a cytolytic virus, such as a Vaccinia virus, or ethanol. The method can additionally comprise contacting the ductal epithelium with a cytokine or hematopoietic growth factor, such as GM-CSF.
  • In another embodiment of the prophylactic method of the present invention, the ductal epithelium of an exocrine gland, such as a mammary gland, is treated prophylactically for cancer so as to inhibit the formation of cancer of ductal epithelial origin. The method comprises contacting, preferably by ductal cannulation, the ductal epithelium of the exocrine gland with an epithelium-destroying agent to destroy less than all of the ductal epithelium so as to inhibit the formation of cancer of ductal epithelial origin. Preferably, up to about 70%, 80%, 85%, 90% or 95% of the ductal epithelium is destroyed.
  • The epithelium-destroying agent is preferably a vector comprising a thymidine kinase gene, such as that from Herpes simplex, and ganciclovir, which can be brought into contact with the ductal epithelium by any suitable means, preferably by ductal-cannulation or by systemic administration, a vector comprising a HPRT gene and HAT nucleotide, which can be brought into contact with the ductal epithelium by any suitable means, preferably by ductal cannulation or by systemic administration, a vector comprising a gene, which upon transformation of a cell of the ductal epithelium and expression therein, induces apoptosis or death of the transformed cell, such as bclxs, ethanol, or a cytolytic virus, such as Vaccinia virus.
  • The above method can additionally comprise the administration of a cytokine or hematopoietic growth factor, such as GM-CSF. The GM-CSF can be brought into contact with the ductal epithelium of the mammary gland by any suitable means, such as by ductal cannulation of GM-CSF or a vector comprising a gene encoding GM-CSF, in which case the vector can be the same vector as the one encoding the thymidine kinase, HPRT or apoptosis-inducing gene, or it can be systemically administered.
  • This embodiment of the prophylactic method can be used to treat any exocrine gland. However, it is particularly useful in the treatment of the mammary gland.
  • The above-described prophylactic method of treating a mammary gland is particularly useful in treating a mammary gland in a mammal at risk for developing breast cancer. The mammary gland can be characterized as one that has never had a tumor, one that had a tumor previously but the tumor is no longer detectable due to other prior therapeutic treatment, or one that has an incipient or occult tumor, preneoplasia or ductal hyperplasia. Normally, hyperplasias and incipient and occult tumors are not detectable by means of physical examination or radiography. Accordingly, the prophylactic method will find use in cases where there is reason to take some prophylactic measures, such as when there are known inherited factors predisposing to cancers, where there are suspicious lesions present in a breast with the potential for developing into a malignancy, where there has been exposure to carcinogenic agents in the environment, where age predisposes to a cancer, where cancer of another gland, e.g., the mammary gland of the contralateral breast, suggests a propensity for developing cancer, or where there is a fear or suspicion of metastasis.
  • Therapeutic Method
  • The therapeutic method of the present invention is a method of treating the ductal epithelium of an exocrine gland therapeutically for a disease that affects the ductal epithelium of the exocrine gland. The method comprises contacting, preferably by ductal cannulation, the ductal epithelium of the exocrine gland with an epithelium-destroying agent so as to destroy cells of the ductal epithelium affected by the disease.
  • In one embodiment of the therapeutic method of the present invention, the ductal epithelium of a mammary gland is locally treated therapeutically for cancer so as to destroy cancerous and noncancerous cells of the ductal epithelium and inhibit the spread of cancer. The method comprises contacting, preferably by ductal cannulation, the ductal epithelium of the mammary gland with an epithelium-destroying agent, which need not, and preferably does not, specifically target cancerous cells. The agent preferably is a vector comprising a thymidine kinase gene, such as a Herpes simplex thymidine kinase gene, and ganciclovir, a vector comprising a HPRT gene and HAT nucleotide, a cytolytic virus, such as a Vaccinia virus, or ethanol. The method can additionally comprise contacting the ductal epithelium with a cytokine or hematopoietic growth factor, such as GM-CSF.
  • In the therapeutic method, the epithelial-destroying agent should destroy all of the diseased or malignant epithelium. In addition, the ductal epithelium immediately surrounding the diseased/malignant epithelium also preferably should be destroyed.
  • Combined Therapeutic/Prophylactic Method
  • The present invention also provides a method of treating the ductal epithelium of a mammary gland both therapeutically and prophylactically for cancer. The method comprises treating the mammary gland therapeutically with any given therapeutic method, such as those currently known and used in the art. Examples of such methods include surgical removal of the cancerous tissue, radiation therapy and chemotherapy. The method further comprises contacting, either concomitantly with or subsequently to the therapeutic treatment, the ductal epithelium of the mammary gland, e.g., by ductal cannulation, with an epithelium-destroying agent, which preferably does not specifically target cancerous cells, so as to destroy any remaining cancerous cells and noncancerous cells and inhibit the spread of cancer. The epithelium-destroying agent is preferably a vector comprising a thymidine kinase gene, such as a Herpes simplex thymidine kinase gene, combined with ganciclovir, a vector comprising a HPRT gene combined with HAT nucleotide, a cytolytic virus, such as a Vaccinia virus, or ethanol. The method can additionally comprise contacting the ductal epithelium with a cytokine or hematopoietic growth factor, such as GM-CSF.
  • Alternative Embodiments of Prophylactic & Therapeutic Methods
  • Although preferred embodiments have been described above, the methods of the present invention can be used to treat the ductal epithelium of any exocrine gland. Examples of exocrine glands, other than the mammary gland, which can be treated with the present inventive methods include, among others, the prostate, liver, gall bladder, pancreas, kidneys, sweat glands, and salivary glands. The methods are especially useful in the prophylactic and therapeutic treatment of the ductal epithelium of mammary glands.
  • Similarly, the methods can be used to treat an exocrine gland for any disease that affects the exocrine ductal epithelium. The methods are particularly useful in the treatment of cancer, including the stages of hyperplasia, adenoma, carcinoma in situ, and carcinoma, of ductal epithelial origin.
  • Any method can be used to destroy the ductal epithelium. It is preferred, however, that the destruction is limited to the ductal epithelium or a part thereof.
  • Any method of contacting the ductal epithelium can be used to effect local treatment. Preferably, ductal cannulation is used. Although any duct or lobule can be cannulated, it is preferred that the central canal or duct be cannulated. Ductal cannulation also enables direct injection of a tumor mass, if desired.
  • Any epithelium-destroying agent can be used to destroy the ductal epithelium of an exocrine gland. The agent preferably should not destroy cells other than cells of the ductal epithelium and preferably should not result in side effects, the adversity of which outweigh the benefits of destruction of the ductal epithelium. In no event should the methods be used to destroy completely the ductal epithelium of an exocrine gland in the prophylactic/therapeutic treatment of a given disease, wherein the complete destruction of the ductal epithelium, in and of itself, would result in death of the mammal so treated.
  • Examples of agents that can be used in the context of the prophylactic and therapeutic methods of the present invention include cytotoxic agents. Any cytotoxic agent known in the art and suitable for contacting the ductal epithelium of an exocrine gland of a mammal can be used. In addition to ethanol and GCV described above, other examples of cytotoxic agents and their prodrugs include genistein, okadaic acid, 1-β-D-arabinofuranosyl-cytosine, arabinofuranosyl-5-azacytosine, cisplatin, carboplatin, actinomycin D, asparaginase, bis-chloro-ethyl-nitroso-urea, bleomycin, chlorambucil, cyclohexyl-chloro-ethyl-nitroso-urea, cytosine arabinoside, daunomycin, etoposide, hydroxyurea, melphalan, mercaptopurine, mitomycin C, nitrogen mustard, procarbazine, teniposide, thioguanine, thiotepa, vincristine, 5-fluorouracil, 5-fluorocytosine, adriamycin, cyclophosphamide, methotrexate, vinblastine, doxorubicin, leucovorin, taxol, anti-estrogen agents such as tamoxifen, intracellular antibodies against oncogenes, the flavonol quercetin, Guan-mu-tong extract, retinoids such as fenretinide, nontoxic retinoid analogues such as N-(4-hydroxyphenyl)-retinamide (HPR), and monoterpenes such as limonene, perillyl alcohol and sobrerol. Preferably, the agent is locally administered, especially if administration of the agent is accompanied by toxic side effects. Otherwise, the agent can be administered by any suitable route, such as systemic administration.
  • A cytolytic virus also can be used as an agent. Any cytolytic virus can be used as long as the organism mounts a rapid immunological response to it such that the virus cannot cause disease if it escapes the ductal epithelium. Examples of cytolytic viruses include Vaccinia viruses and Sindbis viruses, which can also be used as vectors. Preferably, a Vaccinia virus is used. Due to lack of mucosal immunity, Vaccinia infectious particles enter and lyse the breast epithelial cells, yet stromal immunity destroys the particles as soon as they leave the ductile tree of the exocrine gland, thereby preventing cytolysis beyond the ductal epithelium. The advantages of Vaccinia administration are that it eliminates the need for high titer virus, the need to induce cell division in the breast, and the need to administer a drug to effect cell death.
  • A vector comprising a suicide gene also can be used as an agent, in conjunction with an agent that destroys the ductal epithelium of an exocrine gland. The vector comprising a suicide gene, upon transformation of a cell of the ductal epithelium and expression therein, renders the transformed cell sensitive to the epithelium-destroying agent, increases the sensitivity of the transformed cell to the agent, converts the agent from a prodrug to an active drug, activates the conversion of the agent from a prodrug to an active drug, enhances the effect of the agent or, itself, produces a protein that is cytotoxic. A preferred suicide gene for use in the present inventive methods is the one described above, i.e., a thymidine kinase, such as the one from Herpes simplex, which phosphorylates GCV, which, in turn, inhibits DNA replication. Another example of a suicide gene is cytosine deaminase, which is used in conjunction with 5-fluorocytosine. If the vector comprising the suicide gene is administered locally to the ducts, the cytotoxic agent or precursor can be administered systemically, since only transfected cells will be affected. In this regard, the bystander effect, i.e., the death of neighboring uninfected cells, presumably due to transfer of toxic byproducts through gap junctions between cells in the same compartment, obviates the need for every cell in the ductal epithelium, which is to be destroyed, to be infected. However, sufficient time must be allowed between contacting the ductal epithelium with the suicide gene and the prodrug, for example, to achieve efficient killing of the breast epithelial cells.
  • A vector comprising an apoptosis-inducing gene also can be used as an agent that destroys the ductal epithelium of an exocrine gland (Vaux, Cell 76:777-779 (1994)). Examples of apoptosis-inducing genes include ced genes, myc genes (overexpressed), the bclxs gene, the bax gene, and the bak gene. The apoptosis-inducing gene causes death of transfected cells, i.e., by inducing programmed cell death. For example, the bclxs gene, bax gene, or bak gene can be used to inhibit bcl-2 or bcl-xL, leading to apoptosis. Where necessary, a vector comprising an apoptosis-inducing gene can be used in combination with an agent that inactivates apoptosis inhibitors such as bcl-z, p35, IAP, NAIP, DAD1 and A20 proteins.
  • Suicide and apopotosis genes can be administered by way of a viral vector, such as an adenoviral or retroviral vector. Adenoviral vectors enable the generation of high titer recombinant viruses (1011/ml) and the efficient transduction of postmitotic cells because adenoviral DNA exists as an episome in the nucleus (Verma, Molecular Medicine 1:2-3 (1994)).
  • According to one preferred embodiment, the gene can be under the transcriptional regulation of a Rous sarcoma viral promoter. Alternatively, it can be under the control of an epithelial tissue-specific or cell-specific promoter.
  • Uptake of recombinant virus can be facilitated by pretreatment or simultaneous treatment with polybrene or, for example, in the case of a retrovirus, attachment of the functional fragment of an antibody to the viral particle.
  • In another embodiment, the apoptosis gene or suicide gene can be present in a recombinant microorganism, which will express the gene. One particularly preferred microorganism for this purpose is the bacterium Listeria monocytogenes.
  • Other methods known in the art for introduction of raw DNA into cells can be used in the methods of the present invention. Alternatively, liposomes, complexes between polypeptide ligands for receptors on mammary ductal epithelial cells, including complexes of antibodies and functional fragments thereof, and plasmids can be used (Mulligan, Science 260:926-931 (1993)). Epithelial cell-specific promoters, such as whey acidic protein (wap), can be used to target expression of a given gene, e.g., a suicide gene, in ductal epithelial cells. Use can also be made of wild-type tumor suppressor genes, such as p53 or Mcs-1 (rat), homeobox genes expressed in normal cells but not in cancerous cells, and the maspin gene.
  • Additionally, the ductal epithelium can be contacted with an agent to effect the scavenging of epithelial cells destroyed in accordance with the present invention, e.g., a cytokine/growth factor. Suitable cytokines/growth factors include GM-CSF, G-CSF, IL-2, IL-4, IL-6, IL-7, hCG, TNF-α, INF-α and INF-γ. Such factors can be contacted with the ductal epithelium directly or by expression of a vector comprising a gene encoding the factor, in which case the vector can be the same one that comprises a suicide gene, for example. The factors stimulate a potent, long-lasting, and specific cell immunity, requiring both CD4 and CD8 cells. The immune response is designed to scavenge destroyed ductal epithelial cells by generating autoimmunity towards epithelial cell antigens.
  • The ductal epithelium is preferably contacted with the agent by introduction of the agent through the central canal or duct of the exocrine ductal epithelium, such as by ductal cannulation. However, in the case of the mammary gland, for example, there are 6-9 major ducts that emanate from the nipple and serially branch into other ducts, terminating in lobulo-alveolar structures (Russo et al. (1990), supra). Accordingly, in some circumstances, such as those in which even more localized treatment is necessary or desired, for example, by the choice of anti-cancer agent, it may be preferable to contact the ductal epithelium of the exocrine gland through one of the other ducts or through a lobulo-alveolar structure as opposed to the central canal or duct. In this regard, ductal cannulation enables intratumoral injection.
  • The methods of the present invention can be combined with other methods of prophylactic and therapeutic treatment in addition to those cited above, such as methods that target destruction of cancer cells, e.g., by targeting of cell-surface markers, receptor ligands, e.g., ligands to gastrin-releasing peptide-like receptors, tumor-associated antigens, e.g., the 57 kD cytokeratin or the antigen recognized by the monoclonal antibody GB24, the extracellular matrix glycoprotein tamascin, antisense oncogenes such as c-fos, homeobox genes that are expressed in cancer cells but not normal cells, tumor-infiltrating lymphocytes that express cytokines, RGD-containing peptides and proteins, which are administered following surgery, lipophilic drug-containing liposomes to which are covalently conjugated monoclonal antibodies for targeting to cancer cells, low fat diet, moderate physical exercise and hormonal modulation. For prostate cancer, anti-testosterone agents can be used as well as an inhibitor of cell proliferation produced by prostatic stromal cells and CCAM, an epithelial cell adhesion molecule.
  • The following examples are presented to illustrate the present invention, not to limit its scope. The examples make use of the rat mammary tumor model, which has been deemed an appropriate experimental model for understanding breast cancer in humans (Sukumar et al., Mutation Res. 333 (1-2):37-44 (1995); Russo et al., supra). In fact, 90-100% of female rats develop mammary tumors in this model when they are administered the carcinogen NMU at 55 days of age (Sukumar, Cancer Cells 4:199-204 (1990)).
  • EXAMPLES Example 1
  • This example demonstrates the successful delivery of virus and other agents into the mammary ductile tree by a single injection through the teat.
  • ADV/CMV-β-gal (from Dr. William Burns, Johns Hopkins University) is an adenoviral 5 vector constructed with a 13-galactosidase gene controlled by a cytomegaloviral promoter. It was delivered into the mammary gland by injection of a viral suspension in 20 μl of 0.2% trypan blue in Tris buffer through the teat of a rat. The nipple was extruded, and the sphincter removed by excising the nipple. In the rat, the muscle prevents fluid from regurgitating into the breast and had to be excised in order to visualize the ductal opening and administer the agent. Trypan blue was used as a tracking dye to ensure correct delivery to the ductile tree. Injection about 30 days postpartum resulted in the mammary epithelial tree being clearly visible. Ethyl alcohol (70%) was also successfully delivered by a single injection through the teat.
  • Example 2
  • This example demonstrates that adenovirus can efficiently transduce human mammary epithelial cells in vitro.
  • ADV/CMV-β-gal was used to transduce HBLIOO mammary epithelial cells in vitro. The β-gal enzyme in this construct contains a nuclear localization signal and results in dense nuclear staining .HBL100 cells (102) were plated in 24-well plates, and transduced with virus at various doses and stained with X-gal 48 hrs later. Essentially all cells were infected at a moi=104.
  • This experiment also has been performed in human mammary tumor cells MCF-7 (American Type Culture Collection (ATCC), Rockville, Md.), human mammary epithelial cells MCF-10A (ATCC), and two rat mammary cancer cell lines, RBA (from Dr. Leonard Cohen) and 37-2 (from Dr. C. Marcelo Aldaz) with the same results. More efficient adenoviral constructs have been used, thereby achieving 100% infection at a moi=103. These experiments demonstrate successful infection by and expression of adenovirus carrying the lacZ indicator gene, which permits staining the cells blue by the expression of the enzyme β-galactosidase.
  • Example 3
  • This example demonstrates that infection with an AdHS-tk construct followed by GCV treatment effectively kills mammary tumor cells in vitro.
  • RBA and NMU68 are two rat mammary tumor cell lines derived from a DMBA- and a NMU-induced tumor, respectively [DMBA=dimethylbenz [a] anthracene, NMU=N′-nitro N′-methylurea]. Each cell line was plated at a density of 5×102 in 48-well plates (1.1 cm) and allowed to settle overnight. The next morning, they were transduced with AdHS-tk (Chen et al., PNAS (USA) 91:3054-57 (1994); obtained from S. Woo and E. Aguilar-Cordova, Baylor College of Medicine, Houston, Tex.) at titers of 0, 100, 500, and 1000 moi, and then, 6 hrs later, GCV (10 pg/ml) was added to the culture media. The cells were maintained in the presence of GCV for 3 days and then counted using trypan blue exclusion as a measure of cell viability. The cell numbers were normalized to the growth of cells in the absence of GCV. The results are shown in FIG. 1. More than 80% of the cells of each cell line were killed at a moi of 103.
  • Example 4
  • This example demonstrates the prophylactic effect of the method of the present invention.
  • The mammary glands (6 on each side) of six virgin 50 day old Sprague Dawley rats were injected with AdHS-tk on the left side and trypan blue on the right side or left untreated. One rat remained completely untreated with the virus or GCV and served as a positive control for NMU-induced tumorigenesis. On the day of surgery, rats were given an intramuscular injection of 5 μg estradiol valerate and were anesthetized with an isofluorane/O2 mixture. The nipples were cannulated with a 33 gauge needle. Twenty Al of AdHS-tk diluted in trypan blue carrier (1 mM MgCl2, 20 μg/ml polybrene in 10% glycerol, 0.4% trypan blue in saline) at a concentration of 5×107 particles/μl were injected into the duct. Carrier control mammary glands received 20 μl of trypan blue carrier alone. An animal was considered treated when at least three glands were successfully injected with trypan blue and another three glands were successfully injected with AdHS-tk. The remaining glands were left untreated. Twelve hours later, the rats were injected with 125 mg/kg body weight GCV twice daily for three days. The rats were then given a second intramuscular injection of 5 μg estradiol valerate and intraperitoneal injections of 100 μg/kg body weight GCV once daily for three days. Five to seven days after GCV treatment, the rats were given an intravenous injection of NMU dissolved in 0.05% acetic acid (Ash Stevens, Colo.; 50 mg NMU/kg body weight) and were subsequently monitored for general health and the appearance of tumors at weekly intervals for 8 months. The results were as follows:
    No. of Total % of Glands
    Treatment No. of Tumors Glands with Tumor
    Untreated 5 12 41.7
    Control
    Trypan Blue and 5 17 29.4
    Ganciclovir
    No injection; 5 20 25.0
    Ganciclovir
    AdHS-tk and  2* 35 5.7
    Ganciclovir

    *The two tumors (size < 5 mm) in this group were detected during necropsy at the termination of the experiment 8 months later. The difference between tumors appearing in treated versus control glands was significant by Chi2 analysis (p < 0.01).
  • The above results show that the method of the present invention inhibits the formation of cancer of ductal epithelial origin in this rat model, in which NMU induces the formation of mammary tumors in 90-100% of female rats of similar age. Surprisingly and unexpectedly, this prophylactic effect was achieved without extensive destruction of the mammary glands. These data demonstrate that selective destruction of epithelial cells, e.g., key stem cells, can be sufficient to provide prophylactic protection against carcinogen-induced tumor formation in the ductal epithelium of the mammary gland.
  • Example 5
  • This example demonstrates the efficient transfection of mammary epithelial cells in vivo.
  • Lytic Vaccinia virus (106 Vaccinia-HA, which also carries lacZ, in 20 μl 0.2% trypan blue) was injected into the mammary glands through the teat of 45 day old virgin rats. Contralateral control glands were injected with 0.2% trypan blue. The glands were excised after 3 days. Frozen mammary gland sections were stained with X-gal and counterstained with eosin. When Vaccinia-HA was injected via the rat teat, it was able to infect the epithelial cells. At 3 days post-infection, the X-gal staining was confined primarily to the epithelial cells.
  • Example 6
  • This example demonstrates the cytotoxicity of 20 Vaccinia/HA on HBL100 cells in vitro.
  • HBL100 cells (from ATCC, Rockville, Md.) were plated in DME:F12 medium (50% Dulbecco's Modified Eagles Medium: 50%Ham's F12 Supplement) containing 10% fetal bovine serum and 10 μg/ml insulin at a density of 5×104 cells/well and incubated at 37° C. overnight. Vaccinia/HA, at concentrations of 0 moi, 0.1 moi, or 1.0 moi, was added to the culture medium, and the cells were incubated at 37° C. for at least 3 days. More than 90% of the cells were dead within 72 hrs of infection at 0.1 moi.
  • Example 7
  • This example shows the death of rat mammary tumor cells in culture by infection with Vaccinia/HA.
  • Cells of rat mammary cancer cell line RBA were plated in growth medium at a density of 5×104 cells/well and incubated at 37° C. overnight. Vaccinia virus engineered to express β-galactosidase and hemagglutinin genes (Vaccinia/HA) was added to the culture medium at concentrations of 0 moi, 0.1 moi, or 1.0 moi and incubated at 37° C. for at least 3 days. Up to 90% of the cells were lysed within 72 hours of injection at 1.0 moi.
  • Example 8
  • This example demonstrates the destruction of mammary epithelium by transfection with Vaccinia/HA in vivo.
  • The mammary glands of 45-day old virgin rats were injected through the teat with 1×107 particles of Vaccinia/HA in 20 μl 0.2% trypan blue (tracking dye). Contralateral control glands were injected with 0.2% trypan blue. The glands were excised after 3 days, fixed in chloroform:methanol:acetic acid and stained in iron-hematoxylin. Branching structures of a whole-mounted mammary gland injected with tracking dye alone were visible up to the end buds and alveoli. Also visible as brown bodies were the mammary lymph nodes. Examination of a whole-mounted mammary gland of the same rat receiving Vaccinia/HA in trypan blue on the contralateral side revealed that only about 30% of the ducts remained. In addition, the lymph nodes were considerably enlarged, denoting the mounting of an immune response to clear the Vaccinia from the vicinity.
  • All publications, patents, and patent applications cited herein are hereby incorporated by reference to the same extent as if each individual document were individually and specifically indicated to be incorporated by reference and were set forth in its entirety herein.
  • While this invention has been described with emphasis upon preferred embodiments, it will be obvious to those of ordinary skill in the art that the preferred embodiments may be varied. It is intended that the invention may be practiced otherwise than as specifically described herein. Accordingly, this invention includes all modifications encompassed within the spirit and scope of the appended claims.

Claims (5)

1. A method for treating or preventing breast cancer, which method comprises cannulating a duct of a mammary gland in need of therapeutic or prophylactic treatment for breast cancer and locally administering into the duct an epithelium-destroying agent in an amount sufficient to destroy at least a portion of the ductal epithelium, whereupon breast cancer is treated or prevented.
2. The method of claim 1, wherein the epithelium-destroying agent selectively destroys cells of the ductal epithelium.
3. The method of claim 1, wherein the portion of the ductal epithelium that is destroyed is up to about 70%.
4. The method of claim 1, wherein the duct is cannulated through a nipple.
5. A method of treating or preventing breast cancer, which method comprises identifying a duct in a nipple of a mammary gland in need of therapeutic or prophylactic treatment, directly accessing cells of the ductal epithelium of the duct by cannulating the duct through the nipple, and locally administering into the duct an epithelium-destroying agent in an amount sufficient to destroy selectively at least a portion of the ductal epithelium substantially without destroying other cells of the mammary gland, whereupon breast cancer is treated or prevented.
US11/467,838 1995-08-03 2006-08-28 Prophylactic and therapeutic treatment of the ductal epithelium of a mammary gland for cancer Abandoned US20060292113A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/467,838 US20060292113A1 (en) 1995-08-03 2006-08-28 Prophylactic and therapeutic treatment of the ductal epithelium of a mammary gland for cancer

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US2892995P 1995-08-03 1995-08-03
US08/692,001 US5763415A (en) 1995-08-03 1996-08-02 Destruction of the epithelium of an exocrine gland in the prophylactic and therapeutic treatment of cancer
US09/093,145 US6153184A (en) 1995-08-03 1998-06-08 Destruction of the epithelium of an exocrine gland in the prophylactic and therapeutic treatment of cancer
US09/240,206 US6330472B2 (en) 1995-08-03 1999-01-29 Prophylactic and therapeutic treatment of the ductal epithelium for a mammary gland for cancer
US9971901A 2001-10-05 2001-10-05
US11/179,322 US7196070B2 (en) 1995-08-03 2005-07-12 Prophylactic and therapeutic treatment of the ductal epithelium of a mammary gland for cancer
US11/467,838 US20060292113A1 (en) 1995-08-03 2006-08-28 Prophylactic and therapeutic treatment of the ductal epithelium of a mammary gland for cancer

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/179,322 Continuation US7196070B2 (en) 1995-08-03 2005-07-12 Prophylactic and therapeutic treatment of the ductal epithelium of a mammary gland for cancer

Publications (1)

Publication Number Publication Date
US20060292113A1 true US20060292113A1 (en) 2006-12-28

Family

ID=26704265

Family Applications (8)

Application Number Title Priority Date Filing Date
US08/692,001 Expired - Lifetime US5763415A (en) 1995-08-03 1996-08-02 Destruction of the epithelium of an exocrine gland in the prophylactic and therapeutic treatment of cancer
US09/093,145 Expired - Lifetime US6153184A (en) 1995-08-03 1998-06-08 Destruction of the epithelium of an exocrine gland in the prophylactic and therapeutic treatment of cancer
US09/240,206 Expired - Lifetime US6330472B2 (en) 1995-08-03 1999-01-29 Prophylactic and therapeutic treatment of the ductal epithelium for a mammary gland for cancer
US09/634,483 Expired - Lifetime US6559130B1 (en) 1995-08-03 2000-08-08 Destruction of the epithelium of an exocrine gland in the prophylactic and therapeutic treatment of cancer
US09/746,588 Expired - Lifetime US6656918B2 (en) 1995-08-03 2000-12-21 Prophylactic and therapeutic treatment of the ductal epithelium of a mammary gland for cancer
US09/971,901 Expired - Fee Related US6932777B2 (en) 1995-08-03 2001-10-05 Prophylactic and therapeutic treatment of the ductal epithelium of a mammary gland for cancer
US11/179,322 Expired - Fee Related US7196070B2 (en) 1995-08-03 2005-07-12 Prophylactic and therapeutic treatment of the ductal epithelium of a mammary gland for cancer
US11/467,838 Abandoned US20060292113A1 (en) 1995-08-03 2006-08-28 Prophylactic and therapeutic treatment of the ductal epithelium of a mammary gland for cancer

Family Applications Before (7)

Application Number Title Priority Date Filing Date
US08/692,001 Expired - Lifetime US5763415A (en) 1995-08-03 1996-08-02 Destruction of the epithelium of an exocrine gland in the prophylactic and therapeutic treatment of cancer
US09/093,145 Expired - Lifetime US6153184A (en) 1995-08-03 1998-06-08 Destruction of the epithelium of an exocrine gland in the prophylactic and therapeutic treatment of cancer
US09/240,206 Expired - Lifetime US6330472B2 (en) 1995-08-03 1999-01-29 Prophylactic and therapeutic treatment of the ductal epithelium for a mammary gland for cancer
US09/634,483 Expired - Lifetime US6559130B1 (en) 1995-08-03 2000-08-08 Destruction of the epithelium of an exocrine gland in the prophylactic and therapeutic treatment of cancer
US09/746,588 Expired - Lifetime US6656918B2 (en) 1995-08-03 2000-12-21 Prophylactic and therapeutic treatment of the ductal epithelium of a mammary gland for cancer
US09/971,901 Expired - Fee Related US6932777B2 (en) 1995-08-03 2001-10-05 Prophylactic and therapeutic treatment of the ductal epithelium of a mammary gland for cancer
US11/179,322 Expired - Fee Related US7196070B2 (en) 1995-08-03 2005-07-12 Prophylactic and therapeutic treatment of the ductal epithelium of a mammary gland for cancer

Country Status (1)

Country Link
US (8) US5763415A (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070189968A1 (en) * 1999-06-11 2007-08-16 Annette Bianchi Gel composition for filling a breast milk duct prior to surgical excision of the duct or other breast tissue

Families Citing this family (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6168779B1 (en) * 1997-09-16 2001-01-02 The Regents Of The University Of California Methods and kits for identifying ductal orifices
US6890311B2 (en) 1997-09-16 2005-05-10 The Regents Of The University Of California Methods for performing medical procedures within a breast duct
US20040092602A1 (en) * 1998-05-07 2004-05-13 Steiner Mitchell S. Method for treatment and chemoprevention of prostate cancer
US20040176470A1 (en) * 1998-05-07 2004-09-09 Steiner Mitchell S. Method for treatment and chemoprevention of prostate cancer
US8197430B1 (en) * 1998-05-22 2012-06-12 Biopheresis Technologies, Inc. Method and system to remove cytokine inhibitor in patients
US6620382B1 (en) 1998-05-22 2003-09-16 Biopheresis Technologies, Llc. Method and compositions for treatment of cancers
US6391026B1 (en) 1998-09-18 2002-05-21 Pro Duct Health, Inc. Methods and systems for treating breast tissue
JP2002526422A (en) * 1998-10-02 2002-08-20 ウィンディー ヒル テクノロジー インコーポレイテッド Methods for identifying, diagnosing, and treating breast cancer
US6413228B1 (en) * 1998-12-28 2002-07-02 Pro Duct Health, Inc. Devices, methods and systems for collecting material from a breast duct
IL143988A0 (en) * 1999-01-20 2002-04-21 Univ California Compositions and methods for intraductal gene therapy
WO2000043548A1 (en) * 1999-01-21 2000-07-27 Board Of Regents Of The University Of Nebraska Therapeutic and diagnostic applications of prostatic acid phosphatase in prostate cancer
US6610484B1 (en) 1999-01-26 2003-08-26 Cytyc Health Corporation Identifying material from a breast duct
US6638727B1 (en) * 1999-01-26 2003-10-28 Cytyc Health Corporation Methods for identifying treating or monitoring asymptomatic patients for risk reduction or therapeutic treatment of breast cancer
JP2002541855A (en) * 1999-04-21 2002-12-10 ジェンザイム・コーポレーション Adenovirus vectors containing nucleic acids encoding immunomodulatory molecules
US6642009B2 (en) * 1999-05-17 2003-11-04 Cytyc Health Corporation Isolated ductal fluid sample
WO2002007604A1 (en) 2000-07-25 2002-01-31 David Hung Method and device for obtaining prostatic material
EP1785727A3 (en) * 2000-08-08 2007-08-29 Cytyc Corporation Identification of viral agents in breast ducts and antiviral therapy therefore
EP1307746B1 (en) * 2000-08-08 2007-04-18 Cytyc Corporation Identification of viral agents in breast ducts and antiviral therapy therefore
WO2002072762A2 (en) * 2001-03-08 2002-09-19 Advanced Cell Technology, Inc. Use of rna interference for the creation of lineage specific es and other undifferentiated cells and production of differentiated cells in vitro by co-culture
US20020142941A1 (en) * 2001-03-30 2002-10-03 Pro Duct Health, Inc. Intraductal treatment targeting methylated promoters in breast cancer
US6680342B2 (en) * 2001-09-20 2004-01-20 Mayo Foundation For Medical Education And Research Methods and compositions for inhibiting the proliferation of prostate cancer cells
US7157499B2 (en) * 2001-10-30 2007-01-02 Zaycor Industries Corp. Medical application of oxidized monoterpenes
US20080249183A1 (en) * 2001-11-29 2008-10-09 Steiner Mitchell S Treatment of androgen-deprivation induced osteoporosis
AU2002360670A1 (en) * 2001-12-19 2003-07-09 Wilk Patent Development Corporation Method and related composition employing nanostructures
CA2477828A1 (en) * 2002-03-15 2003-09-25 Cytyc Health Corporation Method of diagnosis and treatment of breast lesions
CA2565215C (en) * 2004-04-30 2014-04-15 Biopheresis Technologies, Inc. Method and system to remove soluble tnfr1, tnfr2, and il2 in patients
US7250550B2 (en) * 2004-10-22 2007-07-31 Wright Medical Technology, Inc. Synthetic bone substitute material
US20060270641A1 (en) * 2005-05-31 2006-11-30 Steiner Mitchell S Method for chemoprevention of prostate cancer
TW200819540A (en) 2006-07-11 2008-05-01 Genelux Corp Methods and compositions for detection of microorganisms and cells and treatment of diseases and disorders
WO2008011726A1 (en) * 2006-07-27 2008-01-31 Ottawa Health Research Institute Staged immune-response modulation in oncolytic therapy
US20090098529A1 (en) 2006-10-16 2009-04-16 Nanhai Chen Methods for attenuating virus strains for diagnostic and therapeutic uses

Citations (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4568673A (en) * 1984-03-20 1986-02-04 Wayne State University Compositions inhibiting murine MXT ductal carcinoma
US4623660A (en) * 1977-08-22 1986-11-18 Imperial Chemical Industries Plc Triphenylalkene derivatives
US4939240A (en) * 1983-03-04 1990-07-03 Health Research, Inc. Monoclonal antibodies to human breast carcinoma cells and their use in diagnosis and therapy
US5149527A (en) * 1990-09-18 1992-09-22 Oncotech, Inc. Immunopotentiating protocol for chemotherapy-responsive tumors
US5215904A (en) * 1989-01-27 1993-06-01 Wisconsin Alumni Research Foundation Method for producing a recombinant mammal in vivo
US5236844A (en) * 1990-11-21 1993-08-17 Institut National De La Sante Et De La Recherche Medicale Analytical markers for malignant breast cancer
US5304489A (en) * 1987-02-17 1994-04-19 Genpharm International, Inc. DNA sequences to target proteins to the mammary gland for efficient secretion
US5383849A (en) * 1992-05-11 1995-01-24 Wilson-Cook Medical, Inc. Method for selective endoscopic cannulation of ductal structures
US5437863A (en) * 1992-09-02 1995-08-01 Genetics Institute, Inc. Method of enhancing the growth of gut epithelial cells by administration of a cytokine such as interleukin II
US5472441A (en) * 1993-11-08 1995-12-05 Zomed International Device for treating cancer and non-malignant tumors and methods
US5478556A (en) * 1994-02-28 1995-12-26 Elliott; Robert L. Vaccination of cancer patients using tumor-associated antigens mixed with interleukin-2 and granulocyte-macrophage colony stimulating factor
US5585096A (en) * 1994-06-23 1996-12-17 Georgetown University Replication-competent herpes simplex virus mediates destruction of neoplastic cells
US5651964A (en) * 1990-12-04 1997-07-29 Board Of Regents, The University Of Texas System Methods for the suppression of neu mediated tumors by the adenoviral EIA gene
US5662896A (en) * 1988-03-21 1997-09-02 Chiron Viagene, Inc. Compositions and methods for cancer immunotherapy
US5668122A (en) * 1993-07-28 1997-09-16 Fife; Rose S. Method to treat cancer with tetracyclines
US5677292A (en) * 1994-11-17 1997-10-14 Duquesne University Of The Holy Ghost Derivatives of estra 1,3,5(10)triene-17-one,3-amino compounds and their use
US5744133A (en) * 1986-08-13 1998-04-28 Transgene S.A. Expression of a tumor-specific antigen by a recombinant vector virus and use thereof in preventitive or curative treatment of the corresponding tumor
US5747469A (en) * 1991-03-06 1998-05-05 Board Of Regents, The University Of Texas System Methods and compositions comprising DNA damaging agents and p53
US5780009A (en) * 1995-01-20 1998-07-14 Nexia Biotechnologies, Inc. Direct gene transfer into the ruminant mammary gland
US5833975A (en) * 1989-03-08 1998-11-10 Virogenetics Corporation Canarypox virus expressing cytokine and/or tumor-associated antigen DNA sequence
US5837693A (en) * 1995-03-24 1998-11-17 The Regents Of The University Of California Intravenous hormone polypeptide delivery by salivary gland expression
US5858365A (en) * 1993-10-29 1999-01-12 Trustees Of Boston University Methods for the treatment of wounds using butyric acid salts and derivatives
US5866550A (en) * 1992-10-13 1999-02-02 La Jolla Cancer Research Foundation Method of inhibiting replication of hyperproliferative cells using a nucleic acid encoding EIA
US6096874A (en) * 1990-10-01 2000-08-01 Board Of Regents, The University Of Texas System High affinity tamoxifen derivatives

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SE422885B (en) * 1980-04-11 1982-04-05 Ursus Konsult Ab The electrode device
DE3483054D1 (en) * 1983-03-04 1990-10-04 Health Research Inc MONOCLONAL ANTIBODIES AGAINST HUMAN BREAST CANCER CELLS AND THEIR USE IN DIAGNOSIS AND THERAPY.
US5849298A (en) * 1987-06-24 1998-12-15 Autoimmune Inc. Treatment of multiple sclerosis by oral administration of bovine myelin
US5118611A (en) * 1988-07-25 1992-06-02 Adeza Biomedical Corporation Adenocarcinoma antigen binding methods and reagents
CA2113990A1 (en) * 1991-07-26 1993-02-18 Frederick L. Moolten Cancer therapy utilizing malignant cells
PT698109E (en) * 1993-05-10 2002-12-31 Univ Michigan TRANSFER OF GENES INTO PANCREATIC EPITHELIAL CELLS
US5583104A (en) * 1993-12-06 1996-12-10 Mayo Foundation For Medical Research And Education Treatment of proliferation of bile duct epithelium

Patent Citations (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4623660A (en) * 1977-08-22 1986-11-18 Imperial Chemical Industries Plc Triphenylalkene derivatives
US4939240A (en) * 1983-03-04 1990-07-03 Health Research, Inc. Monoclonal antibodies to human breast carcinoma cells and their use in diagnosis and therapy
US4568673A (en) * 1984-03-20 1986-02-04 Wayne State University Compositions inhibiting murine MXT ductal carcinoma
US5744133A (en) * 1986-08-13 1998-04-28 Transgene S.A. Expression of a tumor-specific antigen by a recombinant vector virus and use thereof in preventitive or curative treatment of the corresponding tumor
US5304489A (en) * 1987-02-17 1994-04-19 Genpharm International, Inc. DNA sequences to target proteins to the mammary gland for efficient secretion
US5662896A (en) * 1988-03-21 1997-09-02 Chiron Viagene, Inc. Compositions and methods for cancer immunotherapy
US5215904A (en) * 1989-01-27 1993-06-01 Wisconsin Alumni Research Foundation Method for producing a recombinant mammal in vivo
US5833975A (en) * 1989-03-08 1998-11-10 Virogenetics Corporation Canarypox virus expressing cytokine and/or tumor-associated antigen DNA sequence
US5149527A (en) * 1990-09-18 1992-09-22 Oncotech, Inc. Immunopotentiating protocol for chemotherapy-responsive tumors
US6096874A (en) * 1990-10-01 2000-08-01 Board Of Regents, The University Of Texas System High affinity tamoxifen derivatives
US5236844A (en) * 1990-11-21 1993-08-17 Institut National De La Sante Et De La Recherche Medicale Analytical markers for malignant breast cancer
US5651964A (en) * 1990-12-04 1997-07-29 Board Of Regents, The University Of Texas System Methods for the suppression of neu mediated tumors by the adenoviral EIA gene
US5747469A (en) * 1991-03-06 1998-05-05 Board Of Regents, The University Of Texas System Methods and compositions comprising DNA damaging agents and p53
US5383849A (en) * 1992-05-11 1995-01-24 Wilson-Cook Medical, Inc. Method for selective endoscopic cannulation of ductal structures
US5437863A (en) * 1992-09-02 1995-08-01 Genetics Institute, Inc. Method of enhancing the growth of gut epithelial cells by administration of a cytokine such as interleukin II
US5866550A (en) * 1992-10-13 1999-02-02 La Jolla Cancer Research Foundation Method of inhibiting replication of hyperproliferative cells using a nucleic acid encoding EIA
US5668122A (en) * 1993-07-28 1997-09-16 Fife; Rose S. Method to treat cancer with tetracyclines
US5858365A (en) * 1993-10-29 1999-01-12 Trustees Of Boston University Methods for the treatment of wounds using butyric acid salts and derivatives
US5472441A (en) * 1993-11-08 1995-12-05 Zomed International Device for treating cancer and non-malignant tumors and methods
US5478556A (en) * 1994-02-28 1995-12-26 Elliott; Robert L. Vaccination of cancer patients using tumor-associated antigens mixed with interleukin-2 and granulocyte-macrophage colony stimulating factor
US5585096A (en) * 1994-06-23 1996-12-17 Georgetown University Replication-competent herpes simplex virus mediates destruction of neoplastic cells
US5677292A (en) * 1994-11-17 1997-10-14 Duquesne University Of The Holy Ghost Derivatives of estra 1,3,5(10)triene-17-one,3-amino compounds and their use
US5780009A (en) * 1995-01-20 1998-07-14 Nexia Biotechnologies, Inc. Direct gene transfer into the ruminant mammary gland
US5837693A (en) * 1995-03-24 1998-11-17 The Regents Of The University Of California Intravenous hormone polypeptide delivery by salivary gland expression

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Stearns et al., Preclinical and Clinical Evaluation of Intraductally Administered Agents in Early Breast Cancer Sci Transl Med 3, 106ra108 (2011) *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070189968A1 (en) * 1999-06-11 2007-08-16 Annette Bianchi Gel composition for filling a breast milk duct prior to surgical excision of the duct or other breast tissue
US20110200695A1 (en) * 1999-06-11 2011-08-18 Annette Bianchi Gel composition for filling a breast milk duct prior to surgical excision of the duct or other breast tissue

Also Published As

Publication number Publication date
US6153184A (en) 2000-11-28
US6656918B2 (en) 2003-12-02
US20050281780A1 (en) 2005-12-22
US20020035356A1 (en) 2002-03-21
US20010005505A1 (en) 2001-06-28
US20010021701A1 (en) 2001-09-13
US6932777B2 (en) 2005-08-23
US5763415A (en) 1998-06-09
US7196070B2 (en) 2007-03-27
US6330472B2 (en) 2001-12-11
US6559130B1 (en) 2003-05-06

Similar Documents

Publication Publication Date Title
US7196070B2 (en) Prophylactic and therapeutic treatment of the ductal epithelium of a mammary gland for cancer
Wakimoto et al. Human glioblastoma–derived cancer stem cells: Establishment of invasive glioma models and treatment with oncolytic herpes simplex virus vectors
Thorne et al. Rational strain selection and engineering creates a broad-spectrum, systemically effective oncolytic poxvirus, JX-963
Zeyaullah et al. Oncolytic viruses in the treatment of cancer: a review of current strategies
Short et al. Gene delivery to glioma cells in rat brain by grafting of a retrovirus packaging cell line
WO2018124835A1 (en) Novel exosome-based anticancer agent
Bouvet et al. Suppression of the immune response to an adenovirus vector and enhancement of intratumoral transgene expression by low-dose etoposide
JPH09504784A (en) Gene therapy for solid tumors, papillomas and warts
US20050095270A1 (en) Drug delivery to the inner ear and methods of using same
O’Leary et al. Novel oncolytic chimeric orthopoxvirus causes regression of pancreatic cancer xenografts and exhibits abscopal effect at a single low dose
Seubert et al. Enhanced tumor therapy using vaccinia virus strain GLV-1h68 in combination with a β-galactosidase-activatable prodrug seco-analog of duocarmycin SA
CA2323067A1 (en) Producer cells for replication selective viruses in the treatment of malignancy
Beljanski et al. The use of oncolytic viruses to overcome lung cancer drug resistance
WO1997005898A1 (en) Delivery of an agent to the ductal epithelium in the prophylactic and therapeutic treatment of cancer
Rosenfeld et al. Pancreatic carcinoma cell killing via adenoviral mediated delivery of the herpes simplex virus thymidine kinase gene
Li et al. Efficacy of multiple administrations of a recombinant adenovirus expressing wild-type p53 in an immune-competent mouse tumor model
Zhuang et al. Allelotype analysis of 2′, 3′-dideoxycytidine-and 1, 3-butadiene-induced lymphomas in B6C3F1 mice
Hirschowitz et al. Adenovirus-mediated expression of melanoma antigen gp75 as immunotherapy for metastatic melanoma
Shibata et al. Massive apoptotic cell death in chemically induced rat urinary bladder carcinomas following in situ HSVtk electrogene transfer
Behbakht et al. Adenovirus-mediated gene therapy of ovarian cancer in a mouse model
US20210402010A1 (en) A COMBINATION THERAPY FOR TREATMENT OF THORACIC CANCER USING Ad-REIC/Dkk-3 AND A CHECKPOINT INHIBITOR
CA2239616A1 (en) Delivery of an agent to the ductal epithelium in the prophylactic and therapeutic treatment of cancer
Kolodkin-Gal et al. Herpes simplex virus delivery to orthotopic rectal carcinoma results in an efficient and selective antitumor effect
EP3733210A1 (en) Cell sheet for gene delivery
US20110044952A1 (en) Amplification of cancer-specific oncolytic viral infection by histone deacetylase inhibitors

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH - DIRECTOR DEITR, MA

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:THE JOHNS HOPKINS UNIVERSITY;REEL/FRAME:049221/0467

Effective date: 20190509