WO1994021115A1 - Broad spectrum tumor suppressor genes, gene products and methods for tumor suppression gene therapy - Google Patents

Broad spectrum tumor suppressor genes, gene products and methods for tumor suppression gene therapy Download PDF

Info

Publication number
WO1994021115A1
WO1994021115A1 PCT/US1994/003211 US9403211W WO9421115A1 WO 1994021115 A1 WO1994021115 A1 WO 1994021115A1 US 9403211 W US9403211 W US 9403211W WO 9421115 A1 WO9421115 A1 WO 9421115A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
protein
gene
tumor
vector
Prior art date
Application number
PCT/US1994/003211
Other languages
French (fr)
Inventor
Hong-Ji Xu
Shi-Xue Xu
William F. Benedict
Original Assignee
Baylor College Of Medicine
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 Baylor College Of Medicine filed Critical Baylor College Of Medicine
Priority to EP94911697A priority Critical patent/EP0693874A4/en
Priority to AU64154/94A priority patent/AU697446B2/en
Priority to JP52137494A priority patent/JP3739787B2/en
Priority to KR1019950704162A priority patent/KR100326136B1/en
Publication of WO1994021115A1 publication Critical patent/WO1994021115A1/en

Links

Classifications

    • 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
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4736Retinoblastoma protein
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • 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

Definitions

  • This invention is in the field of tumor suppressor genes (anti-oncogenes) and relates in general to products and methods for practicing broad- spectrum tumor suppressor gene therapy of various human cancers.
  • the invention relates to methods for treating tumor cells (1) administering vectors comprising a nucleic acid sequence coding for a second in-frame AUG codon-initiated retinoblastoma protein of about 94 kD or (2) administering an effective amount of a protein coded for by the nucleic acid sequence.
  • Cancers and tumors are the second most prevalent cause of death in the United States, causing 450,000 deaths per year. One in three Americans will develop cancer, and one in five will die of cancer (Scientific American Medicine, part 12, I, 1, section dated 1987). While substantial progress has been made in identifying some of the likely environmental and hereditary causes of cancer, the statistics for the cancer death rate indicates a need for substantial improvement in the therapy for cancer and related diseases and disorders. 1.3. Cancer Genes
  • cancer genes i.e., genes that have been implicated in the etiology of cancer, have been identified in connection with hereditary forms of cancer and in a large number of well-studied tumor cells. Study of cancer genes has helped provide some understanding of the process of tumorigenesis. While a great deal more remains to be learned about cancer genes, the presently known cancer genes serve as useful models for understanding tumorigenesis. Cancer genes are broadly classified into “oncogenes” which, when activated, promote tumorigenesis, and “tumor suppressor genes” which, when damaged, fail to suppress tumorigenesis. While these classifications provide a useful method for conceptualizing tumorigenesis, it is also possible that a particular gene may play differing roles depending upon the particular allelic form of that gene, its regulatory elements, the genetic background and the tissue environment in which it is operating.
  • oncogenes are somatic cell genes that are mutated from their wild-type alleles (the art refers to these wild-type alleles as protooncogenes) into forms which are able to induce tumorigenesis under certain conditions.
  • the art refers to these wild-type alleles as protooncogenes
  • the oncogenes ra ⁇ and myc are considered as models for understanding oncogenic processes in general.
  • the ra ⁇ oncogene is believed to encode a cytoplasmic protein
  • the myc oncogene is believed to encode a nuclear protein.
  • Neither the ra ⁇ oncogene nor the myc oncogene alone is able to induce full transformation of a normal cell into a tumor cell, but full tumorigenesis usually occurs when both the ras and myc oncogenes are present and expressed together in the same cell (Weinberg, R.A., .1989, Cancer Research 49:3713-3721, at page 3713).
  • Such collaborative effects have been observed between a number of other studied oncogenes.
  • the collaborative model of oncogene tumorigenesis must be qualified by the observation that a cell expressing the -ras oncogene that is surrounded by normal cells does not undergo full transformation. However, if most of the surrounding cells are also ras-expressing, then the ras oncogene alone is sufficient to induce tumorigenesis in a ras-expressing cell. This observation validates the multiple hit theory of tumorigenesis because a change in the tissue environment of the cell hosting the oncogene may be considered a second hit.
  • An alternative and equally valid hypothesis is that events that collaborate with the activation of an oncogene such as ra ⁇ or myc may include the inactivation of a negative regulatory factor or factors (Weinberg, R.A. , 1989, Cancer Research 49:3713-3721, at 3717; Goodrich, D.W. and Lee, W-H. , 1992, Nature 360:177-179), i.e., a tumor suppressor protein.
  • Tumor suppressor genes are genes that, in their wild-type alleles, express proteins that suppress abnormal cellular proliferation. When the gene coding for a tumor suppressor protein is mutated or deleted, the resulting mutant protein or the complete lack of tumor suppressor protein expression may fail to correctly regulate cellular proliferation, and abnormal cellular proliferation may take place, particularly if there is already existing damage to the cellular regulatory mechanism. A number of well- studied human tumors and tumor cell lines have been shown to have missing or nonfunctional tumor suppressor genes.
  • tumor suppression genes include, but are not limited to, the retinoblastoma susceptibility gene or RB gene, the p53 gene, the deleted in colon carcinoma (DCC) gene and the neurofibromatosis type 1 (NF-1) tumor suppressor gene (Weinberg, R.A. Science, 1991, 254:1138-1146). Loss of function or inactivation of tumor suppressor genes may play a central role in the initiation and/or progression of a significant number of human cancers.
  • tumor suppressor genes are large and growing. The following discussion of tumor suppressor genes is not intended to provide a complete review of all known and putative tumor suppressor genes, but is provided as background to indicate the state of the art and the problems to be overcome before the art is able to provide successful genetic therapy of diseases and disorders characterized by abnormally proliferating cells, e.g., tumor or cancer cells.
  • the Retinoblastoma Gene is one of the better studied tumor suppressor genes.
  • the size of the RB gene complementary DNA (cDNA) permits ready manipulation of the gene, so that insertions of the RB gene have been made into a number of cell lines.
  • the RB gene has been shown to be missing or defective in a majority of retinoblastomas, sarcomas of the soft tissues and bones, and in approximately 20 to 40 percent of breast, lung, prostate and bladder carcinomas (Lee, W-H. , et al., PCT Publ. No. WO 90/05180, at pages 38 and 39; see also, Bookstein, R. and Lee, W-H., 1991, Crit. Rev. Oncog. , 2:211-217; Benedict, W.F. et al., J. Clin. Invest.. 1990, 85:988-993) .
  • the predicted RB gene product has 928 amino acids and an expected molecular weight of 106 kD (Lee et al., 1987, Nature, 329:642-645).
  • the natural factor corresponding to the predicted RB gene expression product has been identified as a nuclear phosphoprotein having an apparent relative molecular mass (Mr) of 110-114 kD (Lee et al., 1987, Nature. 329:642-645) or 110-116 kD (Xu et al., 1989, Onco ⁇ ene 4:807-812).
  • Mr apparent relative molecular mass
  • the literature generally refers to the protein encoded by the RB gene as pllO* 8 .
  • the full length RB protein of 928 amino acids is also referred to as the 115 kD (Yokota et al., 1988, Oncogene, 3:471-475), or 105 kD (Whyte et al . , 1988, Nature. 334:124-129) RB proteins.
  • Various mutations of the RB gene are known. These are generally inactive.
  • pS ⁇ 3 a 56 kD truncated RB protein, designated as pS ⁇ 3 , that is considered to function in the same way as does pllO 113 retains activity (Goodrich et al., 1992, Nature 360:177-179).
  • normal human cells show an RB protein pattern consisting of a lower sharp band with an Mr of 110 kD and a broader, more variable region above this band with an Mr ranging from 110 kD to 116 kD.
  • the 110 kD band is the underphosphorylated RB protein, whereas the broader region represents the phosphorylated RB protein.
  • the heterogeneity of the molecular mass results from a varying degree of phosphorylation (Xu et al., 1989, Oncogene, 4:807-812) .
  • the RB protein shows cyclical changes in phosphorylation. Most RB protein is unphosphorylated during Gl phase, but most (perhaps all) RB molecules are phosphorylated in S and G2 phases (Xu et al., 1989, Oncogene, 4:807-812; DeCaprio et al., 1989, Cell. 58:1085-1095; Buchkovich et al., 1989, Cell. 58:1097-1105; Chen et al., 1989, Cell. 58:1193-1198; Mihara et al., 1989, Science. 246:1300-1303).
  • underphosphorylated RB protein binds to SV40 large T antigen. Given that RB protein binding by large T antigen is probably important for the growth promoting effects of large T antigen, this suggests that the underphosphorylated RB protein is the active form of the RB protein, and the phosphorylated RB protein in S and G2 phases is inactive (Ludlow et al., 1989, Cell. 56:57-65).
  • the RB gene expressing the first in-frame AUG codon-initiated RB protein is also referred to herein as the intact RB gene, the RB 110 gene or the pllO ⁇ coding gene. It has also been observed that lower molecular weight ( ⁇ 100 kD, 98 kD, or 98-104 kD) bands of unknown origin which are immunoreactive to various anti-RB antibodies can be detected in immunoprecipitation and Western blots (Xu et al., 1989, Oncogene. 4:807-812; Furukawa et al., 1990, Proc. Natl. Acad. Sci.. USA. 87:2770-2774; Stein et al., 1990, Science. 249:666-669).
  • the deduced second AUG codon-initiated RB protein would be 98 kD, or 12 kD smaller than the pllO* 8 protein. It has been proposed that the lower molecular weight bands are the underphosphorylated (98 kD) and phosphorylated (98-104 kD) RB protein translated from the second AUG codon of the RB mRNA (Xu et al., 1989, Oncogene, 4:807-812), although no data directly supported this hypothesis.
  • Sections 4.2.1, and Figure 5 infra provide data indicating the non-identity of the 98 kD protein bands of unknown origin and the second AUG codon-initiated protein products. It has been proposed that introduction of a functional RB 110 gene into an RB-minus tumor cell will likely "normalize" the cell. Of course, it is not expected that tumor cells which already have normal RB 110 gene expression ("RB+”) will respond to RB 110 gene therapy, because it is presumed that adding additional RB expression cannot correct a non-RB genetic defect.
  • Neurofibromatosis type 1 or von Recklinghausen neurofibromatosis results from the inheritance of a predisposing mutant allele or from alleles created through new germline mutations (C.J. Marshall, 1991, Cell. 64:313-326).
  • the neurofibromatosis type 1 gene referred to as the NF1 gene, is a relatively large locus exhibiting a mutation rate of around 10 4 . Defects in the NF1 gene result in a spectrum of clinical syndromes ranging from cafe-au-lait spots to neurofibromas of the skin and peripheral nerves to Schwannomas and neurofibrosarcomas.
  • the NF1 gene encodes a protein of about 2485 amino acids that shares structural similarity with three proteins that interact with the products of the ra ⁇ protooncogene (Weinberg et al., 1991, Science. 254:1138-1146 at page 1141).
  • the NF1 amino acid sequence shows sequence homology to the catalytic domain of ra ⁇ GAP, a GTPase-activating protein for p21 ra ⁇ (C.J. Marshall, 1991, Cell. 64:313-326 at pages 320 and 321).
  • NFl in cell cycle regulation
  • yeast C.J. Marshall, (1991, Cell. 64:313-326, bridging pages 320 and 321, and citing to Ballester et al, 1990, Cell. 63:851-859).
  • C.J. Marshall 1991, Cell. 64:313-326 at page 321; Weinberg et al., 1991, Science. 254:1138-1146 at page 1141).
  • the p53 Gene Somatic cell mutations of the p53 gene are said to be the most frequently mutated gene in human cancer (Weinberg et al., 1991, Science. 254:1138-1146 at page 1143) .
  • the normal or wild-type p53 gene is a negative regulator of cell growth, which, when damaged, favors cell transformation (Weinberg et al. supra) .
  • the p53 expression product is found in the nucleus, where it may act in parallel with or cooperatively with pllO 8 . This is suggested by a number of observations, for example, both p53 and pllO* 8 proteins are targeted for binding or destruction by the oncoproteins of SV40, adenovirus and human papillomavirus.
  • Tumor cell lines deleted for p53 have been successfully treated with wild-type p53 vector to reduce tu origenicity (Baker, S.J., et al., 1990, Science. 249:912-915) .
  • the introduction of either p53 or RB U0 into cells that have not undergone lesions at these loci does not affect cell 5 proliferation (Marshall, C.J., 1991, Cell, 64:313-326 at page 321; Baker, S.J., et al., 1990, Science. 249:912-915; Huang, H.-J.S., et al., 1988 Science. 242:1563-1566).
  • Such experiments suggest that sensitivity of cells to the suppression of their
  • the DCC gene is a more than approximately one million base pair gene coding for a 190-kD transmembrane phosphoprotein which is hypothesized to be a receptor (Weinberg et al., 1991, Science. 254:1138-1146 at page 1141), the loss of which allows the affected cell a growth advantage. It has also been noted that the DCC has partial sequence homology to the neural cell adhesion molecule (Marshall, 1991, Cell. 64:313-326) which might suggest a role for the DCC protogene in regulating cell to cell interactions.
  • the large size and complexity of the DCC gene, together with the complexity of the K-ras, p53 and possibly other genes involved in colon cancer tumorigenesis demonstrates a need for a broad-spectrum tumor suppressor gene and methods of treating colon carcinoma cells which do not depend upon manipulation of the DCC gene or on the identification of other specific damaged genes in colon carcinoma cells.
  • Retroviral vectors in this context are retroviruses from which all viral genes have been removed or altered so that no viral proteins are made in cells infected with the vector. Viral replication functions are provided by the use of retrovirus 'packaging' cells that produce all of the viral proteins but that do not produce infectious virus. Introduction of the retroviral vector DNA into packaging cells results in production of virions that carry vector RNA and can infect target cells, but no further virus spread occurs after infection. To distinguish this process from a natural virus infection where the virus continues to replicate and spread, the term transduction rather than infection is often used.
  • retroviral vectors for gene therapy are the high efficiency of gene transfer into replicating cells, the precise integration of the transferred genes into cellular DNA, and the lack of further spread of the sequences after gene transduction (Miller, A.D., Nature. 1992, 357:455-460) .
  • the potential for production of replication- competent (helper) virus during the production of retroviral vectors remains a concern, although for practical purposes this problem has been solved. So far, all FDA-approved retroviral vectors have been made by using PA317 amphotropic retrovirus packaging cells (Miller, A.D., and Buttimore, C. , Molec. Cell Biol. , 1986, 6:2895-2902).
  • helper virus production Even by stringent assays that allow for amplification of such events (Lynch, CM., and Miller, A.D., J. Viral.. 1991, 65:3887-3890).
  • Other packaging cell lines are available. For example, cell lines designed for separating different retroviral coding regions onto different plasmids should reduce the possibility of helper virus production by recombination. Vectors produced by such packaging cell lines may also provide an efficient system for human gene therapy (Miller, A.D., 1992, Nature, 357:455-460).
  • Non-retroviral vectors have been considered for use in genetic therapy.
  • One such alternative is the adenovirus (Rosenfeld, M.A. , et al., 1992, Cell.
  • adenovirus vectors Major advantages of adenovirus vectors are their potential to carry large segments of DNA (36 Kb genome) , a very high titre (10 11 ml" 1 ) , ability to infect non-replicating cells, and suitability for infecting tissues in situ, especially in the lung.
  • Plasmid DNA should be easy to certify for use in human gene therapy because, unlike retroviral vectors, it can be purified to homogeneity.
  • liposome-mediated DNA transfer several other physical DNA transfer methods such as those targeting the DNA to receptors on cells by complexing the plasmid DNA to proteins have shown promise in human gene therapy (Wu, G.Y., et al., 1991, J. Biol. Chem..
  • the proposed method of treatment requires identification of the damaged tumor suppressor gene, and introduction of the corresponding undamaged gene (including a promoter and a complete encoding sequence) into the affected tumor cells by means of a vector such as a retrovirus able to express the gene product. It is proposed that the incorporated functional gene will convert the target cell to a non-malignant state.
  • the pllO* 8 reconstituted tumor cells still form invasive tumors in nude mice (Xu, H-J., et al., 1991, Cancer Research. 51:4481-4485; Takahashi, R. , et al., 1991, Proc. Natl. Acad. Sci.. USA. 88:5257-5261; Banerjee, A., et al., 1992, Cancer Research. 52:6297-6304).
  • pllO* 8 reconstituted retinoblastoma cells inoculated into an orthotopic site in this instance, the eye
  • consistently produced tumors Xu, H-J., et al., 1991, Cancer Research 51:4481-4485) .
  • Another proposed method of treating cancer by gene therapy is to antagonize the function of an oncogene by placing an artificial gene, constructed to have an inverted nucleotide sequence compared to the oncogene, into a tumor cell (U.S. patent number 4,740,463, issued April 26, 1988 by Weinberg, et al.).
  • retinoblastoma gene is one of those tumor suppressor genes that is readily accessible to study, thus it provides a model for understanding some of the other disadvantages to cancer gene replacement therapy as heretofore understood. It is known that reintroduction of the retinoblastoma tumor suppressor gene into RB-defective tumor cells inhibits the tumor cell growth and suppresses the neoplastic phenotype of the target cells (WO 90/05180, cited supra ; Huang et al., 1988, Science.
  • tumorigenicity is often incomplete.
  • a significant percentage of the RB- reconstituted tumor cells still form small tumors after a longer latency period in nude mouse tumorigenicity assays.
  • Such tumors although retaining normal RB expression, are histologically malignant and invasive (Xu et al., 1991, Cancer Res. , 51:4481-4485; Takahashi et al., 1991, Proc. Natl. Acad. Sci.. USA. 88:5257-5261; Banerjee et al., 1992, Cancer Res.. 52:6297-6304).
  • TSGR tumor suppressor gene resistance
  • RB proteins have an active form (underphosphorylated protein) and an inactive form (phosphorylated protein) . Therefore, RB-positive tumor cells may have inherited or acquired the ability to phosphorylate RB proteins to the inactive state and allow tumor cell proliferation to continue.
  • conversion of RB-minus cells with plasmid or virus vectors coding for the pllO* 8 protein provides only incomplete suppression, or even exacerbation of a percentage of the malignant cell population because the pllO* 8 protein remains phosphorylated and inactive in some of the target cells.
  • the tumor cells expressing the RB 110 gene may simply have again inactivated the RB 110 gene by mutation in subsequent cell divisions (Lee et al., 1990, Immunol. Ser. 51:169-200, at page 188) .
  • the tumor cells expressing the RB 110 gene may simply have again inactivated the RB 110 gene by mutation in subsequent cell divisions (Lee et al., 1990, Immunol. Ser. 51:169-200, at page 188) .
  • the second in-frame AUG codon-initiated retinoblastoma suppressor protein of about 94 kD (p94 RB ) is a broad- spectrum tumor suppressor, and that insertion of a gene capable of expressing this protein, or the protein itself, into an abnormally proliferating cell, such as a cancer or tumor cell, causes that cell to enter a senescent-like state, terminating the proliferation.
  • the cell so-treated simply stops replicating and dies.
  • the cell may possess any type of genetic defect, known or unknown, so that there is no need to determine the exact nature of the genetic defect associated with the abnormal proliferation. Further, the population of treated cells exhibits an unexpectedly much lower incidence of TSGR resurgence and exacerbation of malignancy than do cells treated with any other tumor suppressor gene. The method is repeated as needed.
  • the invention provides p94 RB encoding vectors and p94 RB proteins for use in treatment of tumors or cancers, and methods of preparing p94 RB proteins suitable for use in methods of treatment.
  • the invention also provides methods of treatment for mammals such as humans, as well as methods of treating abnormally proliferating cells, such as cancer or tumor cells.
  • the invention contemplates treating abnormally proliferating cells, or mammals having a disease characterized by abnormally proliferating cells by any suitable method known to permit a host cell compatible p94 RB encoding vector or a p94 ⁇ protein to enter the cells to be treated so that suppression of proliferation is achieved.
  • the invention comprises a method of treating a disease characterized by abnormally proliferating cells, in a mammal, by administering an expression vector coding for p94 RB to the mammal having a disease characterized by abnormally proliferating cells, inserting the expression vector into the abnormally proliferating cells, and expressing p94 RB in the abnormally proliferating cells in an amount effective to suppress proliferation of those cells.
  • the expression vector is inserted into the abnormally proliferating cells by viral infection or transduction, liposome-mediated transfection, polybrene-mediated transfection, CaP04 mediated transfection and electroporation. The treatment is repeated as needed.
  • the invention comprises a method of treating abnormally proliferating cells of a mammal by inserting a p94 RB encoding expression vector into the abnormally proliferating cells and expressing p94 RB therein in amounts effective to suppress proliferation of those cells. The treatment is repeated as needed.
  • the invention provides a DNA molecule able to suppress growth of an abnormally proliferating cell.
  • the DNA molecule encodes a p94 RB protein having an amino acid sequence substantially according to SEQ ID NO:3, provided that the DNA molecule does not also code for a pllO RB protein.
  • the DNA molecule has the DNA sequence of SEQ ID NO:l, and is expressed by an expression vector.
  • the expression vector may be any host cell-compatible vector.
  • the vector is preferably selected from the group consisting of a retroviral vector, an adenoviral vector and a herpesviral vector.
  • the invention provides a p94 RB protein having an amino acid sequence substantially according to SEQ ID NO:3.
  • the invention provides a method of producing a p94 RB protein by the steps of: inserting a compatible expression vector comprising a p94 RB encoding gene into a host cell and causing the host cell to express p94 RB protein.
  • the invention comprises a method of treating abnormally proliferating cells of a mammal ex vivo by the steps of: removing a tissue sample in need of treatment from a mammal, the tissue sample comprising abnormally proliferating cells; contacting the tissue sample in need of treatment with an effective dose of an p94 RB encoding expression vector; expressing the p94 RB in the abnormally proliferating cells in amounts effective to suppress proliferation of the abnormally proliferating cells.
  • the treatment is repeated as necessary; and the treated tissue sample is returned to the original or another mammal.
  • the tissue treated ex vivo is blood or bone marrow tissue.
  • the invention comprises a method of treating a disease characterized by abnormal cellular proliferation in a mammal by a process comprising the steps of administering p94 RB protein to a mammal having a disease characterized by abnormally proliferating cells, such that the p94 RB protein is inserted into the abnormally proliferating cells in amounts effective to suppress abnormal proliferation of the cells.
  • the p94 RB protein is liposome encapsulated for insertion into cells to be treated. The treatment is repeated as necessary.
  • the invention comprises a method of treating abnormally proliferating cells of a mammal ex vivo by a process comprising the steps of removing a tissue sample in need of treatment from a mammal, the tissue sample comprising abnormally proliferating cells contacting the tissue sample in need of treatment with an effective dose of a p94 RB protein. The treatment is repeated as necessary, and then the treated tissue is returned to the mammal or placed into another mammal.
  • the tumor or cancer cells to be treated are cells having one or more genetically defective tumor suppressor genes and oncogenes selected from the group consisting of an RB, a p53, a c-myc, an N-ras and a c-yes-1 gene.
  • the tumor or cancer cells are cells having no detectable genetic defect of a tumor suppressor gene selected from the group consisting of an RB gene and a p53 gene.
  • the tumor or cancer cells are lung carcinoma cells.
  • the p94 RB encoding expression vector or the p94 RB protein are administered by means of aerosol delivery of liposome- encapsulated p94 RB encoding expression vector or p94 RB protein into a lung in need of such treatment.
  • cancer or tumor are clinically descriptive terms which encompass a myriad of diseases characterized by cells that exhibit unchecked and abnormal cellular proliferation.
  • tumor when applied to tissue, generally refers to any abnormal tissue growth, i.e., excessive and abnormal cellular proliferation.
  • a tumor may be "benign” and unable to spread from its original focus, or
  • malignant and capable of spreading beyond its anatomical site to other areas throughout the hostbody.
  • cancer is an older term which is generally used to describe a malignant tumor or the disease state arising therefrom.
  • the art refers to an abnormal growth as a neoplasm, and to a malignant abnormal growth as a malignant neoplasm.
  • abnormal cellular proliferation is the result of a failure of one or more of the mechanisms controlling cell growth and division.
  • the mechanisms controlling cell growth and division include the genetic and tissue-mediated regulation of cell growth, mitosis and differentiation. These mechanisms are thought to act at the cell nucleus, the cell cytoplasm, the cell membrane and the tissue-specific environment of each cell. The process of transformation of a cell from a normal state to a condition of excessive or abnormal cellular proliferation is called tumorigenesis.
  • tumorigenesis is usually a multistep progression from a normal cellular state to, in some instances, a full malignancy. It is therefore believed that multiple "hits" upon the cell regulatory mechanisms are required for full malignancy to develop. Thus, in most instances, it is believed that there is no single cause of excessive proliferation, but that these disorders are the end result of a series of cumulative events.
  • a malignant tumor or cancer capable of unchecked and rapid spread throughout the body is the most feared and usually the deadliest type of tumor, even so-called benign tumors or growths can cause significant morbidity and mortality by their inappropriate growth.
  • a benign tumor can cause significant damage and disfigurement by inappropriate growth in cosmetically sensitive areas, or by exerting pressure on central or peripheral nervous tissue, blood vessels and other critical anatomical structures.
  • a broad-spectrum tumor suppressor gene is a genetic sequence coding for a protein that, when inserted into and expressed in an abnormally proliferating host cell, e.g., a tumor cell, suppresses abnormal proliferation of that cell irrespective of the cause of the abnormal proliferation.
  • the second in-frame AUG (ATG in DNA) codon-initiated retinoblastoma gene disclosed herein exemplifies such a broad-spectrum tumor suppressor gene and is referred to herein as the p94 RB coding gene, as the RB 94 gene or as a DNA molecule coding for pRB 94 .
  • the p94 RB coding gene comprises the nucleotide sequence from exon 3, nucleotide 355 to exon 27, nucleotide 264.
  • the p94 RB encoding gene by definition excludes that portion of the RB n0 gene upstream from the second in-frame AUG start codon.
  • Figures 1A-1F show the DNA sequence of the RB 94 gene wherein the ATG codon begins at nucleotide 19 of that figure (SEQ ID NO:l; SEQ ID NO:2) .
  • a broad-spectrum tumor suppressor protein (including phosphoproteins, lipoproteins, glycoproteins and other protein-based derivatives) is a substance that when injected into, absorbed by or caused to be expressed in any abnormally proliferating cell, reduces or completely suppresses abnormal cellular proliferation.
  • the protein expressed by the second in-frame AUG codon-initiated retinoblastoma gene disclosed herein exemplifies such a broad- spectrum tumor suppressor protein. It is a phosphoprotein of about 94 kD relative molecular mass, and is also referred to herein as p94 RB (SEQ ID NO:3).
  • any other fragment of a tumor suppressor protein e.g., the third or fourth AUG codon-initiated retinoblastoma protein of about 90 kD and 83 kD, respectively, also has the property of suppressing abnormal cellular proliferation.
  • Figures 2A-2F Amino acid sequence of the 94 kDa therapeutic RB protein (SEQ ID N0:3).
  • Figure 3 Construction of baculovirus expression vector for the 94 kDa therapeutic RB protein synthesis; *R.S. is recombination sequence.
  • Figures 4A and 4B Intracellular localization of recombinant baculovirus-produced pllO* 3 and p94 RB in insect cells: Figure 4A shows mock-infected Sf9 cells; Figure 4B shows cells producing pllO ⁇ ; and Figure 4C shows cells producing p94 RB ; note that protein is localized to the nucleus in Figures 4B and 4C. Protein localization is by anti-RB immunochemical staining.
  • Figure 5 A diagram of complex formation of baculovirus-expressed and subsequently purified pllO* 8 and p94 RB proteins with SV40 T antigen.
  • the immunoaffinity chromatography purified proteins were mixed with an equal amount of T antigen, and aliquots of the mixture were immunoprecipitated with PAB419 anti-T antibody, followed by Western blotting. The blot was sequentially incubated with MAb-1 anti-RB antibody and PAB419 antibody.
  • Lane 1 lysate of T antigen immortalized W138 VA13 fibrobrasts was used as a control; lane 2, purified pllO* 8 ; lane 3, co- precipitation of T-Ag with pllO* 8 ; lane 4, purified p94 RB ; lane 5, co-precipitation of T-Ag with p94 ⁇ .
  • Figures 6A and 6B Construction of recombinant plasmids for high-level expression of pllO* 8 (pCMV-f- RB35) and p94 RB (pCMV-s-RB42) proteins in human cells using cytolomegalovirus promoter/enhancer:
  • Figure 6A is an explanatory drawing of the pllO* 3 coding cDNA;
  • Figure 6B provides maps of the pllOTM and p94 RB expression plasmids where pCMV-f-RB35 codes for pllO* 3 and pCMV-s-RB42 codes for p94 RB .
  • pCMV-s-RB42 has most of pllO 83 coding region deleted upstream of the second ATG.
  • Figures 7A and 7B Construction of recombinant plasmids for expression of pllO 0 (pBA-f-RB33) and p94 RB (p ⁇ A-s-RB34) proteins in human cells using ⁇ -actin promoter:
  • Figure A is a map of the pllO* 8 coding plasmid, pBA-f-RB33;
  • Figure B is a map of the p94 RB coding plasmid, pBA-s-RB34. Note that pBA-s-RB34 has most of the pllO* 8 coding region deleted upstream of the second ATG.
  • Figures 8A, 8B and 8C Morphological effects of pllO* 8 and p94 RB expression on RB-defective bladder carcinoma cell line 5637 (ATCC HTB9) :
  • Figure 8A is mock-transfected HTB9 cells;
  • Figure 8B is pllO 88 expressing HTB9 transfectants;
  • Figure 8C is p94 RB - expressing HTB9 transfectants.
  • Arrows indicate examples for RB-positive immunostained cells. Note that the pllO* 8 expressing cells of Figure 8B appear normal, but that the p94 RB expressing cells of Figure 8C are senescent.
  • FIG. 9 Half-life analysis of pllOTM and p94 RB proteins in RB-reconstituted bladder carcinoma cell line, 5637.
  • the bladder tumor cells were transfected in multiple dishes with either pllO** (p ⁇ A-f-RB33) or p94 ⁇ (pBA-s-RB34) expression plasmids. Twenty-four hours after transfection the cells were labeled with [ 35 S]-methionine and chased with excess unlabeled methionine for 0, 6, 12 and 24 hours, respectively.
  • pllO* 8 and p94 RB proteins were determined by immunoprecipitation: the left side of the figure (0-12 hours) shows the half-life of pllO* 8 is less than 6 hours; the right side of the figure (0-24 hours) shows the half-life of p94 RB is about 12 hours.
  • FIG. 10 Western blot analysis of exogenous pllO ⁇ and p94 RB proteins in transiently transfected 5637 cells showing the distinct underphosphorylation state of the p94 ⁇ protein: lane 1 shows normal human fibroblast cell line, WI-38; lane 2 shows parental RB- minus bladder carcinoma cell line, 5637; lane 3 shows 5637 cells transfected with pllO ⁇ -expressing plasmid; lane 4 shows 5637 cells transfected with p94 RB - expressing plasmid.
  • FIGS 11A-11C Expression of the human full- length RB protein, pllO* 8 ( Figure 11B) and the broad- spectrum tumor suppressor protein, p94 RB ( Figure 11C) in normal (non-tumorigenic) mouse fibroblast cells via retrovirus plasmid vectors. Both the pllO* 8 - and p94 RB - expressing cells (arrows) have normal viable morphology similar to the parental cells Figure 11A) .
  • Figures 12A-12D Expression of the human pllO 88 ( Figure 12C) and p94 RB ( Figure 12D) proteins in mouse urinary bladder mucous membranes in vivo .
  • Liposomes were mixed with the pllO**- and p94 RB - expressing plasmids, respectively and infused directly into the mouse bladder via a catheter.
  • transitional epithelia expressing the pllOTM Figure 12C, arrows
  • p94 RB Figure 12D, arrows
  • the present invention is based upon the unexpected discovery that p94 RB expressed by an expression vector in any abnormally proliferating target cell, e.g., a cancer or tumor cell, causes the suppression of the abnormal proliferation.
  • the treatment has been effective with all tested tumor cell lines and is not limited to treatment of RB-minus tumor cells.
  • the p94 RB protein remains in the active, underphosphorylated form, and has a half-life in the target cell which is two to three times longer than that of pllO**.
  • a synergistic combination of accumulation of p94 RB together with its tendency to remain in an underphosphorylated, active form serves to terminate the cell replication cycle in target tumor cells.
  • the property of suppressing cell growth and inducing senescence or killing any abnormally proliferating cell irrespective of its genetic defect, is nevertheless completely unanticipated and unexpected.
  • a gene coding for the second in- frame AUG codon-initiated RB protein i.e., p94 RB
  • p94 RB was expressed by a baculovirus vector in insect host cells as a stable nuclear phosphoprotein.
  • the resulting unphosphorylated forms of p94 RB were able to form a specific complex with SV40 T antigen, providing an important verification that the p94 RB protein shares many functional properties of the naturally occurred piio** protein, i.e., phosphorylation, viral oncoprotein association and nuclear tethering (Templeton et al., 1991, Proc. Natl. Acad. Sci.. USA. 88:3033-3037) .
  • the effects of transfection by either first or second in-frame AUG codon-initiated RB protein expression plasmid were compared on a number of well known human tumor cell lines.
  • the tested cell lines included: an RB-defective human bladder carcinoma cell line, 5637 (ATCC HTB9) ; RB-defective human breast carcinoma cell line, MDA-MB-468 (ATCC HTB132) ; RB- defective human non-small cell lung carcinoma cell line, H2009 (Kratzke, R.A. , et al., 1992, The Journal of Biological Chemistry.
  • RB- defective human prostate carcinoma cell line DU145 (ATCC HTB81) ; RB-defective human osteosarcoma cell line, Saos-2 (ATCC HTB85) ; RB-defective human fibrosarcoma metastatic to lung cell line, Hs913T (ATCC HTB152) ; human cervix adenocarcinoma cell line, HeLa (ATCC CCL2) and human fibrosarcoma cell line, HT1080 (ATCC CCL121) . Both the HeLa and HT1080 cell lines have normal pll0 RB expression.
  • the study also demonstrated that the RB-minus tumor cells expressing the second in-frame AUG codon- initiated RB protein, p94 RB , did not progress through the cell cycle, as evidenced by their failure to incorporate [ 3 H]-thymidine into DNA.
  • the percentage of cells undergoing DNA replication was only slightly lower in cells producing the intact RB protein (pllO 1 ⁇ ) than in cells that were RB-negative.
  • the RB- defective bladder carcinoma cell line, 5637 failed to phosphorylate the second in-frame AUG codon-initiated RB protein as shown by Western blot analysis.
  • the intact RB protein (pllO* 3 ) expressed in transfected 5637 cells were fully phosphorylated.
  • the half-life of the second in-frame AUG codon-initiated RB protein, p94 RB was shown to be two- to three-fold greater than the intact RB protein (pllO* 8 ) . Therefore, the accumulation of only unphosphorylated (active) p94 RB proteins may account for the failure of transiently transfected 5637 tumor cells to enter S phase, and this in turn may cause these tumor cells to senesce and die.
  • p94 RB protein has its preferentially associated cellular proteins as compared to lOO* 8 . This difference in associated proteins may also contribute to the unique broad-spectrum tumor cell growth suppressing functions of p94 RB protein.
  • an advantage of the present invention is that the methods and products herein disclosed can be used for therapeutic treating tumors having no specific tumor suppressor gene defects, which provides a significant advantage over previous techniques for human tumor suppressor gene therapy.
  • Table 1 on the following page, provides a summary of the identification of the tested tumor cell lines, their tumor origin and genetic defects.
  • John Wiley & Sons, New York may be used to construct p ⁇ * 3 encoding gene expression vectors consisting of appropriate transcriptional/translational control signals and the desired RB cDNA sequence downstream from the first in- frame AUG codon, that is unable to code for pllO* 8 .
  • These methods may include in vitro DNA recombinant and synthetic techniques and in vivo genetic recombination.
  • Expression of a nucleic acid sequence encoding a p94 RB may be regulated by a second nucleic acid sequence so that the p94 RB is expressed in a host infected or transfected with the recombinant DNA molecule.
  • expression of p94 RB may be controlled by any promoter/enhancer element known in the art.
  • the promoter activation may be tissue specific or inducible by a metabolic product or administered substance.
  • Promoters/enhancers which may be used to control p94 RB gene expression include, but are not limited to, the native RB promoter, the cytomegalovirus (CMV) promoter/enhancer (Karasuyama, H. , et al., 1989, J. Exp. Med.. 169:13), the human ⁇ -actin promoter (Gunning, P., et al., 1987, Proc. Natl. Acad. Sci. USA. 84:4831-4835), the glucocorticoid-inducible promoter present in the mouse mammary tumor virus long terminal repeat (MMTV LTR) (Klessig, D.F., et al. , 1984, Mol. Cell Biol..
  • CMV cytomegalovirus
  • MMTV LTR mouse mammary tumor virus long terminal repeat
  • Rous sarcoma virus (Yamamoto et al., 1980, Cell 22:787-797), the herpes simplex virus (HSV) thymidine kinase promoter/enhancer (Wagner et al., 1981, Proc. Natl. Acad. Sci. U.S.A. 78:1441-1445), the regulatory sequences of the metallothionein gene (Brinster et al., 1982, Nature 296:39-42), the adenovirus promoter (Yamada et al., 1985, Proc. Natl.
  • Expression vectors compatible with mammalian host cells for use in genetic therapy of tumor or cancer cells include, but are not limited to: plasmids, retroviral vectors, adenovirus vectors, herpes viral vectors, and non-replicative avipox viruses, as disclosed, for example, by U.S. Patent No. 5,174,993.
  • a plasmid vector derived from pHBAPr-1-neo was constructed for expression of p94 RB in mammalian cells by placing the coding sequence for p94 ⁇ under control of the human ⁇ - actin gene promoter (Gunning, P. et al., Proc. Natl. Acad. Sci..
  • a plasmid vector derived from pCMV-Neo-Ba (Baker, S.J., et al., Science. 1990, 249:912-915), was constructed for expression of p94 RB in mammalian cells by placing the coding sequence for p94 RB under control of the cytomegalovirus (CMV) promoter/enhancer sequences.
  • CMV cytomegalovirus
  • pLLRNL Miller, A.D., et al., 1985, Proc. Natl. Acad. Sci.. USA. 5:431
  • pLLRNL Miller, A.D., et al., 1985, Proc. Natl. Acad. Sci.. USA. 5:431
  • p94 RB protein under the control of the MuLV LTR promoter, the CMV promoter, the ⁇ -actin promoter or any other effective promoter.
  • an adenovirus type 5 (Ad5) deletion mutant, Ad-dl324, and a plasmid, pTG5955 are used to construct an adenovirus vector able to infect mammalian cells and express p94 RB protein under the control of the adenovirus type 2 (Ad2) major late promoter, the CMV promoter, the ⁇ -actin promoter or any other effective promoter.
  • expression vectors compatible with host cells suitable for production of p94 RB may be constructed to express p94 RB protein in those compatible host cells. These include but are not limited to mammalian cells infected with a virus (e.g., adenovirus, retrovirus, herpes simplex virus, avipox virus) ; insect cells infected with a virus (e.g. , baculovirus) ; microorganisms such as yeasts containing yeast vectors, or bacteria transformed with bacteriophage DNA, plasmid DNA, or cosmid DNA.
  • the expression controlling elements of vectors vary in their strengths and specifications. Depending on the host-vector system utilized, any one of a number of suitable transcription and translation elements may be used.
  • the produced p94 RB may be purified from host cells by affinity chromatography, electrophoresis, high-performance liquid chromatography (HPLC) or any other methods known to the art.
  • an engineered derivative of Autographa California Multiple Nuclear Polyhedrosis Virus (“AcMNPV”) was used to produce p94 RB protein in cultured Fall Army worm Spondoptera frugiperda cells (Sf9 cells) with a strong temporally regulated promoter of the polyhedron gene whose product represents 50% or more of total cellular proteins during a lytic infection.
  • the baculovirus-expressed p94 ⁇ protein was subsequently purified by im unoaffinity chromatography.
  • Expression vectors containing p94 RB coding inserts can be identified by three general approaches: (a) nucleic acid hybridization, (b) presence or absence of "marker" gene functions, and (c) expression of inserted sequences.
  • the presence of a p94 RB coding gene inserted in an expression vector can be detected by nucleic acid hybridization using probes comprising sequences that are homologous/complementary to the inserted p94 RB coding gene.
  • Such hybridization can be carried out under stringent or nonstringent conditions, depending upon the size and sequence of the probe selected.
  • the expression vector/host system can be identified and selected based upon the presence or absence of certain "marker" gene functions (e.g., thymidine kinase activity, resistance to antibiotics, viral occlusion formation in a baculovirus vector infected insect cell, etc.) caused by introduction of the expression vector into the host cell.
  • certain "marker" gene functions e.g., thymidine kinase activity, resistance to antibiotics, viral occlusion formation in a baculovirus vector infected insect cell, etc.
  • the expression vector containing the p94 RB coding gene can be identified by the presence of the marker gene function (geneticin resistance) .
  • expression vectors containing a p94 RB coding gene can be identified by assaying the p94 RB coding gene products expressed by the vectors.
  • assays can be based, for example, on the physical or functional properties of the p94 RB gene products in in vitro or in vivo assay systems including metabolic radiolabelling by [ 35 S] methionine, SDS-polyacrylamide gel electrophoresis, binding with a specific antibody, and phosphorylation by a protein kinase.
  • An appropriate p94 RB coding expression vector i.e., a vector which contains the necessary elements for the transcription and translation of the p94 RB protein-coding sequence may be introduced into a host cell.
  • a host cell may be any cell type compatible with the vector for expressing and producing p94 RB .
  • the host cell is a mammalian tumor cell to be treated.
  • the host cell is a human tumor cell to be treated.
  • Expression of the p94 RB in a host cell may be transient, permanent, or inducible.
  • the necessary transcriptional and translational signals can also be supplied by the native RB gene and/or its flanking regions.
  • a variety of vector/host systems may be utilized to express the p94 RB protein-coding sequence in a tumor cell to be treated. These include but are not limited to mammalian cell systems transfected, infected or transduced with a plasmid, or a virus (e.g., adenovirus, retrovirus, herpes simplex virus, avipox virus) .
  • the expression elements of vectors vary in their strengths and specificities. Depending on the host cell to be treated, any one or more of a number of suitable transcription and translation elements may be used. 3.3.3.
  • the p94 RB encoding gene construct of the present invention may be placed by methods well known to the art into an expression vector such as a plasmid or viral expression vector.
  • a plasmid expression vector may be introduced into a tumor cell by calcium phosphate transfection, liposome (for example, LIPOFECTIN)-mediated transfection, DEAE Dextran- mediated transfection, polybrene-mediated transfection, electroporation and any other method of introducing DNA into a cell.
  • a viral expression vector may be introduced into a target cell in an expressible form by infection or transduction.
  • a viral vector includes, but is not limited to: a retrovirus, an adenovirus, a herpes virus and an avipox virus.
  • p94 RB When p94 RB is expressed in any abnormally proliferating cell, the cell replication cycle is arrested, thereby resulting in senescence and cell death and ultimately, reduction in the mass of the abnormal tissue, i.e., the tumor or cancer.
  • a vector able to introduce the gene construct into a target cell and able to express p94 RB therein in cell proliferation-suppressing amounts can be administered by any effective method.
  • a physiologically appropriate solution containing an effective concentration of active vectors can be administered topically, intraocularly, parenterally, orally, intranasally, intravenously, intramuscularly, subcutaneously or by any other effective means.
  • the vector may be directly injected into a target cancer or tumor tissue by a needle in amounts effective to treat the tumor cells of the target tissue.
  • a cancer or tumor present in a body cavity such as in the eye, gastrointestinal tract, genitourinary tract (e.g., the urinary bladder) , pulmonary and bronchial system and the like can receive a physiologically appropriate composition (e.g., a solution such as a saline or phosphate buffer, a suspension, or an emulsion, which is sterile except for the vector) containing an effective concentration of active vectors via direct injection with a needle or via a catheter or other delivery tube placed into the cancer or tumor afflicted hollow organ.
  • a physiologically appropriate composition e.g., a solution such as a saline or phosphate buffer, a suspension, or an emulsion, which is sterile except for the vector
  • Any effective imaging device such as X-ray, sonogra , or fiberoptic visualization system may be used to locate the target tissue and guide the needle or catheter tube.
  • a physiologically appropriate solution containing an effective concentration of active vectors can be administered systemically into the blood circulation to treat a cancer or tumor which cannot be directly reached or anatomically isolated.
  • target tumor or cancer cells can be treated by introducing p94 RB protein into the cells by any known method.
  • liposomes are artificial membrane vesicles that are available to deliver drugs, proteins and plasmid vectors both jln vitro or jLn vivo (Mannino, R.J. et al. , 1988, Biotechniques. 6:682-690) into target cells (Newton, A.C. and Huestis, W.H.,
  • p94 RB protein can be encapsulated at high efficiency with liposome vesicles and delivered into mammalian cells in vitro or in vivo.
  • Liposome-encapsulated p94 RB protein may be administered topically, intraocularly, parenterally, intranasally, intratracheally, intrabronchially, intramuscularly, subcutaneously or by any other effective means at a dose efficacious to treat the abnormally proliferating cells of the target tissue.
  • the liposomes may be administered in any physiologically appropriate composition containing an effective concentration of encapsulated p94 RB protein.
  • the gene construct and vectors of the present invention are effective in inhibiting the growth or mitosis or both of any type of tumor cell.
  • the gene construct of the invention has demonstrated effectiveness in treating tumor cells of carcinomas and sarcomas.
  • the gene construct of the invention has demonstrated effectiveness in suppressing replication and inducing cell senescence followed by cell death in the following tumor cell types: bladder carcinoma, lung carcinoma, breast carcinoma, prostate carcinoma, fibrosarcoma, osteosarcoma and cervix carcinoma.
  • the gene construct of the invention has demonstrated effectiveness in suppressing replication and inducing cell senescence followed by cell death in the tumor cells having the following identified genetic defects: tumor suppressor gene RB and p53 mutation, oncogene myc activation, and oncogene N-ras and c-yes-1 activation.
  • the gene construct of the invention has demonstrated effectiveness in suppressing replication and inducing cell senescence followed by cell death in the tumor cells having normal endogenous tumor suppressor RB 110 and/or p53 gene expression.
  • the gene construct of the invention is able to suppress replication in lymphomas, leukemia and in tumor cells having tumor suppressor gene DCC and NFl genetic defects, as well as in other tumor cell types in which the genetic defects are unknown or have yet to be identified. 3.3.5. Ex Vivo Treatment of Tumor or Cancer Tissues
  • a tumor cell is transduced with a retrovirus vector, an adenovirus vector, a plasmid vector or any other appropriate vector capable of expressing the p94 RB protein in that tumor cell.
  • the cancer cell may be present in a blood or bone marrow sample collected from a leukemia patient.
  • a dose of p94 RB protein expressing retrovirus vector or adenovirus vector or plasmid vector or any other appropriate vector is administered to the sample of blood or bone marrow at a dose sufficient to transduce enough cells in the sample to produce a reduction in tumor cell numbers.
  • the cell proliferation of the treated cancer cells will be slowed or terminated followed by a process similar to normal cellular differentiation or cell senescence.
  • Analo-gously, blood or bone marrow or other tissue is treated ex vivo using an effective dose of a lipsome- encapsulated p94 RB protein. Thereafter the sample may be returned to the donor or infused into another recipient.
  • a retroviral vector, an adenovirus vector, a plasmid vector, or any other appropriate vector capable of expressing the p94 RB protein can be administered in vivo to a cancer by a wide variety of manipulations. All such manipulations have in common the goal of placing the vector in sufficient contact with the target tumor to permit the vector to transduce or transfect the tumor cells.
  • cancers present in the epithelial linings of hollow organs may be treated by infusing the vector suspension into a hollow fluid filled organ, or by spraying or misting into a hollow air filled organ.
  • the tumor cell may be present in or among the epithelial tissue in the lining of pulmonary bronchial tree, the lining of the gastrointestinal tract, the lining of the female reproductive tract, genito-urinary tract, bladder, the gall bladder and any other organ tissue accessible to contact with the vector.
  • the cancer may be located in or on the lining of the central nervous system, such as, for example, the spinal cord, spinal roots or brain, so that vectors infused in the cerebrospinal fluid will contact and transduce the cells of the tumor in that space.
  • the cancer is a solid tumor.
  • the vector can be administered to the tumor by direct injection of the vector suspension into the tumor so that vectors will contact and transduce or transfect the tumor cells inside the tumor.
  • the cancer may be a cancer of the blood, blood forming organs or any organ directly perfused by the blood, so that vectors injected into the blood stream will contact and treat the cells of the cancer.
  • the cancer may be a leukemia, a lymphoma or other tumor type and the tumor cell may be present in the blood, the bone marrow, the spleen, the thymus, the liver and any other blood perfused organ.
  • the vector is administered in a composition comprising the vector together with a carrier or vehicle suitable for maintaining the transduction or transfection efficiency of the chosen vector and promoting a safe infusion.
  • a carrier may be a pH balanced physiological buffer, such as a phosphate, citrate or bicarbonate buffer, a saline solution, a slow release composition and any other substance useful for safely and effectively placing the vector in contact with abnormally proliferating cells to be treated.
  • a carrier may be a pH balanced physiological buffer, such as a phosphate, citrate or bicarbonate buffer, a saline solution, a slow release composition and any other substance useful for safely and effectively placing the vector in contact with abnormally proliferating cells to be treated.
  • AcMNPV Autographa California Multiple Nuclear Polyhedrosis Virus
  • the coding sequence of a foreign gene can easily be placed under the transcriptional control of the polyhedron promoter, resulting in a high level of expression.
  • such proteins may be correctly folded and contain appropriate post-translational modifications like those proteins in the native higher eukaryotes.
  • the resulted DNA molecule has the nucleotide sequence of Figure 1 (SEQ ID NO:l; SEQ ID NO:2), which is also referred to herein as the second in-frame AUG codon-initiated RB protein gene, or the p94 RB encoding gene.
  • the coded-for protein has the sequence of Figure 2 (SEQ ID NO:3) and is referred to herein as the second in-frame AUG codon-initiated RB protein, or the p94 RB protein.
  • the recombinant transfer vector was constructed with insertion of the p94 RB gene into the pVL1393 plasmid so that the p94 RB gene was placed under the control of the polyhedron gene promoter.
  • the resulting pVL-s-RB plasmid contains no additional AUG start codon upstream from the p94 M translation initiation site at nucleotide +19, and thus encodes a nonfusion p94 RB protein.
  • the same strategy was employed to construct a pllO* 3 expression vector which was designated pVL/lst AUG-RB.
  • Transfer of RB cDNAs from the recombinant vectors to the viral genome was accomplished by co-transfecting wild-type AcMNPV virus DNA with pVL-s- RB plasmid DNA or pVL/lst AUG-RB plasmid DNA.
  • the recombinant viruses were subjected to three rounds of plaque purification to obtain a pure stock of RB- containing baculovirus, designated AcMNPV-RB94 and AcMNPV-RBllO, respectively.
  • pllO 1 " 1 and p94 RB Proteins were purified from baculovirus-infected insect cells by immunoaffinity chromatography. Briefly, insect cells were harvested 24 hours after the virus infection and lysed at 4°C with EBC buffer (50 mM Tris-HCl, pH ⁇ .O, 120 mM NaCI, 0.5% NP-40, 50 / ig/ml aprotinin) .
  • EBC buffer 50 mM Tris-HCl, pH ⁇ .O, 120 mM NaCI, 0.5% NP-40, 50 / ig/ml aprotinin
  • the lysate was clarified by centrifugation and the pllO 8 - or p94 RB - containing supernatant was incubated with biotinylated WL-1 polyclonal anti-RB antibodies (Xu, H-J., et al., 1989, Oncogene, 4:807-812) at 4°C overnight.
  • biotinylation of rabbit IgGs using succinimide ester followed the methods described by Bayer and Wilchek (Baylor, E.A. and Wilchek, M. , 1980, Methods Biochem. Anal.. 26:1-45).
  • the RB protein-IgG- biotin complex was collected on a streptavidin agarose gel column. Purified pllO* 3 or p94 RB were eluted from separate columns using 100 mM glycine (pH 2.2) and neutralized with 1M of phosphate (pH 8.0).
  • the AcMNPV-RBllO and AcMNPV-RB94 infected Sf9 cells were immunostained with MAb-1 anti-RB monoclonal antibody 24h after infection. As shown in Figure 4, intense staining was found exclusively in the nuclei of cells infected with either AcMNPV-RBllO (panel B) or AcMNPV-RB94 (panel C).
  • pllO 1 ⁇ and p94 RB proteins purified from baculovirus-infected insect cells by immunoaffinity chromatography were tested for their ability to form a specific complex with SV40 T antigen. Briefly, equal amounts of p94 RB or pllO* 3 and T antigen were mixed and aliquots of the mixture were immunoprecipitated with PAB419 anti-T antibody. As shown in Figure 5, mixing of p94 RB (or pllOTM) with T antigen in vitro resulted in the co-immunoprecipitation of both under- and hypo- phosphorylated p94 RB (lane 5), or pllO 8 (lane 3) with PAB419.
  • pllO ⁇ or p94 RB protein can form a specific complex with SV40 T antigen.
  • the AcMNPV-RB94 virus-infected insect cells appear to make hyperphosphorylated p94 RB (lane 4) , which was unable to form complexes with SV40 T antigen (compare lane 4 with lane 5) .
  • the second in-frame AUG codon-initiated p94 RB protein produced in recombinant virus-infected insect cells is a artificial but stable nuclear phosphoprotein with its under- and hypo-phosphorylated forms being able to assemble specific complex with SV40 T antigen, as does the naturally occurring RB protein species, pllO 1 ⁇ .
  • RB retinoblastoma
  • Hind II, at nucleotide +7 and the restriction enzyme, Seal, at nucleotide 3,230 The A of the second in-frame AUG codon of the full length RB cDNA open reading frame was designated nucleotide +19).
  • the resulted 3,230 bp RB cDNA fragment had two blunt ends. Conversion of the blunt ends to restriction enzyme BamHI sites was done by ligation of a synthetic BamHI oligonucleotide linker (GGGATCCC) to each blunt end of the fragment followed by digestion with the BamHI enzyme.
  • GGGATCCC synthetic BamHI oligonucleotide linker
  • the desired RB cDNA fragment was inserted into the BamHI cloning site of a plasmid vector, pUC19, and propagated in the Escherichia coli strain, DH5 alpha bacterial cells.
  • the recombinant plasmid was purified from a single DH5 alpha transformant and designated plasmid pUC-s-RB.
  • This plasmid contains the desired RB cDNA fragment of 3,230 bp coding for the second in-frame AUG codon-initiated RB protein of 816 amino acids.
  • RB cDNA plasmid Encoding the First In-Frame AUG Codon-initiated RB Protein.
  • the full length RB cDNA plasmid was digested with the restriction enzyme, Acyl at nucleotide -322 and Seal at nucleotide 3,230.
  • the Acyl ends (overhang 5'-CG) were repaired by "filling in” the ends with the Klenow fragment of E. coli DNA polymerase I in the presence of all 4 dNTPs to generate blunt ends. Conversion of the blunt ends to restriction enzyme BamHI sites was done as described above.
  • the resulted RB cDNA fragment of 3,552 bp was inserted into the plasmid pUC19 and propagated in the Escherichia coli strain DH5 alpha, which was subsequently purified from a single DH5 alpha transformant and designated plasmid pUC-f-RB.
  • This plasmid contains the RB cDNA fragment of 3,552 bp coding for the first in-frame AUG codon- initiated RB protein of 928 amino acids.
  • the RB cDNA fragment of 3,230 bp coding for the second in-frame AUG codon-initiated RB protein of 816 amino acids (p94 RB ) was recovered from plasmid pUC-s-RB following the restriction enzyme, BamHI digestion, and re-inserted into the unique BamHI site of an expression plasmid, pHBAPr-1-neo (Gunning, P., et al., Proc. Natl. Acad. Sci.. USA. 1987, 84:4831-4835) in a orientation that the p94 RB coding sequence was under the direct control of the ⁇ -actin gene promoter.
  • a plasmid vector with the correct insert orientation was selected by restriction endonuclease mapping after propagation in DH5 alpha Escherichia coli host cells, and was designated p ⁇ A-s-RB34 ( Figure 7B) .
  • the corresponding DH5 alpha strain that contains plasmids P/5A-S-RB34 was thereafter designated DHB-S-RB34 (ATCC 69241, patent depository, American Type culture Collection) .
  • the plasmid vector p ⁇ A-s-RB34 contains no additional AUG codon between the ⁇ -actin gene promoter and the second in-frame AUG codon of the RB coding sequence, and thus encodes a non-fusion p94 RB protein.
  • the plasmid vector p ⁇ A-s-RB34 also confers a dominant selectable marker (geneticin resistance) in eukaryotic cells through expression of the neomycin phosphotransferase (neo) under separate control of an SV40 early promoter ( Figure 7, sv-neo) .
  • the RB cDNA fragment of 3,552 bp coding for the first in-frame AUG codon-initiated RB protein of 928 amino acids (pllO RB ) was recovered from plasmid pUC-f-RB and re-inserted into the expression plasmid pHBAPr-1-neo downstream from the ⁇ -actin gene promoter.
  • the resulting plasmid vector was designated p ⁇ A-f-RB33 ( Figure 7A) .
  • the plasmid vector p ⁇ A-f-RB33 contains no additional AUG codon between the ⁇ -actin gene promoter and the first in-frame AUG codon of the RB coding sequence, and thus encodes a non-fusion pllO* 8 protein.
  • an expression plasmid pCMV-Neo- Bam (Baker, S.J., et al., Science. 1990, 249:912-915) was used in place of plasmid pH ⁇ APr-1-neo.
  • the vector included cytomegalovirus (CMV) promoter/enhancer sequences, which could drive expression of the insert at the BamHI site, and splicing and polyadenylation sites derived from the rabbit ⁇ -globin gene, which ensured proper processing of the transcribed insert in the cells.
  • CMV cytomegalovirus
  • a pBR322 origin of replication and ⁇ - lactamase gene facilitated propagation of the plasmid in E. coli .
  • the plasmid conferred geneticin resistance (a selectable marker in eukaryotic cells) through expression of the neomycin phosphotransferase (neo) under the control of a herpes simplex virus (HSV) thymidine kinase promoter.
  • HSV herpes simplex virus
  • retroviral vector pLLRNL (Miller, A.D., Law, M.-F., Verma, I.M., Molec. Cell Biol. , 1985, 5:431) and amphotropic retrovirus packaging cell line, PA317 (ATCC CRL9078) (Miller, A.D., and Buttimore, C. , Molec. Cell Biol.. 1986, 6:2895-2902) are used.
  • a plasmid p4.95BT or F7 containing the full-length RB gene cDNA is digested with the restriction enzyme Hind II at nucleotide +7 (the A of the second in-frame AUG codon of the full-length RB cDNA open reading frame was designated nucleotide +19) .
  • Conversion of the Hind II site to restriction enzyme Hind III site is done by ligation of a synthetic Hind III oligonucleotide linker (CCAAGCTTGG) to the blunt ends of the linear plasmid DNA, followed by digestion with the Hind III enzyme.
  • the linear plasmid DNA is further digested with restriction enzyme, Seal, at nucleotide 3,230.
  • the resulted RB cDNA fragment of 3,230 bp codes for the second in-frame AUG codon-initiated RB protein of 816 amino acids (p94 ⁇ ) .
  • This fragment has a 5'-Hind III site (cohesive end) and a 3'-Seal site (blunt end) , which facilitates its insertion into the retroviral vector, pLLRNL.
  • the vector pLLRNL is digested with two sets of restriction enzymes: Hind III/ClaI and Smal/Clal to delete the luciferase gene. Appropriate fragments are recovered from the agarose gel following electrophoresis, and ligated with the RB cDNA fragment of 3,230 bp to form a new vector, pLRB94RNL, in which the p94 RB expression is under the control of the long terminal repeat sequences of Moloney murine leukemia virus (MuLV LTRs) .
  • the basic protocol for construction of the retroviral vector, pLRB94RNL is modified from Huang, H.-J.S., et al., 1988, Science. 242:1563-1566.
  • the vector pLLRNL is digested with a single restriction enzyme, Hind III, to delete the luciferase gene, as well as the Rous sarcoma virus promoter (RSV) .
  • An appropriate DNA fragment is recovered from the p94 RB expression plasmid, pCMV-s-RB42 (or p ⁇ A-s-RB34) .
  • the recovered DNA fragment which contains the 3,230 bp RB cDNA fragment and 5'-flanking CMV promoter (or ⁇ -actin promoter), is inserted into the Clal restriction site of the retroviral vector. Conversion between the restriction enzyme sites is done by the methods as described ⁇ upra in Section 4.3.7.
  • the p94 RB gene is under the control of an internal promoter (the CMV promoter or ⁇ -actin promoter) , while the Tn5 neomycin-resistance gene (Neo) is under the control of the MuLV LTRs.
  • An internal promoter the CMV promoter or ⁇ -actin promoter
  • Teo Tn5 neomycin-resistance gene
  • a safe and efficient amphotropic packaging cell line is necessary for transfer of retroviral vector genes into human cancer cells.
  • the virus packaging methods are modified from the method of Miyanohara et al., Proc. Natl. Acad. Sci. , USA. 1988, 85:6538-6542. For this protocol, the PA317 packaging cell line is used. This packaging cell line has received prior approval for use in human gene therapy clinical trials.
  • the retroviral vector (pLRB94RNL) DNA is transfected into PA317 packaging cells by LIPOFECTIN reagent (GIBCO BRL Life Technologies, Inc., Gaithersburg, MD) or electroporation methods as described in Sections 4.4.1. infra . Single colonies are isolated by selection in G418-containing medium (400 ⁇ g/ml) and expanded into mass cultures.
  • PA317 clones To titer the virus produced by selected PA317 clones, dilutions of cell-free culture medium from each PA317 clone are applied to 208F rat fibroblasts (indicator cells) in the presence of POLYBRENE (Sigma, 4 ⁇ g/ml) and G418 selection (400 ⁇ g/ml) is started 24 hours after infection. After two weeks, G418-resistant colonies are visualized by Giemsa staining and viral titers are determined (colony-forming units per milliliter, cfu/ml) . PA317 clones producing high virus titers are then assayed for human p94 RB protein expression by Western immunoblotting as described previously
  • the recombinant adenovirus Ad-RB94 is constructed from the adenovirus type 5 (Ad5) deletion mutant,
  • Ad-dl324 and a plasmid, pTG5955, in which the human
  • CFTR cDNA has been replaced by the human RB cDNA fragment of 3,230 bp coding for the p94 RB protein.
  • the plasmid pTG5955 containing the RB cDNA insert is linearized by restriction enzyme Clal cleavage and co-transfected with the large fragment of Clal-cut Ad-dl324 DNA into 293 (ATCC CRL1573) cells to allow homologous recombination to occur, followed by replication and encapsidation of recombinant adenoviral DNA into infectious virions and the formation of plaques.
  • Ad-RB94 adenovirus plaques containing the human RB cDNA
  • Ad-RB94 viruses are propagated in 293 cells and recovered 36 hours after infection.
  • the viral preparation is purified by CsCl density centrifugation, and stored in virus dialysis buffer (10 mM Tris-Hcl, pH7.4; 1 mM MgCl 2 ) at 4°C for immediate use; or stored at -70°C prior to use (with the addition of 10% glycerol) .
  • the basic protocol for construction of the recombinant adenovirus Ad-RB94 is modified from Rosenfeld, M.A., et al.. Cell. 1992, 68:143-155.
  • the p94 RB expressing plasmid vectors pCMV-s-RB42 or pBA-s-RB34 are used to form complexes with liposomes, and directly treat tumor cells in vivo (or ex vivo) . In this procedure, as described in
  • Human tumor cells having known RB gene deficiencies were treated with the p94 RB plasmid vector p ⁇ A-s-RB34 (or pCMV-s-RB42) .
  • These include: 1) human bladder carcinoma cell line, 5637, (ATCC HTB9) ; 2) human breast carcinoma cell line, MDA-MB-468 (ATCC HTB132) ; 3) human non-small cell lung carcinoma cell line, H2009 (Kratzke, R.A. , et al., 1992, The Journal of Biological Chemistry.
  • human prostate carcinoma cell line DU145 (ATCC HTB81) ; 5) human osteosarcoma cell line, Saos2 (ATCC HTB85) ; and 6) human fibrosarcoma metastatic to lung cell line, HS913T (ATCC HTB152) .
  • tumor cells were transiently transfected with the plasmid DNA p ⁇ A-s-RB34 (or pCMV-s-RB42) via LIPOFECTIN reagent (GIBCO BRL Life Technologies, Inc. Gaithersberg, MD) . Similar results have been obtained from transfection using calcium phosphate or electroporation methods.
  • Tumor cells were seeded in 100-mm dishes in appropriate growth medium supplemented with serum. The cells were incubated at 37°C in a 5% C0 2 environment until the cells were 40-60% confluent. This usually took 18-24 hours, but the time varied among cell types.
  • Solution A for each dish of cells to be transfected, 5-10 ⁇ g of plasmid DNA were diluted into a final volume of lOO ⁇ l with serum-free medium;
  • Solution B for each dish of cells to be transfected, 30-50 ⁇ l of LIPOFECTIN reagent was diluted into a final volume of lOO ⁇ l with serum- free medium.
  • the two solutions were combined, mixed gently, and incubated at room temperature for 10-15 min.
  • the LIPOFECTIN reagent interacted spontaneously with plasmid DNA to form a lipid-DNA complex. While the lipid-DNA complex was forming, the cells were washed twice with 6 ml of serum-free medium.
  • tumor cells were transfected with the plasmid DNA p ⁇ A-f-RB33 or pCMV-f-RB35 which expresses the pllO* 8 .
  • the following assays were used to evaluate the growth inhibitory effects of introducing p94 RB versus pllO 0 expression in RB defective tumor cells:
  • the tumor cells were labeled with [ 3 H]-thymidine for 2 hours, then transferred to polylysine-coated slides, fixed and immunocytochemically stained with a monoclonal anti-RB antibody, MAb-l (Triton Biosciences, Inc. Alameda, CA) .
  • MAb-l Monoclonal anti-RB antibody
  • the RB-positive transfected cells were counted under the microscope.
  • the slides were then coated with Kodak NTB2 autoradiographic emulsion and exposed for 7-10 days.
  • the [ 3 H]-thymidine labeling and RB protein immunocytochemical staining were done according to the methods previously described (Xu et al., Oncogene. 1991, 6:1139-1146).
  • the tumor cells were replated at a density of 10 5 cells per 100 mm dish with selected medium containing G418 of 400-600 ⁇ g/ml. Cells were cultured for 2 to 3 weeks and colonies of >100 cells were scored. The data are illustrated in Table 3. Cells treated with plasmid vectors expressing p94 RB formed approximately four-fold fewer colonies than those transfected with pllO* 8 plasmid vectors. The difference was statistically significant (p ⁇ 0.05 by t-test) .
  • Table 3 Growth inhibitory effects of introducing pi 10TM and p94TM expression into RB-defective bladder carcinoma cell line, 5637 (HTB9). Each number represents 6 to 11 dishes.
  • the HTB9 transfectants were also immunostained with MAb-1 anti-RB monoclonal antibody about 24 hours after transfection. The staining results are illustrated in Figure 8.
  • Two RB+ human cell lines (i.e., having no RB gene defect) , including a human fibrosarcoma cell line, HT1080 (ATCC CCL121) , and human cervix carcinoma cell line, HeLa (ATCC CCL2) were treated with the p94 ⁇ protein expression plasmid, pCMV-s-RB42, using the LIPOFECTIN reagent as described supra .
  • these cell lines were also transfected with the pllO* 8 protein expression plasmid, pCMV-f-RB35.
  • the colony formation assay as described ⁇ upra was used to evaluate the growth inhibitory effects of introducing exogenous p94 RB versus pllO* 8 expression in RB + tumor cells.
  • Table 4 Growth inhibitory effects of introducing pi 10TM and p94TM expression into RB-positive human fibrosarcoma cell line, HT1080 and the RB positive human cervix carcinoma cell line, HeLa.
  • the RB expression was under the control of cytomegalovirus (CMV) promoter.
  • CMV cytomegalovirus
  • transfected bladder carcinoma cell line, 5637 (ATCC HTB9) was measured by pulse-labeling of transfected 5637 cells with [ 35 S]-methionine followed by a chase with excess unlabeled methionine ( Figure 9) .
  • the bladder tumor cells were transfected in multiple dishes with either pllO 8 ( Figure 9, left) or p94 RB ( Figure 9, right) expression plasmids. Twenty- four hours after transfection the cells were labeled with [ 35 S]-methionine and chased with excess unlabeled methionine for 0, 6, 12 and 24 hours, respectively. RB proteins were determined by immunoprecipitation.
  • p94 RB protein in the transfected 5637 cells was determined to be 12 hours. In contrast, the half-life of pllO* 3 protein was 4-6 hours. Therefore, p94 RB protein expressed in host tumor cells has a slower turnover, which is believed to contribute to its efficacy as a suppressor of both RB+ and RB- tumor cell replication.
  • the comparative phosphorylation states of pllO* 8 and p94 RB in transiently transfected 5637 cells were determined by Western blot analysis: cell-lysates were made from WI-38, parental 5637 and p ⁇ A-f-RB33 (expressing pllO* 8 , Section 4.3.5) or pBA-s-RB34
  • Tumor cells from RB minus human bladder carcinoma cell line, 5637 (ATCC HTB9) and RB + human bladder carcinoma cell line, SCaBER (ATCC HTB3) are injected directly into the bladders of female athymic (nu/nu) nude mice (6 to 8 weeks of age) by a catheter as initially reported by Jones and his colleagues (Ahlering, T.E., et al., Cancer Res. , 1987, 47:6660-6665) .
  • nude mouse bladder tumors Development and progression of the nude mouse bladder tumors are monitored using a fiber-optical system to which a TV monitor is attached.
  • the experimental tumors are subsequently treated with retrovirus vectors expressing the p94 RB .
  • Supernatants with high virus titers are obtained from tissue culture media of selected PA317 clones expressing high level of human p94 RB protein (Section 4.3.7) and confirmed as free of replication- competent virus prior to use.
  • the retroviral vector suspension at high titers ranging from 4 x 10 4 to greater than 1 x 10 7 colony-forming unit (cfu)/ml, and more preferably at a titer greater than 1 x 10 6 cfu/ml is then infused directly into the mouse bladders via a catheter to treat the tumors.
  • the skilled artisan will understand that such treatments can be repeated as many times as necessary via a catheter inserted into the bladder.
  • the tumor regression following transferring the p94 RB gene is monitored frequently via the fiber-optic system mentioned above.
  • NCI-H460 (ATCC HTB177) cells which have normal pllO* 8 expression are injected into the right mainstream bronchus of athymic (nu/nu) nude mice (10 5 cells per mouse) . Three days later the mice are inoculated endobronchically with supernatant from the p94 RB , or pllO* 8 retrovirus producer cells daily for three consecutive days. Tumor formation is suppressed in the group of mice treated with the p94 RB retrovirus supernatant. In contrast, in the other group, which is treated with pllO* 8 retrovirus supernatant, the majority of mice develop endobronchial tumors.
  • Non-small cell lung cancer patients having an endobronchial tumor accessible to a bronchoscope, and also having a bronchial obstruction, are initially selected for p94 RB gene therapy. Treatment is administered by bronchoscopy under topical or general anesthesia. To begin the procedure, as much gross tumor as possible is resected endoscopically. A transbronchial aspiration needle (21G) is passed through the biopsy channel of the bronchoscope.
  • the residual tumor site is injected with the appropriate retroviral vector supernatant (Section 4.3.7), adenovirus Ad-RB94 suspension (Section 4.3.8) or p94 RB -expressing plasmid vector- liposome complexes (Section 4.3.4 and 4.3.6) at a volume of 5 ml to 10 ml.
  • Protamine is added at a concentration of 5 ⁇ q/ml .
  • the injections of therapeutic viral or plasmid supernatant comprising one or more of the vectors are administered around and within the tumor or tumors and into the submucosa adjacent to the tumor. The injections are repeated daily for five consecutive days and monthly therafter. The treatment may be continued as long as there is no tumor progression. After one year the patients are evaluated to deteri e whether it is appropriate to continue therapy.
  • the patients wear a surgical mask for 24 hours following injection of the viral supernatant. All medical personnel wear masks routinely during bronchoscopy and injection of the viral supernatant. Anti-tussive is prescribed as necessary.
  • target tumor or cancer cells are treated by introducing p94 RB protein into cells in need of such treatment by any known method.
  • liposomes are artificial membrane vesicles that have been extensively studied for their usefulness as delivery vehicles of drugs, proteins and plasmid vectors both in vitro or in vivo (Mannino, R.J. et al., 1988, Biotechnigues. 6:682- 690) . Proteins such as erythrocyte anion transporter (Newton, A.C. and Huestis, W.H. , Biochemistry.
  • small-particle aerosols provide a method for the delivery of drugs for treatment of respiratory diseases.
  • drugs can be administered in small-particle aerosols by using liposomes as a vehicle. Administered via aerosols, the drugs are deposited rather uniformly on the surface of the nasopharynx, the traceheobronchial tree and in the pulmonary area (Knight, V. and Gilbert, B., 1988, European Journal of Clinical Microbiology and Infectious Diseases. 7:721-731) .
  • the therapeutic p94 RB protein is purified, for example, from recombinant baculovirus AcMNPV-RB94 infected insect cells by immunoaffinity chromatography (Sections 4.1 and 4.2) or any other convenient source.
  • the p94 RB protein is mixed with liposomes and incorporated into the liposome vesicles at high efficiency.
  • the encapsulated p94 RB is active. Since the aerosol delivery method is mild and well-tolerated by normal volunteers and patients, the p94 RB -containing liposomes can be administered to treat patients suffering from lung cancers of any stage and/or to prevent lung cancers in high-risk population.
  • the p94 RB protein- containing liposomes are administered by nasal inhalation or by a endotracheal tube via small- particle aerosols at a dose sufficient to suppress abnormal cell proliferation. Aerosolization treatments are administered to a patient for 30 minutes, three times daily for two weeks, with repetition as needed. The p94 RB protein is thereby delivered throughout the respiratory tract and the pulmonary area. The treatment may be continued as long as necessary. After one year the patent's overall condition will be evaluated to determine if continued therapy is appropriate.
  • the retroviral vector, pLRB94RNL, expressing p94 RB protein as described supra in section 4.3.7. was introduced into normal mouse fibroblast-derived retrovirus-packaging cell line, PA317 (ATCC CRL9078) by LIPOFECTIN reagent (GIBCO BRL Life Technologies, Inc., Gaithersburg, MD) . Single cell colonies were isolated by selection in G418-containing medium and expanded into mass cultures. These clonal cells had been maintained over a one-year period of continuous culture, and stably expressed high levels of p94 RB protein as determined by immunocytochemical staining ( Figure 11) or by Western immunoblotting.
  • the retroviral plasmid vector, pLRB94RNL, expressing p94 ⁇ protein as described supra in section 4.3.7. was mixed with DMRIE/DOPE Liposomes (VICAL, Inc., San Diego, CA) and infused directly into the mouse urinary bladders via a catheter. Forty-eight hours after treatment, the mice were sacrificed and bladders excised. As demonstrated by immunohistochemical staining of the p94 RB protein in paraffin-embedded tissue sections from the mouse bladders ( Figure 12) , the liposome-encapsulated p94 RB expressing retroviral plasmid vectors penetrated the mucosa of mouse bladders and expressed p94 RB protein in the great majority of the transitional cells.
  • transitional epithelia expressing the p94 RB were histologically normal ( Figure 12, panel D, arrows), and were indistinguishable from the mucosa in untreated mouse bladders or mouse bladders treated with liposomes only ( Figure 12) .
  • the results from such animal experiments strongly suggest that the p94 RB treatment, unlike the conventional cytotoxic cancer therapy, is non-toxic to normal tissues in vivo .
  • GAA GTA TTA CAA ATG GAA GAT GAT CTG GTG ATT TCA TTT CAG TTA ATG 339 Glu Val Leu Gin Met Glu Asp Asp Leu Val He Ser Phe Gin Leu Met 95 100 105
  • GAT GCA AGA TTA TTT TTG GAT CAT GAT AAA ACT CTT CAG ACT GAT TCT 723 Asp Ala Arg Leu Phe Leu Asp His Asp Lys Thr Leu Gin Thr Asp Ser 220 225 230 235
  • GGC AAC TTG ACA AGA GAA ATG ATA AAA CAT TTA GAA CGA TGT GAA CAT 1347 Gly Asn Leu Thr Arg Glu Met He Lys His Leu Glu Arg Cys Glu His 430 435 440
  • ACG CGT GTA AAT TCT ACT GCA AAT GCA GAG ACA CAA GCA ACC TCA GCC 1587 Thr Arg Val Asn Ser Thr Ala Asn Ala Glu Thr Gin Ala Thr Ser Ala 510 515 520
  • AAA AAA AAA GTG TAT CGG CTA GCC TAT CTC CGG CTA AAT ACA CTT TGT GAA 1683 Lys Lys Val Tyr Arg Leu Ala Tyr Leu Arg Leu Asn Thr Leu Cys Glu 540 545 550 555 CGC CTT CTG TCT GAG CAC CCA GAA TTA GAA CAT ATC ATC TGG ACC CTT 1731 Arg Leu Leu Ser Glu His Pro Glu Leu Glu His He He Trp Thr Leu 560 565 570

Abstract

The present invention relates to a broad-spectrum tumor suppressor gene and the protein expressed by that gene in appropriate hostcells. The protein is a second in-frame AUG codon-initiated retinoblasoma protein of about 94 kD relative molecular mass. The present invention also relates to methods of treating a mammal having a disease or disorder characterized by abnormal cellular proliferation, such as a tumor or cancer and methods of treating abnormally proliferating cells, such as tumor or cancer cells. Treatment is accomplished by inserting a host cell compatible p94RB expression vector or an effective amount of p94RB protein into a cell or cells in need of treatment.

Description

BROAD SPECTRUM TUMOR SUPPRESSOR GENES, GENE PRODUCTS AND METHODS FOR TUMOR SUPPRESSION GENE THERAPY
This invention was made in part with United States government support under grant number EY06195 awarded by National Institutes of Health. The United States government has certain rights in the invention.
1. BACKGROUND OF THE INVENTION
1.1 Field of the Invention
This invention is in the field of tumor suppressor genes (anti-oncogenes) and relates in general to products and methods for practicing broad- spectrum tumor suppressor gene therapy of various human cancers. In particular, the invention relates to methods for treating tumor cells (1) administering vectors comprising a nucleic acid sequence coding for a second in-frame AUG codon-initiated retinoblastoma protein of about 94 kD or (2) administering an effective amount of a protein coded for by the nucleic acid sequence.
1.2 Cancer
Cancers and tumors are the second most prevalent cause of death in the United States, causing 450,000 deaths per year. One in three Americans will develop cancer, and one in five will die of cancer (Scientific American Medicine, part 12, I, 1, section dated 1987). While substantial progress has been made in identifying some of the likely environmental and hereditary causes of cancer, the statistics for the cancer death rate indicates a need for substantial improvement in the therapy for cancer and related diseases and disorders. 1.3. Cancer Genes
A number of so-called cancer genes, i.e., genes that have been implicated in the etiology of cancer, have been identified in connection with hereditary forms of cancer and in a large number of well-studied tumor cells. Study of cancer genes has helped provide some understanding of the process of tumorigenesis. While a great deal more remains to be learned about cancer genes, the presently known cancer genes serve as useful models for understanding tumorigenesis. Cancer genes are broadly classified into "oncogenes" which, when activated, promote tumorigenesis, and "tumor suppressor genes" which, when damaged, fail to suppress tumorigenesis. While these classifications provide a useful method for conceptualizing tumorigenesis, it is also possible that a particular gene may play differing roles depending upon the particular allelic form of that gene, its regulatory elements, the genetic background and the tissue environment in which it is operating.
1.3.1. Onco enes The oncogenes are somatic cell genes that are mutated from their wild-type alleles (the art refers to these wild-type alleles as protooncogenes) into forms which are able to induce tumorigenesis under certain conditions. There is presently a substantial literature on known and putative oncogenes and the various alleles of these oncogenes. In order to provide background information and to further the understanding of the scope of the invention, a brief discussion of representative oncogenes is provided.
For example, the oncogenes raε and myc are considered as models for understanding oncogenic processes in general. The raε oncogene is believed to encode a cytoplasmic protein, and the myc oncogene is believed to encode a nuclear protein. Neither the raε oncogene nor the myc oncogene alone is able to induce full transformation of a normal cell into a tumor cell, but full tumorigenesis usually occurs when both the ras and myc oncogenes are present and expressed together in the same cell (Weinberg, R.A., .1989, Cancer Research 49:3713-3721, at page 3713). Such collaborative effects have been observed between a number of other studied oncogenes.
The collaborative model of oncogene tumorigenesis must be qualified by the observation that a cell expressing the -ras oncogene that is surrounded by normal cells does not undergo full transformation. However, if most of the surrounding cells are also ras-expressing, then the ras oncogene alone is sufficient to induce tumorigenesis in a ras-expressing cell. This observation validates the multiple hit theory of tumorigenesis because a change in the tissue environment of the cell hosting the oncogene may be considered a second hit. An alternative and equally valid hypothesis is that events that collaborate with the activation of an oncogene such as raε or myc may include the inactivation of a negative regulatory factor or factors (Weinberg, R.A. , 1989, Cancer Research 49:3713-3721, at 3717; Goodrich, D.W. and Lee, W-H. , 1992, Nature 360:177-179), i.e., a tumor suppressor protein.
1.3.2. Tumor Suppressor Genes Tumor suppressor genes are genes that, in their wild-type alleles, express proteins that suppress abnormal cellular proliferation. When the gene coding for a tumor suppressor protein is mutated or deleted, the resulting mutant protein or the complete lack of tumor suppressor protein expression may fail to correctly regulate cellular proliferation, and abnormal cellular proliferation may take place, particularly if there is already existing damage to the cellular regulatory mechanism. A number of well- studied human tumors and tumor cell lines have been shown to have missing or nonfunctional tumor suppressor genes. Examples of tumor suppression genes include, but are not limited to, the retinoblastoma susceptibility gene or RB gene, the p53 gene, the deleted in colon carcinoma (DCC) gene and the neurofibromatosis type 1 (NF-1) tumor suppressor gene (Weinberg, R.A. Science, 1991, 254:1138-1146). Loss of function or inactivation of tumor suppressor genes may play a central role in the initiation and/or progression of a significant number of human cancers.
The list of putative tumor suppressor genes is large and growing. The following discussion of tumor suppressor genes is not intended to provide a complete review of all known and putative tumor suppressor genes, but is provided as background to indicate the state of the art and the problems to be overcome before the art is able to provide successful genetic therapy of diseases and disorders characterized by abnormally proliferating cells, e.g., tumor or cancer cells.
1.3.2.1. The Retinoblastoma Gene The RB gene is one of the better studied tumor suppressor genes. The size of the RB gene complementary DNA (cDNA) , about 4.7 Kb, permits ready manipulation of the gene, so that insertions of the RB gene have been made into a number of cell lines. The RB gene has been shown to be missing or defective in a majority of retinoblastomas, sarcomas of the soft tissues and bones, and in approximately 20 to 40 percent of breast, lung, prostate and bladder carcinomas (Lee, W-H. , et al., PCT Publ. No. WO 90/05180, at pages 38 and 39; see also, Bookstein, R. and Lee, W-H., 1991, Crit. Rev. Oncog. , 2:211-217; Benedict, W.F. et al., J. Clin. Invest.. 1990, 85:988-993) .
Based upon study of the isolated RB cDNA clone, the predicted RB gene product has 928 amino acids and an expected molecular weight of 106 kD (Lee et al., 1987, Nature, 329:642-645). The natural factor corresponding to the predicted RB gene expression product has been identified as a nuclear phosphoprotein having an apparent relative molecular mass (Mr) of 110-114 kD (Lee et al., 1987, Nature. 329:642-645) or 110-116 kD (Xu et al., 1989, Oncoσene 4:807-812). Hence, the literature generally refers to the protein encoded by the RB gene as pllO*8. In this connection, it is noteworthy that measurement of apparent relative molecular mass by SDS-PAGE is frequently inaccurate owing to protein secondary structure. Therefore, the full length RB protein of 928 amino acids is also referred to as the 115 kD (Yokota et al., 1988, Oncogene, 3:471-475), or 105 kD (Whyte et al . , 1988, Nature. 334:124-129) RB proteins. Various mutations of the RB gene are known. These are generally inactive. However, a 56 kD truncated RB protein, designated as pSδ3, that is considered to function in the same way as does pllO113 retains activity (Goodrich et al., 1992, Nature 360:177-179). on SDS-PAGE normal human cells show an RB protein pattern consisting of a lower sharp band with an Mr of 110 kD and a broader, more variable region above this band with an Mr ranging from 110 kD to 116 kD. The 110 kD band is the underphosphorylated RB protein, whereas the broader region represents the phosphorylated RB protein. The heterogeneity of the molecular mass results from a varying degree of phosphorylation (Xu et al., 1989, Oncogene, 4:807-812) .
The RB protein shows cyclical changes in phosphorylation. Most RB protein is unphosphorylated during Gl phase, but most (perhaps all) RB molecules are phosphorylated in S and G2 phases (Xu et al., 1989, Oncogene, 4:807-812; DeCaprio et al., 1989, Cell. 58:1085-1095; Buchkovich et al., 1989, Cell. 58:1097-1105; Chen et al., 1989, Cell. 58:1193-1198; Mihara et al., 1989, Science. 246:1300-1303).
Furthermore, only the underphosphorylated RB protein binds to SV40 large T antigen. Given that RB protein binding by large T antigen is probably important for the growth promoting effects of large T antigen, this suggests that the underphosphorylated RB protein is the active form of the RB protein, and the phosphorylated RB protein in S and G2 phases is inactive (Ludlow et al., 1989, Cell. 56:57-65).
The RB gene expressing the first in-frame AUG codon-initiated RB protein is also referred to herein as the intact RB gene, the RB110 gene or the pllO^ coding gene. It has also been observed that lower molecular weight (<100 kD, 98 kD, or 98-104 kD) bands of unknown origin which are immunoreactive to various anti-RB antibodies can be detected in immunoprecipitation and Western blots (Xu et al., 1989, Oncogene. 4:807-812; Furukawa et al., 1990, Proc. Natl. Acad. Sci.. USA. 87:2770-2774; Stein et al., 1990, Science. 249:666-669). Considering that the RB110 cDNA open reading frame sequence (McGee, T.L., et al., 1989, Gene. 80:119-128) reveals an in-frame second AUG codon located at exon 3, nucleotides 355-357, the deduced second AUG codon-initiated RB protein would be 98 kD, or 12 kD smaller than the pllO*8 protein. It has been proposed that the lower molecular weight bands are the underphosphorylated (98 kD) and phosphorylated (98-104 kD) RB protein translated from the second AUG codon of the RB mRNA (Xu et al., 1989, Oncogene, 4:807-812), although no data directly supported this hypothesis. Thus, no conclusive observation confirms the actual expression of the RB gene from the second in-frame AUG codon. Further, Sections 4.2.1, and Figure 5 infra provide data indicating the non-identity of the 98 kD protein bands of unknown origin and the second AUG codon-initiated protein products. it has been proposed that introduction of a functional RB110 gene into an RB-minus tumor cell will likely "normalize" the cell. Of course, it is not expected that tumor cells which already have normal RB110 gene expression ("RB+") will respond to RB110 gene therapy, because it is presumed that adding additional RB expression cannot correct a non-RB genetic defect. In fact, it has been shown that in the case of RB+ tumor cell lines, such as the osteosarcoma cell line, U-2 OS, which expresses the normal pllO*8, introduction of an extra pllO*3 coding gene did not change the neoplastic phenotype of such tumor lines (Huang, et al., 1988, Science. 242:1563-1566).
In the only reported exception, introduction of a pllO*8 coding vector into normal human fibroblasts, WS1, which have no known RB or any other genetic defects, led to the cessation of cell growth (WO 91/15580, Research Development Foundation, by Fung et al., PCT application filed 10 April 1991, published 17 October 1991, at page 18) . However, it is believed that these findings were misinterpreted since a plasmid, ppVUO-Neo, producing SV40 T antigen with a well-known growth-promoting effect on host cells was used improperly to provide a comparison with the effect of RBπo expression on cell growth of transfected WS1 fibroblasts (Fung, et al. Id. see Example 2 page 25) . This view is confirmed by the extensive literature, together with similar confirming data provided by the examples presented infra , clearly characterizing RB+ tumor cells as "incurable" by treatment with wild-type RB110 gene. In addition, it is noteworthy that the WS1 cell line per se is a generally recognized non-tumorigenic human diploid fibroblast cell line with limited cell division potential in culture. Therefore, WO91/15580 simply does not provide any method for effectively treating RB+ tumors with an RB110 gene. Thus, there remains a need for a broad-spectrum tumor suppressor gene for treating abnormally proliferating cells having any type of genetic defect.
1.3.2.2. The Neurofibromatosis Gene
Neurofibromatosis type 1 or von Recklinghausen neurofibromatosis results from the inheritance of a predisposing mutant allele or from alleles created through new germline mutations (C.J. Marshall, 1991, Cell. 64:313-326). The neurofibromatosis type 1 gene, referred to as the NF1 gene, is a relatively large locus exhibiting a mutation rate of around 104. Defects in the NF1 gene result in a spectrum of clinical syndromes ranging from cafe-au-lait spots to neurofibromas of the skin and peripheral nerves to Schwannomas and neurofibrosarcomas.
The NF1 gene encodes a protein of about 2485 amino acids that shares structural similarity with three proteins that interact with the products of the raε protooncogene (Weinberg et al., 1991, Science. 254:1138-1146 at page 1141). For example, the NF1 amino acid sequence shows sequence homology to the catalytic domain of raε GAP, a GTPase-activating protein for p21 raε (C.J. Marshall, 1991, Cell. 64:313-326 at pages 320 and 321).
The role of NFl in cell cycle regulation is apparently a complex one that is not yet fully elucidated. For example, it has been hypothesized that it is a suppressor of oncogenically activated p21 ras in yeast (C.J. Marshall, (1991, Cell. 64:313-326, bridging pages 320 and 321, and citing to Ballester et al, 1990, Cell. 63:851-859). On the other hand, other possible pathways for NFl interaction are suggested by the available data (C.J. Marshall, 1991, Cell. 64:313-326 at page 321; Weinberg et al., 1991, Science. 254:1138-1146 at page 1141). At present, no attempts to treat NFl cells with a wild-type NFl gene have been undertaken due to the size and complexity of the NFl locus. Therefore, it would be highly desirable to have a broad-spectrum tumor suppressor gene able to treat NFl and any other type of cancer or tumor.
1.3.3.3. The p53 Gene Somatic cell mutations of the p53 gene are said to be the most frequently mutated gene in human cancer (Weinberg et al., 1991, Science. 254:1138-1146 at page 1143) . The normal or wild-type p53 gene is a negative regulator of cell growth, which, when damaged, favors cell transformation (Weinberg et al. supra) . As noted for the RB protein, the p53 expression product is found in the nucleus, where it may act in parallel with or cooperatively with pllO8. This is suggested by a number of observations, for example, both p53 and pllO*8 proteins are targeted for binding or destruction by the oncoproteins of SV40, adenovirus and human papillomavirus.
Tumor cell lines deleted for p53 have been successfully treated with wild-type p53 vector to reduce tu origenicity (Baker, S.J., et al., 1990, Science. 249:912-915) . However, the introduction of either p53 or RBU0 into cells that have not undergone lesions at these loci does not affect cell 5 proliferation (Marshall, C.J., 1991, Cell, 64:313-326 at page 321; Baker, S.J., et al., 1990, Science. 249:912-915; Huang, H.-J.S., et al., 1988 Science. 242:1563-1566). Such experiments suggest that sensitivity of cells to the suppression of their
10 growth by a tumor suppressor gene is dependent on the genetic alterations that have taken place in the cells. Such a dependency would be further complicated by the observation in certain cancers that alterations in the p53 tumor suppressor or gene locus appear after
15 mutational activation of the ras oncogene (Marshall, C.J., 1991, Cell. 64:313-326; Fearon, E.R. , and Vogelstein, B. , 1990, Cell. 61:759-767).
Therefore, there remains a need for a broad- spectrum tumor suppressor gene that does not depend on
20 the specific identification of each mutated gene causing abnormal cellular proliferation.
1.3.3.4. The Deleted in Colon Carcinoma Gene (DCC.
_5 The multiple steps in the tumorigenesis of colon cancer are readily monitored during development by colonoscopy. The combination of colonoscopy with the biopsy of the involved tissue has uncovered a number of degenerative genetic pathways leading to the result
30 of a malignant tumor. One well studied pathway begins with large polyps of which 60% of the cells carry a mutated, activated allele of K-ras. A majority of these tumors then proceed to the inactivation-mutation of the gene referred to as the deleted in colon
35 carcinoma (DCC) gene, followed by the inactivation of the p53 tumor suppressor gene. The DCC gene is a more than approximately one million base pair gene coding for a 190-kD transmembrane phosphoprotein which is hypothesized to be a receptor (Weinberg et al., 1991, Science. 254:1138-1146 at page 1141), the loss of which allows the affected cell a growth advantage. It has also been noted that the DCC has partial sequence homology to the neural cell adhesion molecule (Marshall, 1991, Cell. 64:313-326) which might suggest a role for the DCC protogene in regulating cell to cell interactions. As can be appreciated, the large size and complexity of the DCC gene, together with the complexity of the K-ras, p53 and possibly other genes involved in colon cancer tumorigenesis demonstrates a need for a broad-spectrum tumor suppressor gene and methods of treating colon carcinoma cells which do not depend upon manipulation of the DCC gene or on the identification of other specific damaged genes in colon carcinoma cells.
1.4 Genetic Therapy: Gene Transfer Methods The treatment of human disease by gene transfer has now moved from the theoretical to the practical realm. The first human gene therapy trial was begun in September 1990 and involved transfer of the adenosine deaminase (ADA) gene into lymphocytes of a patient having an otherwise lethal defect in this enzyme, which produces immune deficiency. The results of this initial trial have been very encouraging and have helped to stimulate further clinical trials (Culver, K.W. , Anderson, W.F., Blaese, R.M. , Hum. Gene. Ther.. 1991, 2:107).
So far all but one of the approved gene transfer trials in humans rely on retroviral vectors for gene transduction. Retroviral vectors in this context are retroviruses from which all viral genes have been removed or altered so that no viral proteins are made in cells infected with the vector. Viral replication functions are provided by the use of retrovirus 'packaging' cells that produce all of the viral proteins but that do not produce infectious virus. Introduction of the retroviral vector DNA into packaging cells results in production of virions that carry vector RNA and can infect target cells, but no further virus spread occurs after infection. To distinguish this process from a natural virus infection where the virus continues to replicate and spread, the term transduction rather than infection is often used.
The major advantages of retroviral vectors for gene therapy are the high efficiency of gene transfer into replicating cells, the precise integration of the transferred genes into cellular DNA, and the lack of further spread of the sequences after gene transduction (Miller, A.D., Nature. 1992, 357:455-460) . The potential for production of replication- competent (helper) virus during the production of retroviral vectors remains a concern, although for practical purposes this problem has been solved. So far, all FDA-approved retroviral vectors have been made by using PA317 amphotropic retrovirus packaging cells (Miller, A.D., and Buttimore, C. , Molec. Cell Biol. , 1986, 6:2895-2902). Use of vectors having little or no overlap with viral sequences in the PA317 cells eliminates helper virus production even by stringent assays that allow for amplification of such events (Lynch, CM., and Miller, A.D., J. Viral.. 1991, 65:3887-3890). Other packaging cell lines are available. For example, cell lines designed for separating different retroviral coding regions onto different plasmids should reduce the possibility of helper virus production by recombination. Vectors produced by such packaging cell lines may also provide an efficient system for human gene therapy (Miller, A.D., 1992, Nature, 357:455-460).
Non-retroviral vectors have been considered for use in genetic therapy. One such alternative is the adenovirus (Rosenfeld, M.A. , et al., 1992, Cell.
68:143-155; Jaffe, H.A. et al., 1992, Nature Genetics 1:372-378; Lemarchand, P. et al., 1992, Proc. Natl. Acad. Sci. USA. 89:6482-6486). Major advantages of adenovirus vectors are their potential to carry large segments of DNA (36 Kb genome) , a very high titre (1011 ml"1) , ability to infect non-replicating cells, and suitability for infecting tissues in situ, especially in the lung. The most striking use of this vector so far is to deliver a human cystic fibrosis transmembrane conductance regulator (CFTR) gene by intratracheal instillation to airway epithelium in cotton rats (Rosenfeld, M.A. , et al.. Cell, 1992, 63:143-155). Similarly, herpes viruses may also prove valuable for human gene therapy (Wolfe, J.H., et al., 1992, Nature Genetics. 1:379-384). Of course, any other suitable viral vector may be used for genetic therapy with the present invention.
The other gene transfer method that has been approved by the FDA for use in humans is the transfer of plasmid DNA in liposomes directly to human cells in situ (Nabel, E.G., et al., 1990, Science. 249:1285- 1288) . Plasmid DNA should be easy to certify for use in human gene therapy because, unlike retroviral vectors, it can be purified to homogeneity. In addition to liposome-mediated DNA transfer, several other physical DNA transfer methods such as those targeting the DNA to receptors on cells by complexing the plasmid DNA to proteins have shown promise in human gene therapy (Wu, G.Y., et al., 1991, J. Biol. Chem.. 266:14338-14342; Curiel, D.T., et al., 1991, Proc. Natl. Acad. Sci. USA. 88:8850-8854). 1.5 Proposed Strategies for Cancer Gene Therapy It has been observed that certain tumor cells return to normal function when fused with normal cells, suggesting that replacement of a missing factor, such as a wild-type tumor suppressor gene expression product may serve to restore a tumor cell to a normal state (reviewed by Weinberg, R.A. , 1989, Cancer Research 49:3713-3721, at 3717).
These observations have led to research aimed at providing genetic treatment of tumor cells having defective tumor suppressor genes. The proposed method of treatment requires identification of the damaged tumor suppressor gene, and introduction of the corresponding undamaged gene (including a promoter and a complete encoding sequence) into the affected tumor cells by means of a vector such as a retrovirus able to express the gene product. It is proposed that the incorporated functional gene will convert the target cell to a non-malignant state. For example, The Regents of the University of California, in Patent Cooperation Treaty patent application (by Lee et al., number WO 90/05180, having an international filing date of 30 October 1989 and published 17 May 1990) , disclose a scheme for identifying an inactive or defective tumor suppressor gene and then replacing such a defective gene with its functional equivalent. In particular, the WO 90/05180 application proposes, based on .in vitro studies, to insert a functional RBU0 gene into an RB-minus tumor cell by means of a retroviral vector in order to render such cells non-malignant.
In addition, international application WO 89/06703 (by Dryja et al. , having an international filing date of 23 January 1989, and published 27 July 1989) proposes the treatment of retinoblastoma defective tumors by administering a retinoblastoma gene expression product. In this connection, it has been reported that the introduction of the RB110 gene into RB-minus retinoblastoma, osteosarcoma, bladder and prostate carcinoma cells resulted in cells showing reduced tumorigenicity in nude mice, but probably not a reduced cell growth rate. The results varied depending on the particular parental cell line (Goodrich et al., 1992, Cancer Research 52:1968-1973; Banerjee, A., et al., 1992, Cancer Research. 52:6297- 6304; Takahashi, R. , et al., 1991, Proc. Natl. Acad. Sci.. USA. 88:5257-5261; Xu, H-J. , et al., 1991, Cancer Research. 51:4481-4485; Bookstein et al, 1990, Science. 247:712-715; Huang, H-J.S., et al., 1988, Science 242, 1563-1566). However, the suppression of tumorigenicity by introduction of the pllO1^ coding gene into RB-minus tumor cells is incomplete. The pllO*8 reconstituted tumor cells still form invasive tumors in nude mice (Xu, H-J., et al., 1991, Cancer Research. 51:4481-4485; Takahashi, R. , et al., 1991, Proc. Natl. Acad. Sci.. USA. 88:5257-5261; Banerjee, A., et al., 1992, Cancer Research. 52:6297-6304). In particular, it has been shown that pllO*8 reconstituted retinoblastoma cells inoculated into an orthotopic site (in this instance, the eye) consistently produced tumors (Xu, H-J., et al., 1991, Cancer Research 51:4481-4485) . These findings, which will be discussed in detail infra , caution that the tumor suppressor gene replacement therapy as heretofore envisioned may simply result in cells that only appear to be "cured". Certainly, the findings of Xu et al. indicate a need for an improved genetic therapy for tumors which avoids these shortcomings.
Another proposed method of treating cancer by gene therapy is to antagonize the function of an oncogene by placing an artificial gene, constructed to have an inverted nucleotide sequence compared to the oncogene, into a tumor cell (U.S. patent number 4,740,463, issued April 26, 1988 by Weinberg, et al.).
All of these proposed solutions also share the deficiency of requiring that the specific genetic defect of the tumor to be treated be identified prior to treatment.
Since the pllO*3 protein product is active in the underphosphorylated state (discussed in detail supra) , and phosphoamino acid analysis has demonstrated only phosphoserine and phosphothreonine but not phosphotyrosine in RB protein (Shew, J-Y., et al., 1989, Ocogene Research. 1:205-213), it has been proposed to make a mutant RB protein with its serine or threonine residues being replaced by alanine or valine or others and that introduction of such a mutant, unphosphorylated RB protein into target cells may lead to growth arrest (International Application WO 91/15580, Research Development Foundation, by Fung et al., at page 20). Unfortunately, in all cases analyzed so far, the human RB protein carrying a point mutation and retaining the unphosphorylated state were invariably inactive proteins and associated with tumorigenesis rather than tumor suppression (Templeton et al., 1991, Proc. Natl. Acad. Sci. , USA, 88:3033- 3037.
1.6 Tumor Suppressor Gene Resistance As the above discussion of gene mutations in tumor cells has indicated, not every cancer gene is a suitable candidate for wild-type gene replacement therapy due to the gene size or complexity or for other reasons. The retinoblastoma gene is one of those tumor suppressor genes that is readily accessible to study, thus it provides a model for understanding some of the other disadvantages to cancer gene replacement therapy as heretofore understood. It is known that reintroduction of the retinoblastoma tumor suppressor gene into RB-defective tumor cells inhibits the tumor cell growth and suppresses the neoplastic phenotype of the target cells (WO 90/05180, cited supra ; Huang et al., 1988, Science. 242:1563-1566; Bookstein et al., 1990, Science. 247:712-715; Xu et al., 1991, Cancer Res.. 51:4481-4485; Takahashi et al., 1991, Proc. Natl. Acad. Sci.. USA. 88:5257-5261; Goodrich et al. , 1992, Cancer Res.. 52:1968-1973; Banerjee et al., 1992, Cancer Res.. 52:6297-6304).
However, the suppression of tumorigenicity is often incomplete. A significant percentage of the RB- reconstituted tumor cells still form small tumors after a longer latency period in nude mouse tumorigenicity assays. Such tumors, although retaining normal RB expression, are histologically malignant and invasive (Xu et al., 1991, Cancer Res. , 51:4481-4485; Takahashi et al., 1991, Proc. Natl. Acad. Sci.. USA. 88:5257-5261; Banerjee et al., 1992, Cancer Res.. 52:6297-6304).
Furthermore, it has been observed that several cell lines derived from such RB-positive tumors have become very tumorigenic and have formed large, progressively growing tumors when subsequently injected into nude mice (Zhou, Y. ; Li, J. ; Xu, K. ; Hu, S-X. ; Benedict, W.F., and Xu, H-J., Proc. Am. Assoc. Cancer Res.. 34:3214, 1993). This phenomenon, which is referred to herein as tumor suppressor gene resistance (TSGR) is a serious obstacle to the successful implementation of any scheme of tumor suppressor gene therapy for human cancers.
Without wishing to be bound by any particular hypothesis or explanation of the TSGR phenomenon, it is believed that the RB gene product exemplifies a possible explanation for TSGR. RB proteins have an active form (underphosphorylated protein) and an inactive form (phosphorylated protein) . Therefore, RB-positive tumor cells may have inherited or acquired the ability to phosphorylate RB proteins to the inactive state and allow tumor cell proliferation to continue. Thus, conversion of RB-minus cells with plasmid or virus vectors coding for the pllO*8 protein provides only incomplete suppression, or even exacerbation of a percentage of the malignant cell population because the pllO*8 protein remains phosphorylated and inactive in some of the target cells.
Alternatively, the tumor cells expressing the RB110 gene may simply have again inactivated the RB110 gene by mutation in subsequent cell divisions (Lee et al., 1990, Immunol. Ser. 51:169-200, at page 188) . Thus, there remains a need for a method of treating tumor cells by gene therapy so that the possibility of further mutation and resurgence of malignancy is avoided.
1.7 Summary of Obstacles to Cancer Gene Therapy In brief, there are at least three major obstacles to be overcome to achieve a practical tumor suppressor gene therapy for tumor cells: i) The necessity to determine the identity and sequence of each defective tumor suppressor gene or oncogene before attempting genetic therapy of that tumor. This is particularly a problem considering the multiple genetic defects found in many tumor cells studied;
2) The size and complexity of certain tumor suppressor genes or oncogenes renders manipulation of certain of these genes difficult; and
3) The possibility that TSGR as described above for the RB110 model system will generate tumor cells that have equal or greater dysfunction than did the original abnormal cells.
Accordingly, there is a need in the art for a genetic therapy for tumor or cancer cells which can safely overcome these problems and provide an effective treatment for all types of tumor cells without the need to determine the exact genetic deficiency of each treated tumor cell and without the risk of TSGR resurgence and exacerbation of the malignancy.
2. SUMMARY OF THE INVENTION
Obstacles to the successful practice of tumor suppressor gene therapy of cancers are avoided by the present invention. In a totally unexpected and surprising discovery, it has been determined that the second in-frame AUG codon-initiated retinoblastoma suppressor protein of about 94 kD (p94RB) is a broad- spectrum tumor suppressor, and that insertion of a gene capable of expressing this protein, or the protein itself, into an abnormally proliferating cell, such as a cancer or tumor cell, causes that cell to enter a senescent-like state, terminating the proliferation. The cell so-treated simply stops replicating and dies. The cell may possess any type of genetic defect, known or unknown, so that there is no need to determine the exact nature of the genetic defect associated with the abnormal proliferation. Further, the population of treated cells exhibits an unexpectedly much lower incidence of TSGR resurgence and exacerbation of malignancy than do cells treated with any other tumor suppressor gene. The method is repeated as needed.
Therefore, the invention provides p94RB encoding vectors and p94RB proteins for use in treatment of tumors or cancers, and methods of preparing p94RB proteins suitable for use in methods of treatment. The invention also provides methods of treatment for mammals such as humans, as well as methods of treating abnormally proliferating cells, such as cancer or tumor cells. Broadly, the invention contemplates treating abnormally proliferating cells, or mammals having a disease characterized by abnormally proliferating cells by any suitable method known to permit a host cell compatible p94RBencoding vector or a p94ω protein to enter the cells to be treated so that suppression of proliferation is achieved.
In one embodiment, the invention comprises a method of treating a disease characterized by abnormally proliferating cells, in a mammal, by administering an expression vector coding for p94RB to the mammal having a disease characterized by abnormally proliferating cells, inserting the expression vector into the abnormally proliferating cells, and expressing p94RB in the abnormally proliferating cells in an amount effective to suppress proliferation of those cells. The expression vector is inserted into the abnormally proliferating cells by viral infection or transduction, liposome-mediated transfection, polybrene-mediated transfection, CaP04 mediated transfection and electroporation. The treatment is repeated as needed.
In another embodiment, the invention comprises a method of treating abnormally proliferating cells of a mammal by inserting a p94RB encoding expression vector into the abnormally proliferating cells and expressing p94RB therein in amounts effective to suppress proliferation of those cells. The treatment is repeated as needed.
In another alternative embodiment, the invention provides a DNA molecule able to suppress growth of an abnormally proliferating cell. The DNA molecule encodes a p94RB protein having an amino acid sequence substantially according to SEQ ID NO:3, provided that the DNA molecule does not also code for a pllORB protein. In a more preferred embodiment, the DNA molecule has the DNA sequence of SEQ ID NO:l, and is expressed by an expression vector. The expression vector may be any host cell-compatible vector. The vector is preferably selected from the group consisting of a retroviral vector, an adenoviral vector and a herpesviral vector. In another alternative embodiment, the invention provides a p94RB protein having an amino acid sequence substantially according to SEQ ID NO:3.
In another alternative embodiment, the invention provides a method of producing a p94RB protein by the steps of: inserting a compatible expression vector comprising a p94RB encoding gene into a host cell and causing the host cell to express p94RB protein.
In another alternative embodiment, the invention comprises a method of treating abnormally proliferating cells of a mammal ex vivo by the steps of: removing a tissue sample in need of treatment from a mammal, the tissue sample comprising abnormally proliferating cells; contacting the tissue sample in need of treatment with an effective dose of an p94RB encoding expression vector; expressing the p94RB in the abnormally proliferating cells in amounts effective to suppress proliferation of the abnormally proliferating cells. The treatment is repeated as necessary; and the treated tissue sample is returned to the original or another mammal. Preferably, the tissue treated ex vivo is blood or bone marrow tissue.
In another alternative embodiment, the invention comprises a method of treating a disease characterized by abnormal cellular proliferation in a mammal by a process comprising the steps of administering p94RB protein to a mammal having a disease characterized by abnormally proliferating cells, such that the p94RB protein is inserted into the abnormally proliferating cells in amounts effective to suppress abnormal proliferation of the cells. In a preferred embodiment, the p94RB protein is liposome encapsulated for insertion into cells to be treated. The treatment is repeated as necessary.
In another alternative embodiment the invention comprises a method of treating abnormally proliferating cells of a mammal ex vivo by a process comprising the steps of removing a tissue sample in need of treatment from a mammal, the tissue sample comprising abnormally proliferating cells contacting the tissue sample in need of treatment with an effective dose of a p94RB protein. The treatment is repeated as necessary, and then the treated tissue is returned to the mammal or placed into another mammal.
In a more preferred embodiment the tumor or cancer cells to be treated are cells having one or more genetically defective tumor suppressor genes and oncogenes selected from the group consisting of an RB, a p53, a c-myc, an N-ras and a c-yes-1 gene.
In a more preferred embodiment the tumor or cancer cells are cells having no detectable genetic defect of a tumor suppressor gene selected from the group consisting of an RB gene and a p53 gene.
In a still more preferred embodiment the tumor or cancer cells are lung carcinoma cells.
In a still more preferred embodiment the p94RB encoding expression vector or the p94RB protein are administered by means of aerosol delivery of liposome- encapsulated p94RB encoding expression vector or p94RB protein into a lung in need of such treatment. 3. DETAILED DESCRIPTION OF THE INVENTION 3.1 Definitions
The terms "cancer" or "tumor" are clinically descriptive terms which encompass a myriad of diseases characterized by cells that exhibit unchecked and abnormal cellular proliferation. The term "tumor", when applied to tissue, generally refers to any abnormal tissue growth, i.e., excessive and abnormal cellular proliferation. A tumor may be "benign" and unable to spread from its original focus, or
"malignant" and capable of spreading beyond its anatomical site to other areas throughout the hostbody. The term "cancer" is an older term which is generally used to describe a malignant tumor or the disease state arising therefrom. Alternatively, the art refers to an abnormal growth as a neoplasm, and to a malignant abnormal growth as a malignant neoplasm.
Irrespective of whether the growth is classified as malignant or benign, the causes of excessive or abnormal cellular proliferation of tumor or cancer cells are not completely clear. Nevertheless, there is persuasive evidence that abnormal cellular proliferation is the result of a failure of one or more of the mechanisms controlling cell growth and division. It is also now believed that the mechanisms controlling cell growth and division include the genetic and tissue-mediated regulation of cell growth, mitosis and differentiation. These mechanisms are thought to act at the cell nucleus, the cell cytoplasm, the cell membrane and the tissue-specific environment of each cell. The process of transformation of a cell from a normal state to a condition of excessive or abnormal cellular proliferation is called tumorigenesis. It has been observed that tumorigenesis is usually a multistep progression from a normal cellular state to, in some instances, a full malignancy. It is therefore believed that multiple "hits" upon the cell regulatory mechanisms are required for full malignancy to develop. Thus, in most instances, it is believed that there is no single cause of excessive proliferation, but that these disorders are the end result of a series of cumulative events.
While a malignant tumor or cancer capable of unchecked and rapid spread throughout the body is the most feared and usually the deadliest type of tumor, even so-called benign tumors or growths can cause significant morbidity and mortality by their inappropriate growth. A benign tumor can cause significant damage and disfigurement by inappropriate growth in cosmetically sensitive areas, or by exerting pressure on central or peripheral nervous tissue, blood vessels and other critical anatomical structures.
A broad-spectrum tumor suppressor gene is a genetic sequence coding for a protein that, when inserted into and expressed in an abnormally proliferating host cell, e.g., a tumor cell, suppresses abnormal proliferation of that cell irrespective of the cause of the abnormal proliferation. The second in-frame AUG (ATG in DNA) codon-initiated retinoblastoma gene disclosed herein exemplifies such a broad-spectrum tumor suppressor gene and is referred to herein as the p94RB coding gene, as the RB94 gene or as a DNA molecule coding for pRB94. According to the nucleotide sequence of the retinoblastoma susceptibility gene (McGee, T.L., et al., 1989, Gene. 80:119-128), the p94RB coding gene comprises the nucleotide sequence from exon 3, nucleotide 355 to exon 27, nucleotide 264. Thus, the p94RB encoding gene by definition excludes that portion of the RBn0 gene upstream from the second in-frame AUG start codon. Figures 1A-1F show the DNA sequence of the RB94 gene wherein the ATG codon begins at nucleotide 19 of that figure (SEQ ID NO:l; SEQ ID NO:2) .
A broad-spectrum tumor suppressor protein (including phosphoproteins, lipoproteins, glycoproteins and other protein-based derivatives) is a substance that when injected into, absorbed by or caused to be expressed in any abnormally proliferating cell, reduces or completely suppresses abnormal cellular proliferation. The protein expressed by the second in-frame AUG codon-initiated retinoblastoma gene disclosed herein exemplifies such a broad- spectrum tumor suppressor protein. It is a phosphoprotein of about 94 kD relative molecular mass, and is also referred to herein as p94RB (SEQ ID NO:3). One of ordinary skill in the art will be able to determine if any other fragment of a tumor suppressor protein, e.g., the third or fourth AUG codon-initiated retinoblastoma protein of about 90 kD and 83 kD, respectively, also has the property of suppressing abnormal cellular proliferation.
3.2 Brief Description of the Figures Figures 1A-1F: Nucleotide sequence of the cDNA fragment encoding the 94 kD therapeutic RB protein (plus strand is SEQ ID NO:l, minus strand is SEQ ID NO:2) .
Figures 2A-2F: Amino acid sequence of the 94 kDa therapeutic RB protein (SEQ ID N0:3). Figure 3: Construction of baculovirus expression vector for the 94 kDa therapeutic RB protein synthesis; *R.S. is recombination sequence.
Figures 4A and 4B:Intracellular localization of recombinant baculovirus-produced pllO*3 and p94RB in insect cells: Figure 4A shows mock-infected Sf9 cells; Figure 4B shows cells producing pllO^; and Figure 4C shows cells producing p94RB; note that protein is localized to the nucleus in Figures 4B and 4C. Protein localization is by anti-RB immunochemical staining.
Figure 5: A diagram of complex formation of baculovirus-expressed and subsequently purified pllO*8 and p94RB proteins with SV40 T antigen. The immunoaffinity chromatography purified proteins were mixed with an equal amount of T antigen, and aliquots of the mixture were immunoprecipitated with PAB419 anti-T antibody, followed by Western blotting. The blot was sequentially incubated with MAb-1 anti-RB antibody and PAB419 antibody. Lane 1, lysate of T antigen immortalized W138 VA13 fibrobrasts was used as a control; lane 2, purified pllO*8; lane 3, co- precipitation of T-Ag with pllO*8; lane 4, purified p94RB; lane 5, co-precipitation of T-Ag with p94ω.
Figures 6A and 6B: Construction of recombinant plasmids for high-level expression of pllO*8 (pCMV-f- RB35) and p94RB (pCMV-s-RB42) proteins in human cells using cytolomegalovirus promoter/enhancer: Figure 6A is an explanatory drawing of the pllO*3 coding cDNA; Figure 6B provides maps of the pllO™ and p94RB expression plasmids where pCMV-f-RB35 codes for pllO*3 and pCMV-s-RB42 codes for p94RB. Note that pCMV-s-RB42 has most of pllO83 coding region deleted upstream of the second ATG.
Figures 7A and 7B: Construction of recombinant plasmids for expression of pllO0 (pBA-f-RB33) and p94RB (pβA-s-RB34) proteins in human cells using β-actin promoter: Figure A is a map of the pllO*8 coding plasmid, pBA-f-RB33; Figure B is a map of the p94RB coding plasmid, pBA-s-RB34. Note that pBA-s-RB34 has most of the pllO*8 coding region deleted upstream of the second ATG.
Figures 8A, 8B and 8C: Morphological effects of pllO*8 and p94RB expression on RB-defective bladder carcinoma cell line 5637 (ATCC HTB9) : Figure 8A is mock-transfected HTB9 cells; Figure 8B is pllO88 expressing HTB9 transfectants; Figure 8C is p94RB- expressing HTB9 transfectants. Arrows indicate examples for RB-positive immunostained cells. Note that the pllO*8 expressing cells of Figure 8B appear normal, but that the p94RB expressing cells of Figure 8C are senescent.
Figure 9. Half-life analysis of pllO™ and p94RB proteins in RB-reconstituted bladder carcinoma cell line, 5637. The bladder tumor cells were transfected in multiple dishes with either pllO** (pβA-f-RB33) or p94ω (pBA-s-RB34) expression plasmids. Twenty-four hours after transfection the cells were labeled with [35S]-methionine and chased with excess unlabeled methionine for 0, 6, 12 and 24 hours, respectively. The pllO*8 and p94RB proteins were determined by immunoprecipitation: the left side of the figure (0-12 hours) shows the half-life of pllO*8 is less than 6 hours; the right side of the figure (0-24 hours) shows the half-life of p94RB is about 12 hours.
Figure 10. Western blot analysis of exogenous pllO^and p94RB proteins in transiently transfected 5637 cells showing the distinct underphosphorylation state of the p94ω protein: lane 1 shows normal human fibroblast cell line, WI-38; lane 2 shows parental RB- minus bladder carcinoma cell line, 5637; lane 3 shows 5637 cells transfected with pllO^-expressing plasmid; lane 4 shows 5637 cells transfected with p94RB- expressing plasmid.
Figures 11A-11C. Expression of the human full- length RB protein, pllO*8 (Figure 11B) and the broad- spectrum tumor suppressor protein, p94RB (Figure 11C) in normal (non-tumorigenic) mouse fibroblast cells via retrovirus plasmid vectors. Both the pllO*8- and p94RB- expressing cells (arrows) have normal viable morphology similar to the parental cells Figure 11A) . Figures 12A-12D. Expression of the human pllO88 (Figure 12C) and p94RB (Figure 12D) proteins in mouse urinary bladder mucous membranes in vivo . Liposomes were mixed with the pllO**- and p94RB- expressing plasmids, respectively and infused directly into the mouse bladder via a catheter. Notably, transitional epithelia expressing the pllO™ (Figure 12C, arrows) or p94RB (Figure 12D, arrows) both retained normal, viable morphology similar to their counterparts in the untreated mouse bladder (Figure 12A) or the mouse bladder treated with liposomes only (Figure 12B) .
3.3 The Invention
The present invention is based upon the unexpected discovery that p94RB expressed by an expression vector in any abnormally proliferating target cell, e.g., a cancer or tumor cell, causes the suppression of the abnormal proliferation. Surprisingly, the treatment has been effective with all tested tumor cell lines and is not limited to treatment of RB-minus tumor cells.
Without wishing to be bound by a particular hypothesis or proposed mechanism of action, it is believed that the p94RB protein remains in the active, underphosphorylated form, and has a half-life in the target cell which is two to three times longer than that of pllO**. Thus, it is possible that a synergistic combination of accumulation of p94RB together with its tendency to remain in an underphosphorylated, active form serves to terminate the cell replication cycle in target tumor cells. However, whatever the mechanism of action, the property of suppressing cell growth and inducing senescence or killing any abnormally proliferating cell, irrespective of its genetic defect, is nevertheless completely unanticipated and unexpected. In order to obtain the broad-spectrum tumor suppressor protein, a gene coding for the second in- frame AUG codon-initiated RB protein, i.e., p94RB, was expressed by a baculovirus vector in insect host cells as a stable nuclear phosphoprotein. The resulting unphosphorylated forms of p94RB were able to form a specific complex with SV40 T antigen, providing an important verification that the p94RB protein shares many functional properties of the naturally occurred piio** protein, i.e., phosphorylation, viral oncoprotein association and nuclear tethering (Templeton et al., 1991, Proc. Natl. Acad. Sci.. USA. 88:3033-3037) .
The effects of transfection by either first or second in-frame AUG codon-initiated RB protein expression plasmid were compared on a number of well known human tumor cell lines. The tested cell lines included: an RB-defective human bladder carcinoma cell line, 5637 (ATCC HTB9) ; RB-defective human breast carcinoma cell line, MDA-MB-468 (ATCC HTB132) ; RB- defective human non-small cell lung carcinoma cell line, H2009 (Kratzke, R.A. , et al., 1992, The Journal of Biological Chemistry. 267:25998-26003); RB- defective human prostate carcinoma cell line, DU145 (ATCC HTB81) ; RB-defective human osteosarcoma cell line, Saos-2 (ATCC HTB85) ; RB-defective human fibrosarcoma metastatic to lung cell line, Hs913T (ATCC HTB152) ; human cervix adenocarcinoma cell line, HeLa (ATCC CCL2) and human fibrosarcoma cell line, HT1080 (ATCC CCL121) . Both the HeLa and HT1080 cell lines have normal pll0RB expression. Each of these cell lines were separately transfected with the pllO*8 coding and the p94RB coding expression plasmids. The results demonstrated that the second in-frame AUG codon-initiated RB protein, p94RB, was a more effective cell growth inhibitor, causing those dividing tumor cells to senesce and die. On the other hand, most normal human cells in vivo are either non-dividing or have the potential to progress into the cell cycle after a long latency period. Therefore, p94RB, as an active cell cycle regulatory factor and a therapeutic reagent is expected to show little or no toxicity when transiently expressed in normal cells in vivo.
The study also demonstrated that the RB-minus tumor cells expressing the second in-frame AUG codon- initiated RB protein, p94RB, did not progress through the cell cycle, as evidenced by their failure to incorporate [3H]-thymidine into DNA. However, the percentage of cells undergoing DNA replication was only slightly lower in cells producing the intact RB protein (pllO1^) than in cells that were RB-negative. Of particular interest was the fact that the RB- defective bladder carcinoma cell line, 5637, failed to phosphorylate the second in-frame AUG codon-initiated RB protein as shown by Western blot analysis. In contrast, the intact RB protein (pllO*3) expressed in transfected 5637 cells were fully phosphorylated. Moreover, the half-life of the second in-frame AUG codon-initiated RB protein, p94RB, was shown to be two- to three-fold greater than the intact RB protein (pllO*8) . Therefore, the accumulation of only unphosphorylated (active) p94RB proteins may account for the failure of transiently transfected 5637 tumor cells to enter S phase, and this in turn may cause these tumor cells to senesce and die. In addition, it has also been found that p94RB protein has its preferentially associated cellular proteins as compared to lOO*8. This difference in associated proteins may also contribute to the unique broad-spectrum tumor cell growth suppressing functions of p94RB protein. Both the fibrosarcoma cell line, HT1080 and cervix carcinoma cell line, HeLa, which have normal RB gene expression, were also successfully treated with the second in-frame AUG codon-initiated RB protein (p94RB) expression plasmid, demonstrating that expression of the p94RB protein in RB+ cancer or tumor cells significantly suppressed the tumor cell growth. Therefore, an advantage of the present invention is that the methods and products herein disclosed can be used for therapeutic treating tumors having no specific tumor suppressor gene defects, which provides a significant advantage over previous techniques for human tumor suppressor gene therapy.
Table 1, on the following page, provides a summary of the identification of the tested tumor cell lines, their tumor origin and genetic defects.
TABLE 1: The Status of Antioncogenes (Tumor Suppressor Genes) and Oncogenes in p94RB-Treated Human Tumor Cells
ANTIONCOGENES
RECIPIENT CELLS TUMOR ORIGIN RB pS3 ONCOGENES
5637 Bladder carcinoma, primary tumor Negative Mutation
DU145 Prostate carcinoma, metastasis to Point Mutation brain mutation
MDA-MB-468 Breast Carcinoma Large Mutation deletion
H2009 Lung carcinoma Mutation Mutation
Hs913T Fibrosarcoma, metastasis to lung Large Negative deletion
Saos2 Osteosarcoma, primary tumor Large Negative deletion
HeLa Cervix carcinoma, primary tumor Normal Negative c-myc activation1
HT1080 Fibrosarcoma, primary tumor Normal Normal N-ras and c-yes-1 activation2,3
Durst, M. , et al. Papillomavirus sequences integrate near cellular oncogenes in some cervical carcinomas. Proc. Natl. Acad. Sci.. USA. 84(4) :1070-1074, 1987.
Brown, R. , et al. A mechanism of activation of an N-ras gene in the human fibrosarcoma cell line HT1080. EMBO J. , 3:1321-1326, 1984.
Sugawara, K. , et al. Distribution of c-yes-1 gene product in various cells and tissues. Br. J. Cancer, 63(4) :508-513, 1991.
3.3.1. Preparation of RB94 Vectors 3.3.1.1. Therapeutic Vectors Any of the methods known to the art for the insertion of DNA fragments into a vector, as described, for example, in Maniatis, T. , Fritsch, E.F., and Sambrook, J. (1989): Molecular Cloning (A Laboratory Manual. , Cold Spring Harbor Laboratory, Cold Spring Harbor, New York; and Ausubel, F.M., Brent, R. , Kingston, R.E., Moore, D.D., Seidman, J.G., Smith, J.A., and Struhl, K. (1992): Current Protocols in Molecular Biology. John Wiley & Sons, New York, may be used to construct pθ *3 encoding gene expression vectors consisting of appropriate transcriptional/translational control signals and the desired RB cDNA sequence downstream from the first in- frame AUG codon, that is unable to code for pllO*8. These methods may include in vitro DNA recombinant and synthetic techniques and in vivo genetic recombination. Expression of a nucleic acid sequence encoding a p94RB may be regulated by a second nucleic acid sequence so that the p94RB is expressed in a host infected or transfected with the recombinant DNA molecule. For example, expression of p94RB may be controlled by any promoter/enhancer element known in the art. The promoter activation may be tissue specific or inducible by a metabolic product or administered substance.
Promoters/enhancers which may be used to control p94RB gene expression include, but are not limited to, the native RB promoter, the cytomegalovirus (CMV) promoter/enhancer (Karasuyama, H. , et al., 1989, J. Exp. Med.. 169:13), the human β-actin promoter (Gunning, P., et al., 1987, Proc. Natl. Acad. Sci. USA. 84:4831-4835), the glucocorticoid-inducible promoter present in the mouse mammary tumor virus long terminal repeat (MMTV LTR) (Klessig, D.F., et al. , 1984, Mol. Cell Biol.. 4:1354-1362), the long terminal repeat sequences of Moloney murine leukemia virus (MuLV LTR) (Weiss, R. , et al., 1985, RNA Tumor Viruses. Cold Spring Harbor Laboratory, Cold Spring Harbor, New York) , the SV40 early region promoter
(Bernoist and Chambon, 1981, Nature 290:304-310), the promoter contained in the 3' long terminal repeat of Rous sarcoma virus (RSV) (Yamamoto et al., 1980, Cell 22:787-797), the herpes simplex virus (HSV) thymidine kinase promoter/enhancer (Wagner et al., 1981, Proc. Natl. Acad. Sci. U.S.A. 78:1441-1445), the regulatory sequences of the metallothionein gene (Brinster et al., 1982, Nature 296:39-42), the adenovirus promoter (Yamada et al., 1985, Proc. Natl. Acad. Sci. U.S.A. 82(11) :3567-71) , and the herpes simplex virus LAT promoter (Wolfe, J.H., et al., 1992, Nature Genetics. 1:379-384) .
Expression vectors compatible with mammalian host cells for use in genetic therapy of tumor or cancer cells, include, but are not limited to: plasmids, retroviral vectors, adenovirus vectors, herpes viral vectors, and non-replicative avipox viruses, as disclosed, for example, by U.S. Patent No. 5,174,993. In a specific embodiment, a plasmid vector derived from pHBAPr-1-neo, was constructed for expression of p94RB in mammalian cells by placing the coding sequence for p94ω under control of the human β- actin gene promoter (Gunning, P. et al., Proc. Natl. Acad. Sci.. USA, 1987, 84:4831-4835). In another specific embodiment, a plasmid vector derived from pCMV-Neo-Ba (Baker, S.J., et al., Science. 1990, 249:912-915), was constructed for expression of p94RB in mammalian cells by placing the coding sequence for p94RB under control of the cytomegalovirus (CMV) promoter/enhancer sequences. In another specific embodiment, a retroviral vector, pLLRNL (Miller, A.D., et al., 1985, Proc. Natl. Acad. Sci.. USA. 5:431) is used to construct a vector able to transduce mammalian cells and express p94RB protein under the control of the MuLV LTR promoter, the CMV promoter, the β-actin promoter or any other effective promoter.
In yet another specific embodiment, an adenovirus type 5 (Ad5) deletion mutant, Ad-dl324, and a plasmid, pTG5955 (Rosenfeld, M.A. , et al.. Cell. 1992, 68:143- 155) are used to construct an adenovirus vector able to infect mammalian cells and express p94RB protein under the control of the adenovirus type 2 (Ad2) major late promoter, the CMV promoter, the β-actin promoter or any other effective promoter.
3.3.1.2. Vectors for Production and
Purification of p94RB Protein
Alternatively, expression vectors compatible with host cells suitable for production of p94RB may be constructed to express p94RB protein in those compatible host cells. These include but are not limited to mammalian cells infected with a virus (e.g., adenovirus, retrovirus, herpes simplex virus, avipox virus) ; insect cells infected with a virus (e.g. , baculovirus) ; microorganisms such as yeasts containing yeast vectors, or bacteria transformed with bacteriophage DNA, plasmid DNA, or cosmid DNA. The expression controlling elements of vectors vary in their strengths and specifications. Depending on the host-vector system utilized, any one of a number of suitable transcription and translation elements may be used. The produced p94RB may be purified from host cells by affinity chromatography, electrophoresis, high-performance liquid chromatography (HPLC) or any other methods known to the art.
In a specific embodiment an engineered derivative of Autographa California Multiple Nuclear Polyhedrosis Virus ("AcMNPV") was used to produce p94RB protein in cultured Fall Army worm Spondoptera frugiperda cells (Sf9 cells) with a strong temporally regulated promoter of the polyhedron gene whose product represents 50% or more of total cellular proteins during a lytic infection. The baculovirus-expressed p94ω protein was subsequently purified by im unoaffinity chromatography.
3.3.1.3. Detection of p94RB Coding
Expression Vectors
Expression vectors containing p94RB coding inserts can be identified by three general approaches: (a) nucleic acid hybridization, (b) presence or absence of "marker" gene functions, and (c) expression of inserted sequences. In the first approach, the presence of a p94RB coding gene inserted in an expression vector can be detected by nucleic acid hybridization using probes comprising sequences that are homologous/complementary to the inserted p94RB coding gene. Such hybridization can be carried out under stringent or nonstringent conditions, depending upon the size and sequence of the probe selected. In the second approach, the expression vector/host system can be identified and selected based upon the presence or absence of certain "marker" gene functions (e.g., thymidine kinase activity, resistance to antibiotics, viral occlusion formation in a baculovirus vector infected insect cell, etc.) caused by introduction of the expression vector into the host cell. For example, if the p94RB coding gene is inserted within a vector having a dominant selectable marker gene, such as a neomycin phosphotransferase gene under separate control of an appropriate promoter, such as an SV40 early promoter, the expression vector containing the p94RB coding gene can be identified by the presence of the marker gene function (geneticin resistance) . In the third approach, expression vectors containing a p94RB coding gene can be identified by assaying the p94RB coding gene products expressed by the vectors. Such assays can be based, for example, on the physical or functional properties of the p94RB gene products in in vitro or in vivo assay systems including metabolic radiolabelling by [35S] methionine, SDS-polyacrylamide gel electrophoresis, binding with a specific antibody, and phosphorylation by a protein kinase.
3.3.2. Expression of 094*^ An appropriate p94RB coding expression vector, i.e., a vector which contains the necessary elements for the transcription and translation of the p94RB protein-coding sequence may be introduced into a host cell. A host cell may be any cell type compatible with the vector for expressing and producing p94RB. In a preferred embodiment, the host cell is a mammalian tumor cell to be treated. In a more preferred embodiment, the host cell is a human tumor cell to be treated. Expression of the p94RB in a host cell may be transient, permanent, or inducible.
The necessary transcriptional and translational signals, including promoter/enhancer sequences can also be supplied by the native RB gene and/or its flanking regions. A variety of vector/host systems may be utilized to express the p94RB protein-coding sequence in a tumor cell to be treated. These include but are not limited to mammalian cell systems transfected, infected or transduced with a plasmid, or a virus (e.g., adenovirus, retrovirus, herpes simplex virus, avipox virus) . The expression elements of vectors vary in their strengths and specificities. Depending on the host cell to be treated, any one or more of a number of suitable transcription and translation elements may be used. 3.3.3. Methods of Treatment The p94RB encoding gene construct of the present invention may be placed by methods well known to the art into an expression vector such as a plasmid or viral expression vector. A plasmid expression vector may be introduced into a tumor cell by calcium phosphate transfection, liposome (for example, LIPOFECTIN)-mediated transfection, DEAE Dextran- mediated transfection, polybrene-mediated transfection, electroporation and any other method of introducing DNA into a cell.
A viral expression vector may be introduced into a target cell in an expressible form by infection or transduction. Such a viral vector includes, but is not limited to: a retrovirus, an adenovirus, a herpes virus and an avipox virus. When p94RB is expressed in any abnormally proliferating cell, the cell replication cycle is arrested, thereby resulting in senescence and cell death and ultimately, reduction in the mass of the abnormal tissue, i.e., the tumor or cancer. A vector able to introduce the gene construct into a target cell and able to express p94RB therein in cell proliferation-suppressing amounts can be administered by any effective method. For example, a physiologically appropriate solution containing an effective concentration of active vectors can be administered topically, intraocularly, parenterally, orally, intranasally, intravenously, intramuscularly, subcutaneously or by any other effective means. In particular, the vector may be directly injected into a target cancer or tumor tissue by a needle in amounts effective to treat the tumor cells of the target tissue.
Alternatively, a cancer or tumor present in a body cavity such as in the eye, gastrointestinal tract, genitourinary tract (e.g., the urinary bladder) , pulmonary and bronchial system and the like can receive a physiologically appropriate composition (e.g., a solution such as a saline or phosphate buffer, a suspension, or an emulsion, which is sterile except for the vector) containing an effective concentration of active vectors via direct injection with a needle or via a catheter or other delivery tube placed into the cancer or tumor afflicted hollow organ. Any effective imaging device such as X-ray, sonogra , or fiberoptic visualization system may be used to locate the target tissue and guide the needle or catheter tube.
In another alternative, a physiologically appropriate solution containing an effective concentration of active vectors can be administered systemically into the blood circulation to treat a cancer or tumor which cannot be directly reached or anatomically isolated.
In yet another alternative, target tumor or cancer cells can be treated by introducing p94RB protein into the cells by any known method. For example, liposomes are artificial membrane vesicles that are available to deliver drugs, proteins and plasmid vectors both jln vitro or jLn vivo (Mannino, R.J. et al. , 1988, Biotechniques. 6:682-690) into target cells (Newton, A.C. and Huestis, W.H.,
Biochemistry. 1988, 27:4655-4659; Tanswell, A.K. et al., 1990, Bioch ica et Biophysica Acta. 1044:269-274; and Ceccoll, J. et al. Journal of Investigative Dermatology. 1989, 93:190-194). Thus, p94RB protein can be encapsulated at high efficiency with liposome vesicles and delivered into mammalian cells in vitro or in vivo.
Liposome-encapsulated p94RB protein may be administered topically, intraocularly, parenterally, intranasally, intratracheally, intrabronchially, intramuscularly, subcutaneously or by any other effective means at a dose efficacious to treat the abnormally proliferating cells of the target tissue. The liposomes may be administered in any physiologically appropriate composition containing an effective concentration of encapsulated p94RB protein.
3.3.4. Tumors Susceptible To Treatment The gene construct and vectors of the present invention are effective in inhibiting the growth or mitosis or both of any type of tumor cell. The gene construct of the invention has demonstrated effectiveness in treating tumor cells of carcinomas and sarcomas. In particular, the gene construct of the invention has demonstrated effectiveness in suppressing replication and inducing cell senescence followed by cell death in the following tumor cell types: bladder carcinoma, lung carcinoma, breast carcinoma, prostate carcinoma, fibrosarcoma, osteosarcoma and cervix carcinoma.
Further, the gene construct of the invention has demonstrated effectiveness in suppressing replication and inducing cell senescence followed by cell death in the tumor cells having the following identified genetic defects: tumor suppressor gene RB and p53 mutation, oncogene myc activation, and oncogene N-ras and c-yes-1 activation.
Furthermore, the gene construct of the invention has demonstrated effectiveness in suppressing replication and inducing cell senescence followed by cell death in the tumor cells having normal endogenous tumor suppressor RB110 and/or p53 gene expression.
In addition, the gene construct of the invention is able to suppress replication in lymphomas, leukemia and in tumor cells having tumor suppressor gene DCC and NFl genetic defects, as well as in other tumor cell types in which the genetic defects are unknown or have yet to be identified. 3.3.5. Ex Vivo Treatment of Tumor or Cancer Tissues
In a preferred embodiment a tumor cell is transduced with a retrovirus vector, an adenovirus vector, a plasmid vector or any other appropriate vector capable of expressing the p94RB protein in that tumor cell. The cancer cell may be present in a blood or bone marrow sample collected from a leukemia patient. A dose of p94RB protein expressing retrovirus vector or adenovirus vector or plasmid vector or any other appropriate vector is administered to the sample of blood or bone marrow at a dose sufficient to transduce enough cells in the sample to produce a reduction in tumor cell numbers. The cell proliferation of the treated cancer cells will be slowed or terminated followed by a process similar to normal cellular differentiation or cell senescence. Analo-gously, blood or bone marrow or other tissue is treated ex vivo using an effective dose of a lipsome- encapsulated p94RB protein. Thereafter the sample may be returned to the donor or infused into another recipient.
3.3.6. In Vivo Treatment of Tumor or Cancer Tissues
Methods of administering viral vectors are well known. In general, the skilled artisan will appreciate that a retroviral vector, an adenovirus vector, a plasmid vector, or any other appropriate vector capable of expressing the p94RBprotein can be administered in vivo to a cancer by a wide variety of manipulations. All such manipulations have in common the goal of placing the vector in sufficient contact with the target tumor to permit the vector to transduce or transfect the tumor cells. In a preferred embodiment, cancers present in the epithelial linings of hollow organs may be treated by infusing the vector suspension into a hollow fluid filled organ, or by spraying or misting into a hollow air filled organ. Thus, the tumor cell may be present in or among the epithelial tissue in the lining of pulmonary bronchial tree, the lining of the gastrointestinal tract, the lining of the female reproductive tract, genito-urinary tract, bladder, the gall bladder and any other organ tissue accessible to contact with the vector. In another preferred embodiment, the cancer may be located in or on the lining of the central nervous system, such as, for example, the spinal cord, spinal roots or brain, so that vectors infused in the cerebrospinal fluid will contact and transduce the cells of the tumor in that space.
In another preferred embodiment, the cancer is a solid tumor. The skilled artisan will appreciate that the vector can be administered to the tumor by direct injection of the vector suspension into the tumor so that vectors will contact and transduce or transfect the tumor cells inside the tumor.
In yet another preferred embodiment, the cancer may be a cancer of the blood, blood forming organs or any organ directly perfused by the blood, so that vectors injected into the blood stream will contact and treat the cells of the cancer. Thus, the cancer may be a leukemia, a lymphoma or other tumor type and the tumor cell may be present in the blood, the bone marrow, the spleen, the thymus, the liver and any other blood perfused organ.
The skilled artisan will understand that the vector is administered in a composition comprising the vector together with a carrier or vehicle suitable for maintaining the transduction or transfection efficiency of the chosen vector and promoting a safe infusion. Such a carrier may be a pH balanced physiological buffer, such as a phosphate, citrate or bicarbonate buffer, a saline solution, a slow release composition and any other substance useful for safely and effectively placing the vector in contact with abnormally proliferating cells to be treated. The invention is further described in the following examples which are in no way intended to limit the scope of the invention.
4. Examples 4.1 Preparation of Vectors for Expression of the Second In-Frame AUG Codon- Initiated RB Protein in Insect Cells
The engineered derivatives of Autographa California Multiple Nuclear Polyhedrosis Virus ("AcMNPV") have been widely employed to produce high levels of accurately processed and biologically active proteins. This baculovirus propagates in cultured Fall Army worm Spondoptera frugiperda cells (Sf9 cells) and has a strong temporarily regulated promoter of the polyhedron gene whose product represents 50% or more of total cellular proteins during a lytic infection.
By in vivo recombination, the coding sequence of a foreign gene can easily be placed under the transcriptional control of the polyhedron promoter, resulting in a high level of expression. In addition, such proteins may be correctly folded and contain appropriate post-translational modifications like those proteins in the native higher eukaryotes.
By site-specific mutagenesis, two BamHl sites were introduced into the RB cDNA at nucleotides +7 and +3230 (the A of the second in-frame AUG codon is designated +19) . The resulted DNA molecule has the nucleotide sequence of Figure 1 (SEQ ID NO:l; SEQ ID NO:2), which is also referred to herein as the second in-frame AUG codon-initiated RB protein gene, or the p94RB encoding gene. The coded-for protein has the sequence of Figure 2 (SEQ ID NO:3) and is referred to herein as the second in-frame AUG codon-initiated RB protein, or the p94RB protein.
In an attempt to achieve maximal production of the second in-frame AUG codon-initiated RB protein in the baculovirus expression system, the recombinant transfer vector was constructed with insertion of the p94RB gene into the pVL1393 plasmid so that the p94RB gene was placed under the control of the polyhedron gene promoter.
As shown in Figure 3, the resulting pVL-s-RB plasmid contains no additional AUG start codon upstream from the p94M translation initiation site at nucleotide +19, and thus encodes a nonfusion p94RB protein. In a parallel study, the same strategy was employed to construct a pllO*3 expression vector which was designated pVL/lst AUG-RB.
Transfer of RB cDNAs from the recombinant vectors to the viral genome was accomplished by co-transfecting wild-type AcMNPV virus DNA with pVL-s- RB plasmid DNA or pVL/lst AUG-RB plasmid DNA. The recombinant viruses were subjected to three rounds of plaque purification to obtain a pure stock of RB- containing baculovirus, designated AcMNPV-RB94 and AcMNPV-RBllO, respectively.
4.2 Purification of pllO1"1 and p94RB Proteins The pllO*3 and p94RB proteins were purified from baculovirus-infected insect cells by immunoaffinity chromatography. Briefly, insect cells were harvested 24 hours after the virus infection and lysed at 4°C with EBC buffer (50 mM Tris-HCl, pHδ.O, 120 mM NaCI, 0.5% NP-40, 50/ig/ml aprotinin) . The lysate was clarified by centrifugation and the pllO8- or p94RB- containing supernatant was incubated with biotinylated WL-1 polyclonal anti-RB antibodies (Xu, H-J., et al., 1989, Oncogene, 4:807-812) at 4°C overnight. The procedures for biotinylation of rabbit IgGs using succinimide ester followed the methods described by Bayer and Wilchek (Baylor, E.A. and Wilchek, M. , 1980, Methods Biochem. Anal.. 26:1-45). The RB protein-IgG- biotin complex was collected on a streptavidin agarose gel column. Purified pllO*3 or p94RB were eluted from separate columns using 100 mM glycine (pH 2.2) and neutralized with 1M of phosphate (pH 8.0).
4.2.1. p94RB Shares Major Biochemical and
Biological Properties With pllO88
Since non-functional mutations of the retinoblastoma protein are characterized by defects in phosphorylation, viral oncoprotein association and nuclear localization (Templeton et al., 1991, Proc. Natl. Acad. Sci.. USA. 88:3033-3037), the functional aspects of the artificial p94RB protein were studied for these characteristics.
First, to determine whether the RB proteins produced in the insect cells with the baculoviruses were associated with the nucleus, the AcMNPV-RBllO and AcMNPV-RB94 infected Sf9 cells were immunostained with MAb-1 anti-RB monoclonal antibody 24h after infection. As shown in Figure 4, intense staining was found exclusively in the nuclei of cells infected with either AcMNPV-RBllO (panel B) or AcMNPV-RB94 (panel C).
The pllO1^ and p94RB proteins purified from baculovirus-infected insect cells by immunoaffinity chromatography were tested for their ability to form a specific complex with SV40 T antigen. Briefly, equal amounts of p94RB or pllO*3 and T antigen were mixed and aliquots of the mixture were immunoprecipitated with PAB419 anti-T antibody. As shown in Figure 5, mixing of p94RB (or pllO™) with T antigen in vitro resulted in the co-immunoprecipitation of both under- and hypo- phosphorylated p94RB (lane 5), or pllO 8 (lane 3) with PAB419. The data demonstrated that either pllO^or p94RB protein can form a specific complex with SV40 T antigen. The AcMNPV-RB94 virus-infected insect cells appear to make hyperphosphorylated p94RB (lane 4) , which was unable to form complexes with SV40 T antigen (compare lane 4 with lane 5) .
The Western blot shown in Figure 5 revealed an apparent relative molecular mass (Mr) of 94 kD for the second in-frame AUG codon-initiated RB protein. On SDS-PAGE, the p94RB protein (Figure 5, lanes 4 and 5) was smaller than the naturally occurring 98 kDa proteins of unknown origin (Xu et al., 1989, Oncogene, 4:807-812) (Figure 5, lane 1). Therefore, the second in-frame AUG codon-initiated RB protein of this invention (p94RB) has not been found to occur naturally in human cells.
It is concluded that the second in-frame AUG codon-initiated p94RB protein produced in recombinant virus-infected insect cells is a artificial but stable nuclear phosphoprotein with its under- and hypo-phosphorylated forms being able to assemble specific complex with SV40 T antigen, as does the naturally occurring RB protein species, pllO1^.
4.3 Construction of Expression Vectors for Mammalian Cells
4.3.1. Subcloning of RB cDNA Fragments
Encoding for the First and Second In-Frame AUG Codon-initiated RB Proteins Subcloning of RB cDNA fragments encoding for the first and second in-frame AUG codon-initiated RB proteins was accomplished by standard methods in the art. The methods for DNA manipulation were modified from Maniatis, T., Fritsch, E.F., and Sambrook, J. (1989) : Molecular Cloning (A Laboratory Manual) .
Cold Spring Harbor Laboratory, Cold Spring Harbor, New York; and Ausubel, F.M. , Brent, R. , Kingston, R.E., Moore, D.D., Seidman, J.G., Smith, J.A. , and Struhl, K. (1992) : Current Protocols in Molecular Biology. John Wiley & Sons, New York.
4.3.2. Preparation of a DNA Molecule
Encoding the Second In-Frame AUG Codon-initiated RB Protein.
A plasmid, p4.95BT (Friend et al., 1987, Proc. Natl. Acad. Sci. USA. 84:9059-9063) or F7 (Takahashi, R. , Hashimoto, T. , Xu, H-J., et al., 1991, Proc. Natl. Acad. Sci. USA. 88:5257-5261) containing the full length retinoblastoma (RB) gene cDNA was digested with the restriction enzyme. Hind II, at nucleotide +7 and the restriction enzyme, Seal, at nucleotide 3,230 (The A of the second in-frame AUG codon of the full length RB cDNA open reading frame was designated nucleotide +19). The resulted 3,230 bp RB cDNA fragment had two blunt ends. Conversion of the blunt ends to restriction enzyme BamHI sites was done by ligation of a synthetic BamHI oligonucleotide linker (GGGATCCC) to each blunt end of the fragment followed by digestion with the BamHI enzyme.
The desired RB cDNA fragment was inserted into the BamHI cloning site of a plasmid vector, pUC19, and propagated in the Escherichia coli strain, DH5 alpha bacterial cells. The recombinant plasmid was purified from a single DH5 alpha transformant and designated plasmid pUC-s-RB. This plasmid contains the desired RB cDNA fragment of 3,230 bp coding for the second in-frame AUG codon-initiated RB protein of 816 amino acids.
4.3.3. Preparation of A DNA Molecule
Encoding the First In-Frame AUG Codon-initiated RB Protein. The full length RB cDNA plasmid was digested with the restriction enzyme, Acyl at nucleotide -322 and Seal at nucleotide 3,230. The Acyl ends (overhang 5'-CG) were repaired by "filling in" the ends with the Klenow fragment of E. coli DNA polymerase I in the presence of all 4 dNTPs to generate blunt ends. Conversion of the blunt ends to restriction enzyme BamHI sites was done as described above. The resulted RB cDNA fragment of 3,552 bp was inserted into the plasmid pUC19 and propagated in the Escherichia coli strain DH5 alpha, which was subsequently purified from a single DH5 alpha transformant and designated plasmid pUC-f-RB. This plasmid contains the RB cDNA fragment of 3,552 bp coding for the first in-frame AUG codon- initiated RB protein of 928 amino acids.
4.3.4. Construction of p94RBExpression
Plasmid Using A Human β-Actin Gene Promoter
The RB cDNA fragment of 3,230 bp coding for the second in-frame AUG codon-initiated RB protein of 816 amino acids (p94RB) was recovered from plasmid pUC-s-RB following the restriction enzyme, BamHI digestion, and re-inserted into the unique BamHI site of an expression plasmid, pHBAPr-1-neo (Gunning, P., et al., Proc. Natl. Acad. Sci.. USA. 1987, 84:4831-4835) in a orientation that the p94RB coding sequence was under the direct control of the β-actin gene promoter. A plasmid vector with the correct insert orientation was selected by restriction endonuclease mapping after propagation in DH5 alpha Escherichia coli host cells, and was designated pβA-s-RB34 (Figure 7B) . The corresponding DH5 alpha strain that contains plasmids P/5A-S-RB34 was thereafter designated DHB-S-RB34 (ATCC 69241, patent depository, American Type culture Collection) .
The plasmid vector pβA-s-RB34 contains no additional AUG codon between the β-actin gene promoter and the second in-frame AUG codon of the RB coding sequence, and thus encodes a non-fusion p94RB protein. The plasmid vector pβA-s-RB34 also confers a dominant selectable marker (geneticin resistance) in eukaryotic cells through expression of the neomycin phosphotransferase (neo) under separate control of an SV40 early promoter (Figure 7, sv-neo) .
4.3.5. Construction of pllO* Expression Plasmid Using A Human β-Actin Gene Promoter
The RB cDNA fragment of 3,552 bp coding for the first in-frame AUG codon-initiated RB protein of 928 amino acids (pllORB) was recovered from plasmid pUC-f-RB and re-inserted into the expression plasmid pHBAPr-1-neo downstream from the β-actin gene promoter. The resulting plasmid vector was designated pβA-f-RB33 (Figure 7A) . The plasmid vector pβA-f-RB33 contains no additional AUG codon between the β-actin gene promoter and the first in-frame AUG codon of the RB coding sequence, and thus encodes a non-fusion pllO*8 protein.
4.3.6. Construction of p94RB and pllO*8 Expression Plasmids Using A Cytomegalovirus Promoter fCMVp.
Alternatively, an expression plasmid, pCMV-Neo- Bam (Baker, S.J., et al., Science. 1990, 249:912-915) was used in place of plasmid pHβAPr-1-neo. The vector included cytomegalovirus (CMV) promoter/enhancer sequences, which could drive expression of the insert at the BamHI site, and splicing and polyadenylation sites derived from the rabbit β-globin gene, which ensured proper processing of the transcribed insert in the cells. A pBR322 origin of replication and β- lactamase gene facilitated propagation of the plasmid in E. coli . The plasmid conferred geneticin resistance (a selectable marker in eukaryotic cells) through expression of the neomycin phosphotransferase (neo) under the control of a herpes simplex virus (HSV) thymidine kinase promoter.
The same strategies as described supra in Sections 4.3.4 and 4.3.5 were employed to transfer the RB cDNA fragments of 3,230 bp and 3,552 bp from plasmids pUC-s-RB and pUC-f-RB, respectively, to the unique BamHI site in the expression vector, pCMV-Neo- Bam. The resulting plasmid vectors were designated by the names of pCMV-s-RB42, expressing the p94RB and pCMV-f-RB35, expressing the pll0RB (Figure 6) . The corresponding Escherichia coli DH5 alpha strain which contains plasmids pCMV-s-RB42 was thereafter designated DHC-S-RB42 (ATCC 69240, patent depository, American Type Culture Collection) .
4.3.7. Construction of p94RB Protein Expression Retrovirus Vectors
For this protocol, retroviral vector, pLLRNL (Miller, A.D., Law, M.-F., Verma, I.M., Molec. Cell Biol. , 1985, 5:431) and amphotropic retrovirus packaging cell line, PA317 (ATCC CRL9078) (Miller, A.D., and Buttimore, C. , Molec. Cell Biol.. 1986, 6:2895-2902) are used.
A plasmid p4.95BT or F7 containing the full-length RB gene cDNA is digested with the restriction enzyme Hind II at nucleotide +7 (the A of the second in-frame AUG codon of the full-length RB cDNA open reading frame was designated nucleotide +19) . Conversion of the Hind II site to restriction enzyme Hind III site is done by ligation of a synthetic Hind III oligonucleotide linker (CCAAGCTTGG) to the blunt ends of the linear plasmid DNA, followed by digestion with the Hind III enzyme. The linear plasmid DNA is further digested with restriction enzyme, Seal, at nucleotide 3,230. The resulted RB cDNA fragment of 3,230 bp codes for the second in-frame AUG codon-initiated RB protein of 816 amino acids (p94ω) . This fragment has a 5'-Hind III site (cohesive end) and a 3'-Seal site (blunt end) , which facilitates its insertion into the retroviral vector, pLLRNL.
The vector pLLRNL is digested with two sets of restriction enzymes: Hind III/ClaI and Smal/Clal to delete the luciferase gene. Appropriate fragments are recovered from the agarose gel following electrophoresis, and ligated with the RB cDNA fragment of 3,230 bp to form a new vector, pLRB94RNL, in which the p94RB expression is under the control of the long terminal repeat sequences of Moloney murine leukemia virus (MuLV LTRs) . The basic protocol for construction of the retroviral vector, pLRB94RNL, is modified from Huang, H.-J.S., et al., 1988, Science. 242:1563-1566.
Alternatively, the vector pLLRNL is digested with a single restriction enzyme, Hind III, to delete the luciferase gene, as well as the Rous sarcoma virus promoter (RSV) . An appropriate DNA fragment is recovered from the p94RB expression plasmid, pCMV-s-RB42 (or pβA-s-RB34) . The recovered DNA fragment, which contains the 3,230 bp RB cDNA fragment and 5'-flanking CMV promoter (or β-actin promoter), is inserted into the Clal restriction site of the retroviral vector. Conversion between the restriction enzyme sites is done by the methods as described εupra in Section 4.3.7. In the resulting p94RB expression retrovirus vector, the p94RB gene is under the control of an internal promoter (the CMV promoter or β-actin promoter) , while the Tn5 neomycin-resistance gene (Neo) is under the control of the MuLV LTRs. A safe and efficient amphotropic packaging cell line is necessary for transfer of retroviral vector genes into human cancer cells. The virus packaging methods are modified from the method of Miyanohara et al., Proc. Natl. Acad. Sci. , USA. 1988, 85:6538-6542. For this protocol, the PA317 packaging cell line is used. This packaging cell line has received prior approval for use in human gene therapy clinical trials.
The retroviral vector (pLRB94RNL) DNA is transfected into PA317 packaging cells by LIPOFECTIN reagent (GIBCO BRL Life Technologies, Inc., Gaithersburg, MD) or electroporation methods as described in Sections 4.4.1. infra . Single colonies are isolated by selection in G418-containing medium (400 μg/ml) and expanded into mass cultures. To titer the virus produced by selected PA317 clones, dilutions of cell-free culture medium from each PA317 clone are applied to 208F rat fibroblasts (indicator cells) in the presence of POLYBRENE (Sigma, 4 μg/ml) and G418 selection (400 μg/ml) is started 24 hours after infection. After two weeks, G418-resistant colonies are visualized by Giemsa staining and viral titers are determined (colony-forming units per milliliter, cfu/ml) . PA317 clones producing high virus titers are then assayed for human p94RB protein expression by Western immunoblotting as described previously
(Xu, H.-J., et al., Oncogene, 1991, 6:1139-1146). Cell-free culture media from selected PA317 clones expressing high level of human p94RB protein are then applied to human cancer cells e_x vivo or in vivo.
4.3.8. Construction of p94RB Protein Expression Adenovirus Vectors
The recombinant adenovirus Ad-RB94 is constructed from the adenovirus type 5 (Ad5) deletion mutant,
Ad-dl324, and a plasmid, pTG5955, in which the human
CFTR cDNA has been replaced by the human RB cDNA fragment of 3,230 bp coding for the p94RB protein. The plasmid pTG5955 containing the RB cDNA insert is linearized by restriction enzyme Clal cleavage and co-transfected with the large fragment of Clal-cut Ad-dl324 DNA into 293 (ATCC CRL1573) cells to allow homologous recombination to occur, followed by replication and encapsidation of recombinant adenoviral DNA into infectious virions and the formation of plaques. Individual plaques are isolated and amplified in 293 cells, viral DNA is isolated and recombinant adenovirus plaques containing the human RB cDNA (Ad-RB94) are identified by restriction cleavage and Southern analysis. Ad-RB94 viruses are propagated in 293 cells and recovered 36 hours after infection. The viral preparation is purified by CsCl density centrifugation, and stored in virus dialysis buffer (10 mM Tris-Hcl, pH7.4; 1 mM MgCl2) at 4°C for immediate use; or stored at -70°C prior to use (with the addition of 10% glycerol) . The basic protocol for construction of the recombinant adenovirus Ad-RB94 is modified from Rosenfeld, M.A., et al.. Cell. 1992, 68:143-155.
4.3.9. Physical DNA Transfer Method An alternative gene transfer method that has been approved for use in humans by the Food and Drug Administration is the transfer of plasmid DNA in liposomes directly to tumor cells .in situ (Nabel, E.G., et al., 1990, Science. 249:1285-1288). Plasmid DNA is easy to certify for use in humans because, unlike retroviral vector, it can be purified to homogeneity.
The p94RB expressing plasmid vectors pCMV-s-RB42 or pBA-s-RB34 are used to form complexes with liposomes, and directly treat tumor cells in vivo (or ex vivo) . In this procedure, as described in
Section 4.4.1 infra , stable integration of the DNA into transfected tumor cells is not required for gene therapy as transient expression may suffice.
4.4. Treatment of Human Tumor Cells In Vitro With p94 RBasπιid Vectors pβA-s-RB34 or pCMV-s- RB42.
4.4.1. Treatment of RB-Defective Human Tumor Cells In Vitro
Human tumor cells having known RB gene deficiencies were treated with the p94RB plasmid vector pβA-s-RB34 (or pCMV-s-RB42) . These include: 1) human bladder carcinoma cell line, 5637, (ATCC HTB9) ; 2) human breast carcinoma cell line, MDA-MB-468 (ATCC HTB132) ; 3) human non-small cell lung carcinoma cell line, H2009 (Kratzke, R.A. , et al., 1992, The Journal of Biological Chemistry. 267:25998-26003); 4) human prostate carcinoma cell line, DU145 (ATCC HTB81) ; 5) human osteosarcoma cell line, Saos2 (ATCC HTB85) ; and 6) human fibrosarcoma metastatic to lung cell line, HS913T (ATCC HTB152) .
For treatment, tumor cells were transiently transfected with the plasmid DNA pβA-s-RB34 (or pCMV-s-RB42) via LIPOFECTIN reagent (GIBCO BRL Life Technologies, Inc. Gaithersberg, MD) . Similar results have been obtained from transfection using calcium phosphate or electroporation methods.
The following procedures for transfection using LIPOFECTIN were modified from the manufacturer's specifications. Tumor cells were seeded in 100-mm dishes in appropriate growth medium supplemented with serum. The cells were incubated at 37°C in a 5% C02 environment until the cells were 40-60% confluent. This usually took 18-24 hours, but the time varied among cell types. The following solution was prepared in 17 x 75 mm polystyrene tubes: Solution A - for each dish of cells to be transfected, 5-10μg of plasmid DNA were diluted into a final volume of lOOμl with serum-free medium; Solution B - for each dish of cells to be transfected, 30-50μl of LIPOFECTIN reagent was diluted into a final volume of lOOμl with serum- free medium. The two solutions were combined, mixed gently, and incubated at room temperature for 10-15 min. The LIPOFECTIN reagent interacted spontaneously with plasmid DNA to form a lipid-DNA complex. While the lipid-DNA complex was forming, the cells were washed twice with 6 ml of serum-free medium. For each transfection, 6 ml of serum-free medium were added to each polystyrene tube containing the lipid-DNA complex. The solution was mixed gently, and the medium-complex was overlayed onto the cells. The dishes were then swirled gently to ensure uniform distribution. The dishes were then incubated at 37°C in a 5% C02 incubator. After 12 to 24 hours the medium-complex was replaced with appropriate growth medium containing 10% fetal calf serum.
In parallel studies, tumor cells were transfected with the plasmid DNA pβA-f-RB33 or pCMV-f-RB35 which expresses the pllO*8. The following assays were used to evaluate the growth inhibitory effects of introducing p94RB versus pllO0 expression in RB defective tumor cells:
1) DNA synthesis in tumor cells treated with plasmid vectors.
After plasmid DNA treatment the tumor cells were labeled with [3H]-thymidine for 2 hours, then transferred to polylysine-coated slides, fixed and immunocytochemically stained with a monoclonal anti-RB antibody, MAb-l (Triton Biosciences, Inc. Alameda, CA) . The RB-positive transfected cells were counted under the microscope. The slides were then coated with Kodak NTB2 autoradiographic emulsion and exposed for 7-10 days. The [3H]-thymidine labeling and RB protein immunocytochemical staining were done according to the methods previously described (Xu et al., Oncogene. 1991, 6:1139-1146). About 400 to 1600 RB-positive and 600 RB-negative tumor cells were assessed for each determination of [3H]-thymidine uptake. The study demonstrated that the RB-defective tumor cells expressing p94RB did not progress through the cell cycle, as evidenced by their failure to incorporate [3H]-thymidine into DNA (Table 2) . However, the percentage of cells undergoing DNA replication was only slightly lower in cells producing pllO*8 than in cells that were RB-negative (Table 2) .
Table 2. Immunocytochemical Staining and pH] Thymidine Incorporation of
RB-Defective Tumor Cells Following Transfection With p94 or pi 10™ Expression Plasmids
Recipient Protein Cells Incorporating [3H] Thymidine Cells Promoter Expressed RB + RB-
5637 β-actin gene pi 10™ 34% 45% promoter p94RB 2.3% 43%
Cytomegalovirus pi 10™ 21 % — promoter/enhancer p94RB 1.8%
MDA- Cytomegalovirus pi 10™ 14% 40% MB-468 promoter/enhancer p94 B 0.5% 39%
H2009 /3-actin gene pl lO™ 19% 26% promoter p94RB 0.1 % 27%
DU145 Cytomegalovirus pi 10™ 23% 33% Promoter/enhancer p94™ 1.0% 33%
Hs913T Cytomegalovirus pi 10™ 18% 34% promoter/enhancer p94™ 0.9% 36%
Saos2 Cytomegalovirus pi 10™ 19% 32% promoter/enhancer p94™ 0.9% 35%
2) Colony formation assay.
Approximately 48 hours after transfection the tumor cells were replated at a density of 105 cells per 100 mm dish with selected medium containing G418 of 400-600 μg/ml. Cells were cultured for 2 to 3 weeks and colonies of >100 cells were scored. The data are illustrated in Table 3. Cells treated with plasmid vectors expressing p94RB formed approximately four-fold fewer colonies than those transfected with pllO*8 plasmid vectors. The difference was statistically significant (p <0.05 by t-test) .
Furthermore, in those colonies that did form after p94ω plasmid DNA treatment, p94RB protein expression was no longer observed. Failure to isolate long-term cultures expressing the p94M protein in treated tumor cells shows that p94RB did suppress tumor cell growth. In contrast, 7 of 48 cell lines (approximately 15%) derived from tumor cells after transfection with the pllO*8 plasmid DNA were found to express the pllO*8 protein. This percentage was consistent with results expected in human cells transfected with a vector containing two independent transcription units and therefore introduction of pllO*8 expression does not exert growth inhibitory effects on RB-defective tumor cells.
Table 3: Growth inhibitory effects of introducing pi 10™ and p94™ expression into RB-defective bladder carcinoma cell line, 5637 (HTB9). Each number represents 6 to 11 dishes.
Vector Type Number of G418-Resistant Colonies Formed
Vector pl lO™ p94] RB
pCMV-Neo-Bam 280 24 pHβAPr-1-neo 33
3) The Effect of p94RB Expression on Cellular Morphology
The HTB9 transfectants were also immunostained with MAb-1 anti-RB monoclonal antibody about 24 hours after transfection. The staining results are illustrated in Figure 8.
As demonstrated in Figure 8, the majority of RB-positive, p94RB-expressing HTB9 cells become very large in size, with lower nucleocytoplasmic ratio, or higher incidence of being multinucleated cells (panel C) , a morphological change frequently associated with cellular senescence. However, such a morphological change has not been seen in group A, mock-transfected HTB9 cells and group B, pll0RB expressing RB-positive HTB9 cells (Figure 8, panels A and B) . 4.4.2. Treatment of Human Tumor Cells Having Normal (pllORB) RB Expression (RB+)
Two RB+ human cell lines (i.e., having no RB gene defect) , including a human fibrosarcoma cell line, HT1080 (ATCC CCL121) , and human cervix carcinoma cell line, HeLa (ATCC CCL2) were treated with the p94ω protein expression plasmid, pCMV-s-RB42, using the LIPOFECTIN reagent as described supra . In parallel studies, these cell lines were also transfected with the pllO*8 protein expression plasmid, pCMV-f-RB35. The colony formation assay as described εupra was used to evaluate the growth inhibitory effects of introducing exogenous p94RB versus pllO*8 expression in RB+ tumor cells. As shown in Table 4, expression of the p94RB protein dramatically inhibited the cell growth of HT1080 and HeLa cells. There was a two- to nine-fold reduction in the number of G418-resistant colonies formed after treated with the plasmid victor pCMV-s-RB42 expressing p94RB, while no such effect was observed by transfection with the pCMV-f-RB35 plasmid (expressing pllO*8 protein) . The difference was statistically significant (the two-tailed P values were less than 0.03 as calculated by the paired t- test) .
Table 4: Growth inhibitory effects of introducing pi 10™ and p94™ expression into RB-positive human fibrosarcoma cell line, HT1080 and the RB positive human cervix carcinoma cell line, HeLa. The RB expression was under the control of cytomegalovirus (CMV) promoter. For each experiment, three 5-cm2 dishes were transfected and the total colonies counted after ten days of selection in G418 (1 mg/ml).
No. of G418-Resistant Colonies Formed
Recipient
Cells Experiment Vector pllO™ p94w
HT1080 1 94 129 14
2 88 122 16
3 100 120 17
4 99 110 15
HeLa 1 24 20 10
2 25 24 9
4.5 Half-Life and Phosphorylation state of the p94RB Protein In Host Cells: The Distinct Properties of p94RB
The half-life of transiently expressed p94RB and pllO*8 proteins in transfected bladder carcinoma cell line, 5637 (ATCC HTB9) was measured by pulse-labeling of transfected 5637 cells with [35S]-methionine followed by a chase with excess unlabeled methionine (Figure 9) .
The bladder tumor cells were transfected in multiple dishes with either pllO8 (Figure 9, left) or p94RB (Figure 9, right) expression plasmids. Twenty- four hours after transfection the cells were labeled with [35S]-methionine and chased with excess unlabeled methionine for 0, 6, 12 and 24 hours, respectively. RB proteins were determined by immunoprecipitation.
The half-life of p94RB protein in the transfected 5637 cells was determined to be 12 hours. In contrast, the half-life of pllO*3 protein was 4-6 hours. Therefore, p94RB protein expressed in host tumor cells has a slower turnover, which is believed to contribute to its efficacy as a suppressor of both RB+ and RB- tumor cell replication.
The comparative phosphorylation states of pllO*8 and p94RB in transiently transfected 5637 cells were determined by Western blot analysis: cell-lysates were made from WI-38, parental 5637 and pβA-f-RB33 (expressing pllO*8, Section 4.3.5) or pBA-s-RB34
(expressing p94RB, Section 4.3.4) plasmid transfected 5637 cells approximately 24 hours after transfection. The basic protocal for Western blot analysis was described in Xu, H-J., et al., 1989, Oncogene. 4:807- 812. Each lane was loaded with 40μl of the lysate corresponding to 4 x 105 cultured cells. Proteins were separated by 8% SDS-PAGE and electroblotted to a PVDF membrane. After blocking with 3% non-fat milk in TBST (lOmM Tris-HCl, pH8.0, 150mM NaCI, 0.05% Tween 20), the membrane was incubated with MAb-1 monoclonal anti- RB antibody at 0.1 g/cm2 overnight. The blot was then probed by the Enhanced Chemiluminescence (ECL) (Amersham Corporation, Arlington Heights, Illinois) immunodetection method. X-ray films were exposed for 2 seconds (Figure 10, lane 1) or 30 seconds (Figure 10, lanes 2-4) .
Of particular interest was the fact that the RB- defective bladder carcinoma cell line, 5637, failed to phosphorylate the p94RB protein as shown by Western blot analysis (Figure 10, lane 4), although the pllO*3 proteins expressed in transfected 5637 cells were fully phosphorylated (Figure 10, lane 3) . Therefore, the presence of only unphosphorylated p94RB proteins may also account for the failure of transfected 5637 tumor cells to enter S phase, and this in turn may cause cellular senescence and cell death. 4.6. Treatment of Human Bladder Cancers In Vivo. The human bladder cancer represents an ideal model for practicing tumor suppressor gene therapy of solid tumors by infusing the p94RB protein expression retroviral vectors into the bladder. The original experimental model of human bladder cancer was established by Dr. Peter A. Jones and his colleagues (Ahlering, T.E., et al.. Cancer Res.. 1987, 47:6660-6665). It has been shown that human bladder tumor cells of RT4 cell line established from a superficial papillary tumor (which usually does not metastasize) produced tumors only locally when injected by a 22-gauge catheter into the bladder of female nude mice. In contrast, the EJ bladder carcinoma cells which were originally isolated from a more aggressive human bladder cancer produced invasive tumors in the nude mouse bladders which metastasized to the lung spontaneously (Ahlering, T.E., et al.. Cancer Res.. 1987, 47:6660-6665). Therefore, this model can be used for treatment of experimental bladder cancer by in vivo gene transfer with retroviral vectors.
Tumor cells from RB minus human bladder carcinoma cell line, 5637 (ATCC HTB9) and RB+ human bladder carcinoma cell line, SCaBER (ATCC HTB3) are injected directly into the bladders of female athymic (nu/nu) nude mice (6 to 8 weeks of age) by a catheter as initially reported by Jones and his colleagues (Ahlering, T.E., et al., Cancer Res. , 1987, 47:6660-6665) .
Development and progression of the nude mouse bladder tumors are monitored using a fiber-optical system to which a TV monitor is attached. The experimental tumors are subsequently treated with retrovirus vectors expressing the p94RB.
Supernatants with high virus titers are obtained from tissue culture media of selected PA317 clones expressing high level of human p94RB protein (Section 4.3.7) and confirmed as free of replication- competent virus prior to use. The retroviral vector suspension at high titers ranging from 4 x 104 to greater than 1 x 107 colony-forming unit (cfu)/ml, and more preferably at a titer greater than 1 x 106 cfu/ml is then infused directly into the mouse bladders via a catheter to treat the tumors. The skilled artisan will understand that such treatments can be repeated as many times as necessary via a catheter inserted into the bladder. The tumor regression following transferring the p94RB gene is monitored frequently via the fiber-optic system mentioned above.
The same procedure as described above is used for treating the human bladder cancer except that the retroviral vector suspension is infused into a human bladder bearing cancer.
4.7. In Vivo Studies Using an Orthotopic Lung Cancer Model
Human large cell lung carcinoma, NCI-H460 (ATCC HTB177) cells which have normal pllO*8 expression are injected into the right mainstream bronchus of athymic (nu/nu) nude mice (105 cells per mouse) . Three days later the mice are inoculated endobronchically with supernatant from the p94RB, or pllO*8 retrovirus producer cells daily for three consecutive days. Tumor formation is suppressed in the group of mice treated with the p94RB retrovirus supernatant. In contrast, in the other group, which is treated with pllO*8 retrovirus supernatant, the majority of mice develop endobronchial tumors. This indicates that the p94RB-expressing retrovirus inhibits growth of RB+ non- small cell lung carcinoma (NSCLC) cells, whereas the piio^-expressing retrovirus does not. 4.8. Treatment of Human Non-Small Cell Lung Cancers In Vivo .
Non-small cell lung cancer patients having an endobronchial tumor accessible to a bronchoscope, and also having a bronchial obstruction, are initially selected for p94RB gene therapy. Treatment is administered by bronchoscopy under topical or general anesthesia. To begin the procedure, as much gross tumor as possible is resected endoscopically. A transbronchial aspiration needle (21G) is passed through the biopsy channel of the bronchoscope.
The residual tumor site is injected with the appropriate retroviral vector supernatant (Section 4.3.7), adenovirus Ad-RB94 suspension (Section 4.3.8) or p94RB-expressing plasmid vector- liposome complexes (Section 4.3.4 and 4.3.6) at a volume of 5 ml to 10 ml. Protamine is added at a concentration of 5 μq/ml . The injections of therapeutic viral or plasmid supernatant comprising one or more of the vectors are administered around and within the tumor or tumors and into the submucosa adjacent to the tumor. The injections are repeated daily for five consecutive days and monthly therafter. The treatment may be continued as long as there is no tumor progression. After one year the patients are evaluated to deteri e whether it is appropriate to continue therapy.
In addition, as a precaution the patients wear a surgical mask for 24 hours following injection of the viral supernatant. All medical personnel wear masks routinely during bronchoscopy and injection of the viral supernatant. Anti-tussive is prescribed as necessary.
4.9 Treatment or Prevention of Human Lung Carcinomas With Liposome-Encapsulated Purified p94RB Protein In yet another alternative, target tumor or cancer cells are treated by introducing p94RB protein into cells in need of such treatment by any known method. For example, liposomes are artificial membrane vesicles that have been extensively studied for their usefulness as delivery vehicles of drugs, proteins and plasmid vectors both in vitro or in vivo (Mannino, R.J. et al., 1988, Biotechnigues. 6:682- 690) . Proteins such as erythrocyte anion transporter (Newton, A.C. and Huestis, W.H. , Biochemistry. 1988, 27:4655-4659), superoxide dismutase and catalase (Tanswell, A.K. et al., 1990, Biochmica et Biophysica Acta, 1044:269-274), and UV-DNA repair enzyme (Ceccoll, J. et al. Journal of Investigative Dermatology. 1989, 93:190-194) have been encapsulated at high efficiency with liposome vesicles and delivered into mammalian cells in vitro or in vivo.
Further, small-particle aerosols provide a method for the delivery of drugs for treatment of respiratory diseases. For example, it has been reported that drugs can be administered in small-particle aerosols by using liposomes as a vehicle. Administered via aerosols, the drugs are deposited rather uniformly on the surface of the nasopharynx, the traceheobronchial tree and in the pulmonary area (Knight, V. and Gilbert, B., 1988, European Journal of Clinical Microbiology and Infectious Diseases. 7:721-731) .
To treat or prevent lung cancers, the therapeutic p94RB protein is purified, for example, from recombinant baculovirus AcMNPV-RB94 infected insect cells by immunoaffinity chromatography (Sections 4.1 and 4.2) or any other convenient source. The p94RB protein is mixed with liposomes and incorporated into the liposome vesicles at high efficiency. The encapsulated p94RB is active. Since the aerosol delivery method is mild and well-tolerated by normal volunteers and patients, the p94RB-containing liposomes can be administered to treat patients suffering from lung cancers of any stage and/or to prevent lung cancers in high-risk population. The p94RB protein- containing liposomes are administered by nasal inhalation or by a endotracheal tube via small- particle aerosols at a dose sufficient to suppress abnormal cell proliferation. Aerosolization treatments are administered to a patient for 30 minutes, three times daily for two weeks, with repetition as needed. The p94RB protein is thereby delivered throughout the respiratory tract and the pulmonary area. The treatment may be continued as long as necessary. After one year the patent's overall condition will be evaluated to determine if continued therapy is appropriate.
4.10 p94RB Treatment is Non-Toxic to Normal Cells In Vitro
The retroviral vector, pLRB94RNL, expressing p94 RB protein as described supra in section 4.3.7. was introduced into normal mouse fibroblast-derived retrovirus-packaging cell line, PA317 (ATCC CRL9078) by LIPOFECTIN reagent (GIBCO BRL Life Technologies, Inc., Gaithersburg, MD) . Single cell colonies were isolated by selection in G418-containing medium and expanded into mass cultures. These clonal cells had been maintained over a one-year period of continuous culture, and stably expressed high levels of p94RB protein as determined by immunocytochemical staining (Figure 11) or by Western immunoblotting. These clones were indistinguishable from their parental normal mouse PA317 cells, or PA317 cells expressing human pllO*8 protein in terms of morphology (Figure 11) and growth rate. The results indicate that p94RB protein expression was non-toxic to normal cells in vi tro . 4.11 p94RB Treatment is Non-Toxic to Normal Tissues In Vivo
The retroviral plasmid vector, pLRB94RNL, expressing p94ω protein as described supra in section 4.3.7. was mixed with DMRIE/DOPE Liposomes (VICAL, Inc., San Diego, CA) and infused directly into the mouse urinary bladders via a catheter. Forty-eight hours after treatment, the mice were sacrificed and bladders excised. As demonstrated by immunohistochemical staining of the p94RB protein in paraffin-embedded tissue sections from the mouse bladders (Figure 12) , the liposome-encapsulated p94RB expressing retroviral plasmid vectors penetrated the mucosa of mouse bladders and expressed p94RB protein in the great majority of the transitional cells. The transitional epithelia expressing the p94RB were histologically normal (Figure 12, panel D, arrows), and were indistinguishable from the mucosa in untreated mouse bladders or mouse bladders treated with liposomes only (Figure 12) . The results from such animal experiments strongly suggest that the p94RB treatment, unlike the conventional cytotoxic cancer therapy, is non-toxic to normal tissues in vivo .
5. Deposit of Microorganisms
The following were deposited on February 10, 1993 with the American Type Culture Collection, 12301 Parklawn Drive, Rockville, Maryland 20852:
Escherichia coli DH5α ATCC Designation DHC-S-RB42 69240
DHB-S-RB34 69241
The present invention is not to be limited in scope by the specific embodiments described herein. Indeed, various modifications of the invention in addition to those described herein will become apparent to those skilled in the art from the foregoing description and accompanying figures. Such modifications are intended to fall within the scope of the claims. Various publications are cited herein, the disclosures of which are incorporated by reference in their entireties.
SEQUENCE LISTING
(1) GENERAL INFORMATION:
(i) APPLICANT: Xu, Hong-Ji Hu, Shi-Xue Benedict, William F.
(ii) TITLE OF INVENTION: BROAD SPECTRUM TUMOR SUPPRESSOR GENES, GENE
PRODUCTS, AND METHODS FOR TUMOR SUPPRESSION GENE THERAPY
(iii) NUMBER OF SEQUENCES: 3
(iv) CORRESPONDENCE ADDRESS:
(A) ADDRESSEE: Pennie & Edmonds
(B) STREET: 1155 Avenue of the Americas
(C) CITY: New York
(D) STATE: New York
(E) COUNTRY: U.S.A.
(F) ZIP: 10036-2711
(v) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: Floppy disk
(B) COMPUTER: IBM PC compatible
(C) OPERATING SYSTEM: PC-DOS/MS-DOS
(D) SOFTWARE: PatentIn Release #1.0, Version #1.25
(vi) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER: To be assigned
(B) FILING DATE: On even date herewith
(C) CLASSIFICATION:
(viii) ATTORNEY/AGENT INFORMATION:
(A) NAME: Poissant, Brian M
(B) REGISTRATION NUMBER: 28,462
(C) REFERENCE/DOCKET NUMBER: 7409-025-228
(ix) TELECOMMUNICATION INFORMATION:
(A) TELEPHONE: (212) 790-9090
(B) TELEFAX: (212) 869-9741/8864
(C) TELEX: 66141 PENNIE
(2) INFORMATION FOR SEQ ID NO:l:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 3232 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: not relevant
(ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 19..2469
(xi) SEQUENCE DESCRIPTION: SEQ ID Nθ:l:
GATCCCGACC TAGATGAG ATG TCG TTC ACT TTT ACT GAG CTA CAG AAA AAC 51
Met Ser Phe Thr Phe Thr Glu Leu Gin Lys Asn 1 5 10 ATA GAA ATC AGT GTC CAT AAA TTC TTT AAC TTA CTA AAA GAA ATT GAT 99 He Glu He Ser Val His Lys Phe Phe Asn Leu Leu Lys Glu He Asp 15 20 25
ACC AGT ACC AAA GTT GAT AAT GCT ATG TCA AGA CTG TTG AAG AAG TAT 147 Thr Ser Thr Lys Val Asp Asn Ala Met Ser Arg Leu Leu Lys Lys Tyr 30 35 40
GAT GTA TTG TTT GCA CTC TTC AGC AAA TTG GAA AGG ACA TGT GAA CTT 195 Asp Val Leu Phe Ala Leu Phe Ser Lys Leu Glu Arg Thr Cys Glu Leu 45 50 55
ATA TAT TTG ACA CAA CCC AGC AGT TCG ATA TCT ACT GAA ATA AAT TCT 243 He Tyr Leu Thr Gin Pro Ser Ser Ser He Ser Thr Glu He Asn Ser 60 65 70 75
GCA TTG GTG CTA AAA GTT TCT TGG ATC ACA TTT TTA TTA GCT AAA GGG 291 Ala Leu Val Leu Lys Val Ser Trp He Thr Phe Leu Leu Ala Lys Gly 80 85 90
GAA GTA TTA CAA ATG GAA GAT GAT CTG GTG ATT TCA TTT CAG TTA ATG 339 Glu Val Leu Gin Met Glu Asp Asp Leu Val He Ser Phe Gin Leu Met 95 100 105
CTA TGT GTC CTT GAC TAT TTT ATT AAA CTC TCA CCT CCC ATG TTG CTC 387 Leu Cys Val Leu Asp Tyr Phe He Lys Leu Ser Pro Pro Met Leu Leu 110 115 120
AAA GAA CCA TAT AAA ACA GCT GTT ATA CCC ATT AAT GGT TCA CCT CGA 435 Lys Glu Pro Tyr Lys Thr Ala Val He Pro He Asn Gly Ser Pro Arg 125 130 135
ACA CCC AGG CGA GGT CAG AAC AGG AGT GCA CGG ATA GCA AAA CAA CTA 483 Thr Pro Arg Arg Gly Gin Asn Arg Ser Ala Arg He Ala Lys Gin Leu 140 145 150 155
GAA AAT GAT ACA AGA ATT ATT GAA GTT CTC TGT AAA GAA CAT GAA TGT 531 Glu Asn Asp Thr Arg He He Glu Val Leu Cys Lys Glu His Glu Cys 160 165 170
AAT ATA GAT GAG GTG AAA AAT GTT TAT TTC AAA AAT TTT ATA CCT TTT 579 Asn He Asp Glu Val Lys Asn Val Tyr Phe Lys Asn Phe He Pro Phe 175 180 185
ATG AAT TCT CTT GGA CTT GTA ACA TCT AAT GGA CTT CCA GAG GTT GAA 627 Met Asn Ser Leu Gly Leu Val Thr Ser Asn Gly Leu Pro Glu Val Glu 190 195 200
AAT CTT TCT AAA CGA TAC GAA GAA ATT TAT CTT AAA AAT AAA GAT CTA 675 Asn Leu Ser Lys Arg Tyr Glu Glu He Tyr Leu Lys Asn Lys Asp Leu 205 210 215
GAT GCA AGA TTA TTT TTG GAT CAT GAT AAA ACT CTT CAG ACT GAT TCT 723 Asp Ala Arg Leu Phe Leu Asp His Asp Lys Thr Leu Gin Thr Asp Ser 220 225 230 235
ATA GAC AGT TTT GAA ACA CAG AGA ACA CCA CGA AAA AGT AAC CTT GAT 771 He Asp Ser Phe Glu Thr Gin Arg Thr Pro Arg Lys Ser Asn Leu Asp 240 245 250
GAA GAG GTG AAT GTA ATT CCT CCA CAC ACT CCA GTT AGG ACT GTT ATG 819 Glu Glu Val Asn Val He Pro Pro His Thr Pro Val Arg Thr Val Met 255 260 265
AAC ACT ATC CAA CAA TTA ATG ATG ATT TTA AAT TCA GCA AGT GAT CAA 867 Asn Thr He Gin Gin Leu Met Met He Leu Asn Ser Ala Ser Asp Gin 270 275 280 CCT TCA GAA AAT CTG ATT TCC TAT TTT AAC AAC TGC ACA GTG AAT CCA 915 Pro Ser Glu Asn Leu He Ser Tyr Phe Asn Asn Cys Thr Val Asn Pro 285 290 295
AAA GAA AGT ATA CTG AAA AGA GTG AAG GAT ATA GGA TAC ATC TTT AAA 963 Lys Glu Ser He Leu Lys Arg Val Lys Asp He Gly Tyr He Phe Lys 300 305 310 315
GAG AAA TTT GCT AAA GCT GTG GGA CAG GGT TGT GTC GAA ATT GGA TCA 1011 Glu Lys Phe Ala Lys Ala Val Gly Gin Gly Cys Val Glu He Gly Ser 320 325 330
CAG CGA TAC AAA CTT GGA GTT CGC TTG TAT TAC CGA GTA ATG GAA TCC 1059 Gin Arg Tyr Lys Leu Gly Val Arg Leu Tyr Tyr Arg Val Met Glu Ser 335 340 345
ATG CTT AAA TCA GAA GAA GAA CGA TTA TCC ATT CAA AAT TTT AGC AAA 1107 Met Leu Lys Ser Glu Glu Glu Arg Leu Ser He Gin Asn Phe Ser Lys 350 355 360
CTT CTG AAT GAC AAC ATT TTT CAT ATG TCT TTA TTG GCG TGC GCT CTT 1155 Leu Leu Asn Asp Asn He Phe His Met Ser Leu Leu Ala Cys Ala Leu 365 370 375
GAG GTT GTA ATG GCC ACA TAT AGC AGA AGT ACA TCT CAG AAT CTT GAT 1203 Glu Val Val Met Ala Thr Tyr Ser Arg Ser Thr Ser Gin Asn Leu Asp 380 385 390 395
TCT GGA ACA GAT TTG TCT TTC CCA TGG ATT CTG AAT GTG CTT AAT TTA 1251 Ser Gly Thr Asp Leu Ser Phe Pro Trp He Leu Asn Val Leu Asn Leu 400 405 410
AAA GCC TTT GAT TTT TAC AAA GTG ATC GAA AGT TTT ATC AAA GCA GAA 1299 Lys Ala Phe Asp Phe Tyr Lys Val He Glu Ser Phe He Lys Ala Glu 415 420 425
GGC AAC TTG ACA AGA GAA ATG ATA AAA CAT TTA GAA CGA TGT GAA CAT 1347 Gly Asn Leu Thr Arg Glu Met He Lys His Leu Glu Arg Cys Glu His 430 435 440
CGA ATC ATG GAA TCC CTT GCA TGG CTC TCA GAT TCA CCT TTA TTT GAT 1395 Arg He Met Glu Ser Leu Ala Trp Leu Ser Asp Ser Pro Leu Phe Asp 445 450 455
CTT ATT AAA CAA TCA AAG GAC CGA GAA GGA CCA ACT GAT CAC CTT GAA 1443 Leu He Lys Gin Ser Lys Asp Arg Glu Gly Pro Thr Asp His Leu Glu 460 465 470 475
TCT GCT TGT CCT CTT AAT CTT CCT CTC CAG AAT AAT CAC ACT GCA GCA 1491 Ser Ala Cys Pro Leu Asn Leu Pro Leu Gin Asn Asn His Thr Ala Ala 480 485 490
GAT ATG TAT CTT TCT CCT GTA AGA TCT CCA AAG AAA AAA GGT TCA ACT 1539 Asp Met Tyr Leu Ser Pro Val Arg Ser Pro Lys Lys Lys Gly Ser Thr 495 500 505
ACG CGT GTA AAT TCT ACT GCA AAT GCA GAG ACA CAA GCA ACC TCA GCC 1587 Thr Arg Val Asn Ser Thr Ala Asn Ala Glu Thr Gin Ala Thr Ser Ala 510 515 520
TTC CAG ACC CAG AAG CCA TTG AAA TCT ACC TCT CTT TCA CTG TTT TAT 1635 Phe Gin Thr Gin Lys Pro Leu Lys Ser Thr Ser Leu Ser Leu Phe Tyr 525 530 535
AAA AAA GTG TAT CGG CTA GCC TAT CTC CGG CTA AAT ACA CTT TGT GAA 1683 Lys Lys Val Tyr Arg Leu Ala Tyr Leu Arg Leu Asn Thr Leu Cys Glu 540 545 550 555 CGC CTT CTG TCT GAG CAC CCA GAA TTA GAA CAT ATC ATC TGG ACC CTT 1731 Arg Leu Leu Ser Glu His Pro Glu Leu Glu His He He Trp Thr Leu 560 565 570
TTC CAG CAC ACC CTG CAG AAT GAG TAT GAA CTC ATG AGA GAC AGG CAT 1779 Phe Gin His Thr Leu Gin Asn Glu Tyr Glu Leu Met Arg Asp Arg His 575 580 585
TTG GAC CAA ATT ATG ATG TGT TCC ATG TAT GGC ATA TGC AAA GTG AAG 1827 Leu Asp Gin He Met Met Cys Ser Met Tyr Gly He Cys Lys Val Lys 590 595 600
AAT ATA GAC CTT AAA TTC AAA ATC ATT GTA ACA GCA TAC AAG GAT CTT 1875 Asn He Asp Leu Lys Phe Lys He He Val Thr Ala Tyr Lys Asp Leu 605 610 615
CCT CAT GCT GTT CAG GAG ACA TTC AAA CGT GTT TTG ATC AAA GAA GAG 1923 Pro His Ala Val Gin Glu Thr Phe Lys Arg Val Leu He Lys Glu Glu 620 625 630 635
GAG TAT GAT TCT ATT ATA GTA TTC TAT AAC TCG GTC TTC ATG CAG AGA 1971 Glu Tyr Asp Ser He He Val Phe Tyr Asn Ser Val Phe Met Gin Arg 640 645 650
CTG AAA ACA AAT ATT TTG CAG TAT GCT TCC ACC AGG CCC CCT ACC TTG 2019 Leu Lys Thr Asn He Leu Gin Tyr Ala Ser Thr Arg Pro Pro Thr Leu 655 660 665
TCA CCA ATA CCT CAC ATT CCT CGA AGC CCT TAC AAG TTT CCT AGT TCA 2067 Ser Pro He Pro His He Pro Arg Ser Pro Tyr Lys Phe Pro Ser Ser 670 675 680
CCC TTA CGG ATT CCT GGA GGG AAC ATC TAT ATT TCA CCC CTG AAG AGT 2115 Pro Leu Arg He Pro Gly Gly Asn He Tyr He Ser Pro Leu Lys Ser 685 690 695
CCA TAT AAA ATT TCA GAA GGT CTG CCA ACA CCA ACA AAA ATG ACT CCA 2163 Pro Tyr Lys He Ser Glu Gly Leu Pro Thr Pro Thr Lys Met Thr Pro 700 705 710 715
AGA TCA AGA ATC TTA GTA TCA ATT GGT GAA TCA TTC GGG ACT TCT GAG 2211 Arg Ser Arg He Leu Val Ser He Gly Glu Ser Phe Gly Thr Ser Glu 720 725 730
AAG TTC CAG AAA ATA AAT CAG ATG GTA TGT AAC AGC GAC CGT GTG CTC 2259 Lys Phe Gin Lys He Asn Gin Met Val Cys Asn Ser Asp Arg Val Leu 735 740 745
AAA AGA AGT GCT GAA GGA AGC AAC CCT CCT AAA CCA CTG AAA AAA CTA 2307 Lys Arg Ser Ala Glu Gly Ser Asn Pro Pro Lys Pro Leu Lys Lys Leu 750 755 760
CGC TTT GAT ATT GAA GGA TCA GAT GAA GCA GAT GGA AGT AAA CAT CTC 2355 Arg Phe Asp He Glu Gly Ser Asp Glu Ala Asp Gly Ser Lys His Leu 765 770 775
CCA GGA GAG TCC AAA TTT CAG CAG AAA CTG GCA GAA ATG ACT TCT ACT 2403 Pro Gly Glu Ser Lys Phe Gin Gin Lys Leu Ala Glu Met Thr Ser Thr 780 785 790 795
CGA ACA CGA ATG CAA AAG CAG AAA ATG AAT GAT AGC ATG GAT ACC TCA 2451 Arg Thr Arg Met Gin Lys Gin Lys Met Asn Asp Ser Met Asp Thr Ser 800 805 810
AAC AAG GAA GAG AAA TGAGGATCTC AGGACCTTGG TGGACACTGT GTACACCTCT 2506 Asn Lys Glu Glu Lys 815 GGATTCATTG TCTCTCACAG ATGTGACTGT ATAACTTTCC CAGGTTCTGT TTATGGCCAC 2566
ATTTAATATC TTCAGCTCTT TTTGTGGATA TAAAATGTGC AGATGCAATT GTTTGGGTGA 2626
TTCCTAAGCC ACTTGAAATG TTAGTCATTG TTATTTATAC AAGATTGAAA ATCTTGTGTA 2686
AATCCTGCCA TTTAAAAAGT TGTAGCAGAT TGTTTCCTCT TCCAAAGTAA AATTGCTGTG 2746
CTTTATGGAT AGTAAGAATG GCCCTAGAGT GGGAGTCCTG ATAACCCAGG CCTGTCTGAC 2806
TACTTTGCCT TCTTTTGTAG CATATAGGTG ATGTTTGCTC TTGTTTTTAT TAATTTATAT 2866
GTATATTTTT TTAATTTAAC ATGAACACCC TTAGAAAATG TGTCCTATCT ATCATCCAAA 2926
TGCAATTTGA TTGACTGCCC ATTCACCAAA ATTATCCTGA ACTCTTCTGC AAAAATGGAT 2986
ATTATTAGAA ATTAGAAAAA AATTACTAAT TTTACACATT AGATTTTATT TTACTATTGG 3046
AATCTGATAT ACTGTGTGCT TGTTTTATAA AATTTTGCTT TTAATTAAAT AAAAGCTGGA 3106
AGCAAAGTAT AACCATATGA TACTATCATA CTACTGAAAC AGATTTCATA CCTCAGAATG 3166
TAAAAGAACT TACTGATTAT TTTCTTCATC CAACTTATGT TTTTAAATGA GGATTATTGA 3226
TAGTGG 3232
(2) INFORMATION FOR SEQ ID NO:2:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 3232 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: not relevant
(ii) MOLECULE TYPE: DNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:2:
GATCCCACTA TCAATAATCC TCATTTAAAA ACATAAGTTG GATGAAGAAA ATAATCAGTA 60
AGTTCTTTTA CATTCTGAGG TATGAAATCT GTTTCAGTAG TATGATAGTA TCATATGGTT 120
ATACTTTGCT TCCAGCTTTT ATTTAATTAA AAGCAAAATT TTATAAAACA AGCACACAGT 180
ATATCAGATT CCAATAGTAA AATAAAATCT AATGTGTAAA ATTAGTAATT TTTTTCTAAT 240
TTCTAATAAT ATCCATTTTT GCAGAAGAGT TCAGGATAAT TTTGGTGAAT GGGCAGTCAA 300
TCAAATTGCA TTTGGATGAT AGATAGGACA CATTTTCTAA GGGTGTTCAT GTTAAATTAA 360
AAAAATATAC ATATAAATTA ATAAAAACAA G GCAAACAT CACCTATATG CTACAAAAGA 420
AGGCAAAGTA GTCAGACAGG CCTGGGTTAT CAGGACTCCC ACTCTAGGGC CATTCTTACT 480
ATCCATAAAG CACAGCAATT TTACTTTGGA AGAGGAAACA ATCTGCTACA ACTTTTTAAA 540
TGGCAGGATT TACACAAGAT TTTCAATCTT GTATAAATAA CAATGACTAA CATTTCAAGT 600
GGCTTAGGAA TCACCCAAAC AATTGCATCT GCACATTTTA TATCCACAAA AAGAGCTGAA 660
GATATTAAAT GTGGCCATAA ACAGAACCTG GGAAAGTTAT ACAGTCACAT CTGTGAGAGA 720
CAATGAATCC AGAGGTGTAC ACAGTGTCCA CCAAGGTCCT GAGATCCTCA TTTCTCTTCC 780 TTGTTTGAGG TATCCATGCT ATCATTCATT TTCTGCTTTT GCATTCGTGT TCGAGTAGAA 840
GTCATTTCTG CCAGTTTCTG CTGAAATTTG GACTCTCCTG GGAGATGTTT ACTTCCATCT 900
GCTTCATCTG ATCCTTCAAT ATCAAAGCGT AGTTTTTTCA GTGGTTTAGG AGGGTTGCTT 960
CCTTCAGCAC TTCTTTTGAG CACACGGTCG CTGTTACATA CCATCTGATT TATTTTCTGG 1020
AACTTCTCAG AAGTCCCGAA TGATTCACCA ATTGATACTA AGATTCTTGA TCTTGGAGTC 1080
ATTTTTGTTG GTGTTGGCAG ACCTTCTGAA ATTTTATATG GACTCTTCAG GGGTGAAATA 1140
TAGATGTTCC CTCCAGGAAT CCGTAAGGGT GAACTAGGAA ACTTGTAAGG GCTTCGAGGA 1200
ATGTGAGGTA TTGGTGACAA GGTAGGGGGC CTGGTGGAAG CATACTGCAA AATATTTGTT 1260
TTCAGTCTCT GCATGAAGAC CGAGTTATAG AATACTATAA TAGAATCATA CTCCTCTTCT 1320
TTGATCAAAA CACGTTTGAA TGTCTCCTGA ACAGCATGAG GAAGATCCTT GTATGCTGTT 1380
ACAATGATTT TGAATTTAAG GTCTATATTC TTCACTTTGC ATATGCCATA CATGGAACAC 1440
ATCATAATTT GGTCCAAATG CCTGTCTCTC ATGAGTTCAT ACTCATTCTG CAGGGTGTGC 1500
TGGAAAAGGG TCCAGATGAT ATGTTCTAAT TCTGGGTGCT CAGACAGAAG GCGTTCACAA 1560
AGTGTATTTA GCCGGAGATA GGCTAGCCGA TACACTTTTT TATAAAACAG TGAAAGAGAG 1620
GTAGATTTCA ATGGCTTCTG GGTCTGGAAG GCTGAGGTTG CTTGTGTCTC TGCATTTGCA 1680
GTAGAATTTA CACGCGTAGT TGAACCTTTT TTCTTTGGAG ATCTTACAGG AGAAAGATAC 1740
ATATCTGCTG CAGTGTGATT ATTCTGGAGA GGAAGATTAA GAGGACAAGC AGATTCAAGG 1800
TGATCAGTTG GTCCTTCTCG GTCCTTTGAT TGTTTAATAA GATCAAATAA AGGTGAATCT 1860
GAGAGCCATG CAAGGGATTC CATGATTCGA TGTTCACATC GTTCTAAATG TTTTATCATT 1920
TCTCTTGTCA AGTTGCCTTC TGCTTTGATA AAACTTTCGA TCACTTTGTA AAAATCAAAG 1980
GCTTTTAAAT TAAGCACATT CAGAATCCAT GGGAAAGACA AATCTGTTCC AGAATCAAGA 2040
TTCTGAGATG TACTTCTGCT ATATGTGGCC ATTACAACCT CAAGAGCGCA CGCCAATAAA 2100
GACATATGAA AAATGTTGTC ATTCAGAAGT TTGCTAAAAT TTTGAATGGA TAATCGTTCT 2160
TCTTCTGATT TAAGCATGGA TTCCATTACT CGGTAATACA AGCGAACTCC AAGTTTGTAT 2220
CGCTGTGATC CAATTTCGAC ACAACCCTGT CCCACAGCTT TAGCAAATTT CTCTTTAAAG 2280
ATGTATCCTA TATCCTTCAC TCTTTTCAGT ATACTTTCTT TTGGATTCAC TGTGCAGTTG 2340
TTAAAATAGG AAATCAGATT TTCTGAAGGT TGATCACTTG CTGAATTTAA AATCATCATT 2400
AATTGTTGGA TAGTGTTCAT AACAGTCCTA ACTGGAGTGT GTGGAGGAAT TACATTCACC 2460
TCTTCATCAA GGTTACTTTT TCGTGGTGTT CTCTGTGTTT CAAAACTGTC TATAGAATCA 2520
GTCTGAAGAG TTTTATCATG ATCCAAAAAT AATCTTGCAT CTAGATCTTT ATTTTTAAGA 2580
TAAATTTCTT CGTATCGTTT AGAAAGATTT TCAACCTCTG GAAGTCCATT AGATGTTACA 2640
AGTCCAAGAG AATTCATAAA AGGTATAAAA TTTTTGAAAT AAACATTTTT CACCTCATCT 2700
ATATTACATT CATGTTCTTT ACAGAGAACT TCAATAATTC TTGTATCATT TTCTAGTTGT 2760
TTTGCTATCC GTGCACTCCT GTTCTGACCT CGCCTGGGTG TTCGAGGTGA ACCATTAATG 2820 GGTATAACAG CTGTTTTATA TGGTTCTTTG AGCAACATGG GAGGTGAGAG TTTAATAAAA 2880
TAGTCAAGGA CACATAGCAT TAACTGAAAT GAAATCACCA GATCATCTTC CATTTGTAAT 2940
ACTTCCCCTT TAGCTAATAA AAATGTGATC CAAGAAACTT TTAGCACCAA TGCAGAATTT 3000
ATTTCAGTAG ATATCGAACT GCTGGGTTGT GTCAAATATA TAAGTTCACA TGTCCTTTCC 3060
AATTTGCTGA AGAGTGCAAA CAATACATCA TACTTCTTCA ACAGTCTTGA CATAGCATTA 3120
TCAACTTTGG TACTGGTATC AATTTCTTTT AGTAAGTTAA AGAATTTATG GACACTGATT 3180
TCTATGTTTT TCTGTAGCTC AGTAAAAGTG AACGACATCT CATCTAGGTC GG 3232
(2) INFORMATION FOR SEQ ID NO:3:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 816 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:3:
Met Ser Phe Thr Phe Thr Glu Leu Gin Lys Asn He Glu He Ser Val 1 5 10 15
His Lys Phe Phe Asn Leu Leu Lys Glu He Asp Thr Ser Thr Lys Val 20 25 30
Asp Asn Ala Met Ser Arg Leu Leu Lys Lys Tyr Asp Val Leu Phe Ala 35 40 45
Leu Phe Ser Lys Leu Glu Arg Thr Cys Glu Leu He Tyr Leu Thr Gin 50 55 60
Pro Ser Ser Ser He Ser Thr Glu He Asn Ser Ala Leu Val Leu Lys 65 70 75 80
Val Ser Trp He Thr Phe Leu Leu Ala Lys Gly Glu Val Leu Gin Met 85 90 95
Glu Asp Asp Leu Val He Ser Phe Gin Leu Met Leu Cys Val Leu Asp 100 105 110
Tyr Phe He Lys Leu Ser Pro Pro Met Leu Leu Lys Glu Pro Tyr Lys 115 120 125
Thr Ala Val He Pro He Asn Gly Ser Pro Arg Thr Pro Arg Arg Gly 130 135 140
Gin Asn Arg Ser Ala Arg He Ala Lys Gin Leu Glu Asn Asp Thr Arg 145 150 155 160
He He Glu Val Leu Cys Lys Glu His Glu Cys Asn He Asp Glu Val 165 170 175
Lys Asn Val Tyr Phe Lys Asn Phe He Pro Phe Met Asn Ser Leu Gly 180 185 190
Leu Val Thr Ser Asn Gly Leu Pro Glu Val Glu Asn Leu Ser Lys Arg 195 200 205
Tyr Glu Glu He Tyr Leu Lys Asn Lys Asp Leu Asp Ala Arg Leu Phe 210 215 220 Leu Asp His Asp Lys Thr Leu Gin Thr Asp Ser He Asp Ser Phe Glu 225 230 235 240
Thr Gin Arg Thr Pro Arg Lys Ser Asn Leu Asp Glu Glu Val Asn Val 245 250 255
He Pro Pro His Thr Pro Val Arg Thr Val Met Asn Thr He Gin Gin 260 265 270
Leu Met Met He Leu Asn Ser Ala Ser Asp Gin Pro Ser Glu Asn Leu 275 280 285
He Ser Tyr Phe Asn Asn Cys Thr Val Asn Pro Lys Glu Ser He Leu 290 295 300
Lys Arg Val Lys Asp He Gly Tyr He Phe Lys Glu Lys Phe Ala Lys 305 310 315 320
Ala Val Gly Gin Gly Cys Val Glu He Gly Ser Gin Arg Tyr Lys Leu 325 330 335
Gly Val Arg Leu Tyr Tyr Arg Val Met Glu Ser Met Leu Lys Ser Glu 340 345 350
Glu Glu Arg Leu Ser He Gin Asn Phe Ser Lys Leu Leu Asn Asp Asn 355 360 365
He Phe His Met Ser Leu Leu Ala Cys Ala Leu Glu Val Val Met Ala 370 375 380
Thr Tyr Ser Arg Ser Thr Ser Gin Asn Leu Asp Ser Gly Thr Asp Leu 385 390 395 400
Ser Phe Pro Trp He Leu Asn Val Leu Asn Leu Lys Ala Phe Asp Phe 405 410 415
Tyr Lys Val He Glu Ser Phe He Lys Ala Glu Gly Asn Leu Thr Arg 420 425 430
Glu Met He Lys His Leu Glu Arg Cys Glu His Arg He Met Glu Ser 435 440 445
Leu Ala Trp Leu Ser Asp Ser Pro Leu Phe Asp Leu He Lys Gin Ser 450 455 460
Lys Asp Arg Glu Gly Pro Thr Asp His Leu Glu Ser Ala Cys Pro Leu 465 470 475 480
Asn Leu Pro Leu Gin Asn Asn His Thr Ala Ala Asp Met Tyr Leu Ser 485 490 495
Pro Val Arg Ser Pro Lys Lys Lys Gly Ser Thr Thr Arg Val Asn Ser 500 505 510
Thr Ala Asn Ala Glu Thr Gin Ala Thr Ser Ala Phe Gin Thr Gin Lys 515 520 525
Pro Leu Lys Ser Thr Ser Leu Ser Leu Phe Tyr Lys Lys Val Tyr Arg 530 535 540
Leu Ala Tyr Leu Arg Leu Asn Thr Leu Cys Glu Arg Leu Leu Ser Glu 545 550 555 560
His Pro Glu Leu Glu His He He Trp Thr Leu Phe Gin His Thr Leu 565 570 575
Gin Asn Glu Tyr Glu Leu Met Arg Asp Arg His Leu Asp Gin He Met 580 585 590
Met Cys Ser Met Tyr Gly He Cys Lys Val Lys Asn He Asp Leu Lys 595 600 605
Phe Lys He He Val Thr Ala Tyr Lys Asp Leu Pro His Ala Val Gin 610 615 620
Glu Thr Phe Lys Arg Val Leu He Lys Glu Glu Glu Tyr Asp Ser He 625 630 635 640
He Val Phe Tyr Asn Ser Val Phe Met Gin Arg Leu Lys Thr Asn He 645 650 655
Leu Gin Tyr Ala Ser Thr Arg Pro Pro Thr Leu Ser Pro He Pro His 660 665 670
He Pro Arg Ser Pro Tyr Lys Phe Pro Ser Ser Pro Leu Arg He Pro 675 680 685
Gly Gly Asn He Tyr He Ser Pro Leu Lys Ser Pro Tyr Lys He Ser 690 695 700
Glu Gly Leu Pro Thr Pro Thr Lys Met Thr Pro Arg Ser Arg He Leu 705 710 715 720
Val Ser He Gly Glu Ser Phe Gly Thr Ser Glu Lys Phe Gin Lys He 725 730 735
Asn Gin Met Val Cys Asn Ser Asp Arg Val Leu Lys Arg Ser Ala Glu 740 745 750
Gly Ser Asn Pro Pro Lys Pro Leu Lys Lys Leu Arg Phe Asp He Glu 755 760 765
Gly Ser Asp Glu Ala Asp Gly Ser Lys His Leu Pro Gly Glu Ser Lys 770 775 780
Phe Gin Gin Lys Leu Ala Glu Met Thr Ser Thr Arg Thr Arg Met Gin 785 790 795 800
Lys Gin Lys Met Asn Asp Ser Met Asp Thr Ser Asn Lys Glu Glu Lys 805 810 815
• -
International Application No- PCT/
Figure imgf000080_0001
-77/2-
Intemational Application No: PCT/
Form PCT/RO/134 (cont.)
American Type Culture Collection
12301 Parklawn Drive Rockville, MD 20852 US
Accession No. Date of Deposit 69241 February 10, 1993

Claims

- 78
We claim: 1. A method of treating a disease characterized by abnormal cellular proliferation in a mammal by a process comprising the steps of: a. administering an effective dose of a p94RB encoding expression vector to a mammal having a disease characterized by abnormally proliferating cells, such that said expression vector is inserted into said abnormally proliferating cells, said expression vector comprising a gene encoding p94RB; and b. expressing p94RB in said abnormally proliferating cells in an amount effective to suppress proliferation of said abnormally proliferating cells; and wherein said p94RB encoding expression vector comprises a p94RB encoding gene, said gene encoding a protein having an amino acid sequence substantially according to SEQ ID NO:3, provided that said protein is not pllORB.
2. The method according to claim 1 wherein said p94RB encoding gene encodes a protein having an amino acid sequence according to SEQ ID NO:3.
3. The method according to claim 1 wherein said p94RB encoding gene has a DNA sequence substantially according to SEQ ID NO:l.
4. The method according to claim 3 wherein said p94RB encoding gene has a DNA sequence according to SEQ ID NO:l.
5. The method according to claim 2 wherein said expression vector is selected from the group consisting of a plasmid and a viral vector. - 79 -
6. The method according to claim 4 wherein said expression vector is selected from the group consisting of a plasmid and a viral vector.
7. The method according to claim 5 wherein said viral vector is selected from a group consisting of a retroviral vector, an adenoviral vector and a herpesviral vector, wherein said p94RB encoding gene is under the control of a promoter selected from the group consisting of a retroviral promoter, an adenoviral promoter, a CMV promoter and a β-actin promoter.
8. The method according to claim 6 wherein said viral vector is selected from a group consisting of a retroviral vector, an adenoviral vector and a herpesviral vector wherein said p94ω encoding gene is under the control of a promoter selected from the group consisting of a retroviral promoter, an adenoviral promoter, a CMV promoter and a β-actin promoter.
9. The method according to claim 8 wherein said expression vector is selected from the group consisting of plasmid pCMV-s-RB42 and plasmid pβA-s- RB34.
10. The method according to claims 1 , 8 or 9 wherein said expression vector is inserted into said abnormally proliferating cells by a method selected from the group consisting of viral infection or transduction, liposome-mediated transfection, polybrene- ediated transfection and CaP04 mediated transfection. - 80 -
11. The method according to claim 1 wherein said abnormally proliferating cells are tumor or cancer cells and said mammal is a human.
12. The method according to claim 11 wherein said tumor or cancer cells are selected from the group consisting of carcinoma and sarcoma cells.
13. The method according to claim 11 wherein tumor or cancer cells are selected from the group consisting of a bladder carcinoma, a lung carcinoma, a breast carcinoma, a prostate carcinoma, a fibrosarcoma, an osteosarcoma, and a cervical carcinoma.
14. The method according to claim 11 wherein said tumor or cancer cells are cells having at least one genetically defective tumor suppressor gene or oncogene selected from the group consisting of an RB, a p53, a c-tπyc an N-ras and a c-yes-1 gene.
15. The method according to claim 11 wherein said tumor or cancer cells have no detectable genetic defect of a tumor suppressor gene, and the tumor suppressor gene is selected from the group consisting of an RB gene and a p53 gene.
16. The method according to claim 12 wherein said carcinoma cells are bladder carcinoma cells and said step of administering said expression vector to treat said bladder carcinoma cells is by means of an infusion of said expression vector into a bladder in need of such treatment.
17. A DNA molecule encoding p94RB having an amino acid sequence substantially according to SEQ ID NO:3, - 81 -
provided that said DNA molecule does not also code for pllO™.
18. The DNA molecule according to claim 17, said DNA molecule coding for a protein having an amino acid sequence according to SEQ ID NO:3.
19. The DNA molecule according to claim 17, said DNA molecule having a DNA sequence substantially according to SEQ ID NO:l, provided that said DNA molecule does not also code for pllO*3.
20. The DNA molecule according to claim 17, said DNA molecule having a DNA sequence according to SEQ ID N0:l.
21. An expression vector comprising said DNA molecule according to claim 18, capable of inserting said p94ω encoding DNA molecule into a mammalian host cell and of expressing p94RB therein.
22. An expression vector comprising said DNA molecule according to claim 20, capable of inserting said p94ω encoding DNA molecule into a mammalian host cell and of expressing p94RB therein.
23. The expression vector according to claim 21, wherein said expression vector is selected from the group consisting of a plasmid and a viral vector.
24. The expression vector according to claim 22, wherein said expression vector is selected from the group consisting of a plasmid and a viral vector.
25. The expression vector according to claim 23 wherein said viral vector is selected from a group consisting of a retroviral vector, an adenoviral 82 -
vector and a herpesviral vector, and wherein said p94ω encoding gene is under the control of a promoter selected from the group consisting of a retroviral promoter, an adenoviral promoter, a CMV promoter and a β-actin promoter.
26. The expression vector according to claim 24 wherein said viral vector is selected from a group consisting of a retroviral vector, an adenoviral vector and a herpesviral vector and wherein said p94RB encoding gene is under the control of a promoter selected from the group consisting of a retroviral promoter, an adenoviral promoter, a CMV promoter and a β-actin promoter.
27. The expression vector according to claim 24 wherein said expression vector is plasmid pCMV-s-RB42 and plasmid pβA-s-RB34.
28. A composition suitable for treating a tumor or cancer in a mammal comprising an effective amount of the expression vector according to claim 21, together with a suitable carrier or vehicle.
29. A composition suitable for treating a tumor or cancer in a mammal comprising an effective amount of the expression vector according to claim 22, together with a suitable carrier or vehicle.
30. A composition according to claims 28 or 29 wherein said carrier or vehicle comprises an encapsulating liposome.
31. A p94RB protein comprising a polypeptide having an amino acid sequence substantially according to SEQ ID NO:3; provided that said protein is not pllO . - 83 -
32. A p94RB protein according to claim 31 comprising a polypeptide having an amino acid sequence according to SEQ ID NO:3.
33. A composition suitable for treating a tumor or cancer in a mammal comprising an effective amount of p94RB according to claim 31, together with a suitable carrier or vehicle.
34. A composition suitable for treating a tumor or cancer in a mammal comprising an effective amount of p94RB according to claim 32, together with a suitable carrier or vehicle.
35. A composition according to claims 33 or 34 wherein said carrier or vehicle comprises an encapsulating liposome.
36. A method of producing a p94RB protein comprising the steps of: a. inserting a compatible expression vector comprising a p94RB encoding gene into a host cell; and b. causing said host cell to express p94RB protein.
37. The method according to claim 36 wherein said host cell is selected from the group consisting of a prokaryotic host cell and a eukaryotic host cell.
38. The method according to claim 32 wherein said eukaryotic host cell is a mammalian host cell and said expression vector is compatible with said mammalian host cell.
39. The method according to claim 38 wherein said expression vector is selected from the group - 84 -
consisting of plasmid pCMV-s-RB42 and plasmid pβA-s- RB34.
40. The method according to claim 37 wherein said host cell is an insect host cell and said expression vector is a plasmid or a viral vector compatible with said insect host cell.
41. The method according to claim 40 wherein said baculovirus vector is AcMNPV-RB94.
42. A method of treating abnormally proliferating cells of a mammal ex vivo by a process comprising the steps of: a. removing a tissue sample in need of treatment from a mammal, said tissue sample comprising abnormally proliferating cells; b. contacting said tissue sample in need of treatment with an effective dose of a p94RB encoding expression vector; c. expressing said p94RB in said abnormally proliferating cells in amounts effective to suppress proliferation of said abnormally proliferating cells; and d. returning said treated tissue sample to said mammal or placing said tissue sample into another mammal.
43. A method of treating a disease characterized by abnormal cellular proliferation in a mammal by administering p94RB protein to a mammal having a disease characterized by abnormally proliferating cells, such that said p94RB protein is inserted into said abnormally proliferating cells in amounts effective to suppress abnormal proliferation of said cells. - 85 -
44. The method according to claim 43 wherein said p94RB protein has an amino acid sequence substantially according to SEQ ID NO:3, provided that said protein is not pllO*8.
45. The method according to claim 43 wherein said p94RB protein has an amino acid sequence according to SEQ ID NO:3.
46. The method according to claim 43 wherein said abnormally proliferating cells are tumor or cancer cells, and said mammal is a human patient.
47. The method according to claim 46 wherein said p94RB protein is encapsulated in a liposome carrier and said p94RB protein is inserted into said abnormally proliferating cells by fusion of said liposome encapsulated p94RB protein with said abnormally proliferating cells.
48. The method according to claim 46 wherein said tumor or cancer cells are selected from the group consisting of a bladder carcinoma, a lung carcinoma, a breast carcinoma, a prostate carcinoma, a fibrosarcoma, an osteosarcoma, and a cervix carcinoma.
49. The method according to claim 46 wherein said tumor or cancer cells are cells having one or more genetically defective tumor suppressor genes and oncogenes selected from the group consisting of an RB, a p53, a c-myc, an N-ras and a c-yes-1 gene.
50. The method according to claim 46 wherein said tumor or cancer cells are cells having no detectable genetic defect of a tumor suppressor gene selected from the group consisting of an RB gene and a p53 gene. - 86 -
51. The method according to claim 48 wherein said tumor or cancer cells are lung carcinoma cells and said step of administering said p94RB protein to treat said lung carcinoma cells is by means of an infusion of said liposome-encapsulated p94RB protein into the respiratory tract and the pulmonary area in need of such treatment.
52. A method of treating abnormally proliferating cells of a mammal ex vivo by a process comprising the steps of: a. removing a tissue sample in need of treatment from a mammal, said tissue sample comprising abnormally proliferating cells; b. contacting said tissue sample in need of treatment with an effective dose of a p94RB protein; and c. returning said treated tissue sample to said mammal or placing said tissue sample into another mammal; and said p94RB protein has the amino acid sequence substantially according to SEQ ID NO:3.
53. A method of treating abnormally proliferating cells by a process comprising the steps of: a. inserting a p94RB encoding expression vector into abnormally proliferating cells of a mammal; and b. expressing said p94RB therein in amounts effective to suppress proliferation of said abnormally proliferating cells and said p94RB encoding expression vector comprises a p94RBencoding gene, said gene encoding a protein having an amino acid sequence substantially according to SEQ ID NO:3.
PCT/US1994/003211 1993-03-25 1994-03-24 Broad spectrum tumor suppressor genes, gene products and methods for tumor suppression gene therapy WO1994021115A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
EP94911697A EP0693874A4 (en) 1993-03-25 1994-03-24 Broad spectrum tumor suppressor genes, gene products and methods for tumor suppression gene therapy
AU64154/94A AU697446B2 (en) 1993-03-25 1994-03-24 Broad spectrum tumor suppressor genes, gene products and methods for tumor suppression gene therapy
JP52137494A JP3739787B2 (en) 1993-03-25 1994-03-24 Wide range of tumor suppressor genes, gene products and tumor suppressor gene therapy
KR1019950704162A KR100326136B1 (en) 1993-03-25 1994-03-24 Broad spectrum tumor suppressor genes, gene products and methods for tumor suppression gene therapy

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US038,760 1993-03-25
US08/038,760 US5496731A (en) 1993-03-25 1993-03-25 Broad-spectrum tumor suppressor genes, gene products and methods for tumor suppressor gene therapy

Publications (1)

Publication Number Publication Date
WO1994021115A1 true WO1994021115A1 (en) 1994-09-29

Family

ID=21901738

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1994/003211 WO1994021115A1 (en) 1993-03-25 1994-03-24 Broad spectrum tumor suppressor genes, gene products and methods for tumor suppression gene therapy

Country Status (9)

Country Link
US (2) US5496731A (en)
EP (1) EP0693874A4 (en)
JP (1) JP3739787B2 (en)
KR (1) KR100326136B1 (en)
AU (1) AU697446B2 (en)
CA (1) CA2159085A1 (en)
IL (1) IL109092A (en)
TW (1) TW379227B (en)
WO (1) WO1994021115A1 (en)

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995016051A2 (en) * 1993-12-03 1995-06-15 Board Of Regents, The University Of Texas System Methods and compositions for the suppression of neu mediated transformation
US5641484A (en) * 1990-12-04 1997-06-24 Board Of Regents, The University Of Texas System Methods for the suppression of neu mediated tumors by adenoviral E1A and SV40 large T antigen
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
WO1997032991A2 (en) * 1996-03-06 1997-09-12 Avigen, Inc. Gene therapy for the treatment of solid tumors using recombinant adeno-associated virus vectors
US5792751A (en) * 1992-04-13 1998-08-11 Baylor College Of Medicine Tranformation of cells associated with fluid spaces
WO1998037091A2 (en) * 1997-02-20 1998-08-27 Board Of Regents, The University Of Texas System Modified retinoblastoma tumor suppressor proteins
WO1998037185A2 (en) * 1997-02-20 1998-08-27 The Board Of Regents Of The University Of Texas System Vectors for controlled gene expression
WO1999002190A1 (en) * 1997-07-11 1999-01-21 Genzyme Corporation Organ-specific targeting of cationic amphiphile/retinoblastoma encoding dna complexes for gene therapy
US5869040A (en) * 1995-06-07 1999-02-09 Biogen, Inc Gene therapy methods and compositions
US5939401A (en) * 1994-12-09 1999-08-17 Genzyme Corporation Cationic amphiphile compositions for intracellular delivery of therapeutic molecules
US5948767A (en) * 1994-12-09 1999-09-07 Genzyme Corporation Cationic amphiphile/DNA complexes
EP1006187A2 (en) * 1995-03-29 2000-06-07 Millennium Pharmaceuticals, Inc. Compositions for the diagnosis, prevention, and treatment of tumor progression
US6197754B1 (en) 1996-10-18 2001-03-06 Board Of Regents, The University Of Texas System Suppression of tumor growth by a mini-E1A gene
US6251597B1 (en) 1996-03-29 2001-06-26 Millennium Pharmaceuticals, Inc. Methods for detecting fohy030
US6312909B1 (en) 1996-03-29 2001-11-06 Millennium Pharmaceuticals, Inc. Compositions and methods for the diagnosis prevention and treatment of tumor progression
US6331524B1 (en) 1994-12-09 2001-12-18 Genzyme Corporation Organ-specific targeting of cationic amphiphile / DNA complexes for gene therapy
US6383814B1 (en) 1994-12-09 2002-05-07 Genzyme Corporation Cationic amphiphiles for intracellular delivery of therapeutic molecules
US6395712B1 (en) 1996-03-20 2002-05-28 Board Of Regents, The University Of Texas System Sensitization of HER-2/neu overexpressing cancer cells to chemotherapy
US6794185B1 (en) 1995-03-29 2004-09-21 Millennium Pharmaceuticals, Inc. fohy030 nucleic acid molecules
US6825320B1 (en) 1995-03-29 2004-11-30 Millenium Pharmaceuticals, Inc. FOHY03 polypeptides

Families Citing this family (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7105156B1 (en) * 1987-09-17 2006-09-12 The Regents Of The University Of California Method of using an adenoviral vector encoding a retinoblastoma protein to treat hyperproliferating cells
CA2042093C (en) * 1990-05-09 2002-12-24 Gyula Hadlaczky Cell line carrying an excess of mammalian centromeres
AU663702B2 (en) 1991-03-06 1995-10-19 Board Of Regents, The University Of Texas System Methods and compositions for the selective inhibition of gene expression
US6410010B1 (en) * 1992-10-13 2002-06-25 Board Of Regents, The University Of Texas System Recombinant P53 adenovirus compositions
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
AU7404994A (en) * 1993-07-30 1995-02-28 Regents Of The University Of California, The Endocardial infusion catheter
US20030060435A1 (en) * 1994-05-31 2003-03-27 Serge Carillo Method of cancer treatment by p53 protein control
ES2204980T3 (en) * 1995-06-30 2004-05-01 Takara Bio Inc. NEW PROMOTER AND GENES EXPRESSION METHOD THAT USES IT.
US5770720A (en) * 1995-08-30 1998-06-23 Barnes-Jewish Hospital Ubiquitin conjugating enzymes having transcriptional repressor activity
CZ296810B6 (en) * 1995-11-30 2006-06-14 Board Of Regents, The University Of Texas System Medicaments for treating tumors
US6392069B2 (en) * 1996-01-08 2002-05-21 Canji, Inc. Compositions for enhancing delivery of nucleic acids to cells
US5789244A (en) * 1996-01-08 1998-08-04 Canji, Inc. Compositions and methods for the treatment of cancer using recombinant viral vector delivery systems
US20040014709A1 (en) * 1996-01-08 2004-01-22 Canji, Inc. Methods and compositions for interferon therapy
US6077697A (en) * 1996-04-10 2000-06-20 Chromos Molecular Systems, Inc. Artificial chromosomes, uses thereof and methods for preparing artificial chromosomes
US20020160970A1 (en) * 1996-04-10 2002-10-31 Gyula Hadlaczky Artificial chromosomes, uses thereof and methods for preparing artificial chromosomes
US20030033617A1 (en) * 1996-04-10 2003-02-13 Gyula Hadlaczky Artificial chromosomes, uses thereof and methods for preparing artificial chromosomes
US6025155A (en) * 1996-04-10 2000-02-15 Chromos Molecular Systems, Inc. Artificial chromosomes, uses thereof and methods for preparing artificial chromosomes
US20030064949A1 (en) * 1998-02-17 2003-04-03 Loretta Nielsen Combined tumor suppressor gene therapy and chemotherapy in the treatment of neoplasms
US20030060434A1 (en) * 1997-02-18 2003-03-27 Loretta Nielsen Combined tumor suppressor gene therapy and chemotherapy in the treatment of neoplasms
KR19980077401A (en) * 1997-04-18 1998-11-16 문우철 Gene therapy for cancer containing wild type p53 gene vector and cationic liposomes
US6663856B1 (en) 1997-06-02 2003-12-16 Thomas Jefferson University Method of inhibiting cancer cell growth using a vector expressing pRb2/P130
EP1007099A4 (en) * 1997-07-11 2004-11-24 Univ Brandeis Method of inducing apoptosis by reducing the level of thiamin
EP1015015B1 (en) 1997-08-21 2007-05-23 Thomas Jefferson University pRb2/p130 PEPTIDE INHIBITORS OF cdk2 KINASE ACTIVITY
US6372720B1 (en) 1998-02-05 2002-04-16 Kenneth J. Longmuir Liposome fusion and delivery vehicle
US6648552B1 (en) * 1999-10-14 2003-11-18 Bechtel Bwxt Idaho, Llc Sensor system for buried waste containment sites
US6841362B1 (en) 2000-02-29 2005-01-11 The Trustees Of Columbia University In The City Of New York Melanoma differentiation associated gene-7 promoter and uses thereof
US7229822B1 (en) * 2000-02-29 2007-06-12 Univ Columbia Melanoma differentation associated gene-5 and vectors and cells containing same
EP2166087B1 (en) * 2000-09-28 2012-11-14 Bioriginal Food & Science Corp. FAD6 fatty acid desaturase family member and uses thereof
JP2005501011A (en) * 2001-06-08 2005-01-13 ユタ ベンチャー ザ セカンド リミテッド パートナーシップ Tissue-specific inner membrane protein
EP1629085A2 (en) * 2003-06-04 2006-03-01 CANJI, Inc. Transfection agents
CN1851455B (en) * 2005-04-22 2010-11-10 复旦大学附属中山医院 Tumour marker-serum protein fingerprint detecting method
US20090054333A1 (en) * 2006-10-17 2009-02-26 Antonio Giordano Peptide inhibitors of cyclin-dependent kinase activity and uses thereof
US7470670B2 (en) * 2006-10-17 2008-12-30 Sbarro Health Research Organization, Inc. Peptide inhibitors of cyclin-dependent kinase activity and uses thereof

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0293266A2 (en) * 1987-05-29 1988-11-30 Research Development Foundation A method for detecting the predisposition to retinoblastoma and a method for detecting a retinoblastoma gene in tumors using a retinoblastoma gene probe
WO1990005180A1 (en) * 1988-10-31 1990-05-17 The Regents Of The University Of California Products and methods for controlling the suppression of the neoplastic phenotype
WO1991015580A1 (en) * 1990-04-10 1991-10-17 Research Development Foundation Gene therapy for cell proliferative diseases
WO1992022640A1 (en) * 1991-06-10 1992-12-23 Research Development Foundation Rb TRANSFERRED CELLS AND METHOD

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5174993A (en) * 1981-12-24 1992-12-29 Health Research Inc. Recombinant avipox virus and immunological use thereof
US4740463A (en) * 1984-04-13 1988-04-26 Massachusetts Institute Of Technology Methods and artificial genes for antagonizing the function of an oncogene
CA1341576C (en) * 1986-08-11 2008-07-08 Thaddeus P. Dryja Diagnosis of retinoblastoma
DE10399032I1 (en) * 1987-08-28 2004-01-29 Health Research Inc Recombinant viruses.
US4942123A (en) * 1987-09-17 1990-07-17 The Regents Of The University Of California ppRB110 -phosphoprotein the retinoblastoma susceptibility gene product
JP3163090B2 (en) * 1988-01-21 2001-05-08 マサチューセッツ アイ アンド イヤー インファーマリー Retinoblastoma diagnostic agent
CA2014473A1 (en) * 1989-04-14 1990-10-14 Hong-Ji Xu Purified, highly specific antibodies against gene products including retinoblastoma and method
CA2091759A1 (en) * 1990-09-17 1992-03-18 Dale E. Bredesen Method and composition for controlling proliferation of cells

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0293266A2 (en) * 1987-05-29 1988-11-30 Research Development Foundation A method for detecting the predisposition to retinoblastoma and a method for detecting a retinoblastoma gene in tumors using a retinoblastoma gene probe
WO1990005180A1 (en) * 1988-10-31 1990-05-17 The Regents Of The University Of California Products and methods for controlling the suppression of the neoplastic phenotype
WO1991015580A1 (en) * 1990-04-10 1991-10-17 Research Development Foundation Gene therapy for cell proliferative diseases
WO1992022640A1 (en) * 1991-06-10 1992-12-23 Research Development Foundation Rb TRANSFERRED CELLS AND METHOD

Non-Patent Citations (19)

* Cited by examiner, † Cited by third party
Title
Cancer Research, Volume 52, Number 22, issued 15 November 1992, A. BANERJEE et al., "Changes in Growth and Tumorigenicity Following Reconstitution of Retinoblastoma Gene Function in Various Human Cancer Cell Types by Microcell Transfer of Chromosome 13", pages 6297-6304, see entire article. *
Cell, Volume 68, issued 10 January 1992, M.A. ROSENFELD et al., "In Vivo Transfer of the Human Cystic Fibrosis Transmembrane Conductance Regulator Gene to the Airway Epithelium", pages 143-155, see entire article. *
Human Gene Therapy, Volume 2, issued 1991, K.W. CULVER et al., "Lymphocyte Gene Therapy", pages 107-109, see entire article. *
Journal of Clinical Investigation, Volume 85, Number 4, issued April 1990, W.F. BENEDICT et al., "Role of the Retinoblastoma Gene in the Initiation and Progression of Human Cancer", pages 988-993, see entire article. *
Nature Genetics, Volume 1, issued August 1992, J.H. WOLFE et al., "Herpesvirus Vector Gene Transfer and Expression of Beta-Glucuronidase in the Central Nervous System of MPS VII Mice", pages 379-384, see entire article. *
Nature, Volume 329, issued 15 October 1987, W.H. LEE et al., "The Retinoblastoma Susceptibility Gene Encodes a Nuclear Phosphoprotein Associated with DNA Binding Activity", pages 642-645, see entire article. *
Nature, Volume 360, issued 12 November 1992, D.W. GOODRICH et al., "Abrogation by c-myc of G1 Phase Arrest Induces by RB Protein but not by p53", pages 177-179, see entire document. *
Oncogene, Volume 4, issued 1989, H.J. XU et al., "The Retinoblastoma Susceptibility Gene Product: a Characteristic Pattern in Normal Cells and Abnormal Expression in Malignant Cells", pages 807-812, see entire article. *
Oncogene, Volume 4, Number 4, issued April 1989, A. T'ANG et al., "Genomic Organization of the Human Retinoblastoma Gene", pages 401-407, see entire article. *
Oncogene, Volume 6, Number 7, issued 1991, H.J. XU et al., "Lack of Nuclear RB Protein Staining in G0/Middle G1 Cells: Correlation to Changes in Total RB Protein Level", pages 1139-1146, see entire article. *
Proceedings of the National Academy of Science, USA, Volume 84, Number 14, issued July 1987, P. GUNNING et al., "A Human Beta-Actin Expression Vector System Directs High-Level Accumulation of Antisense Transcripts", pages 4831-4835, see entire article. *
Proceedings of the National Academy of Science, USA, Volume 84, Number 24, issued December 1987, S.H. FRIEND et al., "Deletions of a DNA Sequence in Retinoblastomas and Mesenchymal Tumors: Organization of the Sequence and its Encoded Protein", pages 9059-9063, see entire article. *
Proceedings of the National Academy of Science, USA, Volume 85, Number 16, issued August 1988, E.Y.H.P. LEE et al., "Molecular Mechanism of Retinoblastoma Gene Inactivation in Retinoblastoma Cell Line Y79", pages 6017-6021, see entire article. *
Proceedings of the National Academy of Science, USA, Volume 88, Number 12, issued 15 June 1991, R. TAKAHASHI et al., "The Retinoblastoma Gene Functions as a Growth and Tumor Suppressor in Human Bladder Carcinoma Cells", pages 5257-5261, see entire article. *
Science, Volume 235, issued 13 March 1987, W.H. LEE et al., "Human Retinoblastoma Susceptibility Gene: Cloning, Identification, and Sequence", pages 1394-1399, see entire article. *
Science, Volume 249, issued 14 September 1990, E.G. NABEL et al., "Site-Specific Gene Expression in Vivo by Direct Gene Transfer into the Arterial Wall", pages 1285-1288, see entire article. *
Science, Volume 254, issued 29 November 1991, R.J. CLEM et al., "Prevention of Apoptosis by a Baculovirus Gene During Infection of Insect Cells", pages 1388-1390, see entire article. *
See also references of EP0693874A4 *
The Journal of Experimental Medicine, Volume 169, Number 1, issued 01 January 1989, H. KARASUYAMA et al., "Autocrine Growth and Tumorigenicity of Interleukin 2-Dependent Helper T Cells Transfected with IL-2 Gene", pages 13-25, see entire article. *

Cited By (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
US5641484A (en) * 1990-12-04 1997-06-24 Board Of Regents, The University Of Texas System Methods for the suppression of neu mediated tumors by adenoviral E1A and SV40 large T antigen
US5643567A (en) * 1990-12-04 1997-07-01 Board Of Regents, The University Of Texas System Methods for the suppression of neu mediated tumors by adenoviral E1A and SV40 large T antigen
US5792751A (en) * 1992-04-13 1998-08-11 Baylor College Of Medicine Tranformation of cells associated with fluid spaces
WO1995016051A2 (en) * 1993-12-03 1995-06-15 Board Of Regents, The University Of Texas System Methods and compositions for the suppression of neu mediated transformation
WO1995016051A3 (en) * 1993-12-03 1995-12-28 Univ Texas Methods and compositions for the suppression of neu mediated transformation
US6331524B1 (en) 1994-12-09 2001-12-18 Genzyme Corporation Organ-specific targeting of cationic amphiphile / DNA complexes for gene therapy
US6383814B1 (en) 1994-12-09 2002-05-07 Genzyme Corporation Cationic amphiphiles for intracellular delivery of therapeutic molecules
US5939401A (en) * 1994-12-09 1999-08-17 Genzyme Corporation Cationic amphiphile compositions for intracellular delivery of therapeutic molecules
US5948767A (en) * 1994-12-09 1999-09-07 Genzyme Corporation Cationic amphiphile/DNA complexes
EP1006187A2 (en) * 1995-03-29 2000-06-07 Millennium Pharmaceuticals, Inc. Compositions for the diagnosis, prevention, and treatment of tumor progression
US6825320B1 (en) 1995-03-29 2004-11-30 Millenium Pharmaceuticals, Inc. FOHY03 polypeptides
US6794185B1 (en) 1995-03-29 2004-09-21 Millennium Pharmaceuticals, Inc. fohy030 nucleic acid molecules
EP1344825A1 (en) * 1995-03-29 2003-09-17 Millennium Pharmaceuticals, Inc. Fohy030 test kits
US6316204B1 (en) 1995-03-29 2001-11-13 Millennium Pharmaceuticals, Inc. Methods for detecting fohy030 polypeptide
EP1006187A3 (en) * 1995-03-29 2000-06-21 Millennium Pharmaceuticals, Inc. Compositions for the diagnosis, prevention, and treatment of tumor progression
US5869040A (en) * 1995-06-07 1999-02-09 Biogen, Inc Gene therapy methods and compositions
WO1997032991A3 (en) * 1996-03-06 1998-02-05 Avigen Inc Gene therapy for the treatment of solid tumors using recombinant adeno-associated virus vectors
US5952221A (en) * 1996-03-06 1999-09-14 Avigen, Inc. Adeno-associated virus vectors comprising a first and second nucleic acid sequence
US6218180B1 (en) 1996-03-06 2001-04-17 Avigen, Inc. Gene therapy for the treatment of solid tumors using recombinant adeno-associated virus vectors
US6531456B1 (en) 1996-03-06 2003-03-11 Avigen, Inc. Gene therapy for the treatment of solid tumors using recombinant adeno-associated virus vectors
WO1997032991A2 (en) * 1996-03-06 1997-09-12 Avigen, Inc. Gene therapy for the treatment of solid tumors using recombinant adeno-associated virus vectors
US6395712B1 (en) 1996-03-20 2002-05-28 Board Of Regents, The University Of Texas System Sensitization of HER-2/neu overexpressing cancer cells to chemotherapy
US7005424B2 (en) 1996-03-20 2006-02-28 Board Of Regents, The University Of Texas System Nucleic acid encoding an E1A gene product sensitizes HER-2/neu overexpressing cancer cells to chemotherapy
US6251597B1 (en) 1996-03-29 2001-06-26 Millennium Pharmaceuticals, Inc. Methods for detecting fohy030
US6312909B1 (en) 1996-03-29 2001-11-06 Millennium Pharmaceuticals, Inc. Compositions and methods for the diagnosis prevention and treatment of tumor progression
US6326356B1 (en) 1996-10-18 2001-12-04 Board Of Regents, The University Of Texas System Suppression of neu overexpression using a mini-E1A gene
US6197754B1 (en) 1996-10-18 2001-03-06 Board Of Regents, The University Of Texas System Suppression of tumor growth by a mini-E1A gene
US6683059B1 (en) 1996-10-18 2004-01-27 Board Of Regents, The University Of Texas System Mini-E1A gene and gene products
WO1998037185A3 (en) * 1997-02-20 1998-11-26 Univ Texas Vectors for controlled gene expression
WO1998037091A3 (en) * 1997-02-20 1998-11-05 Univ Texas Modified retinoblastoma tumor suppressor proteins
WO1998037185A2 (en) * 1997-02-20 1998-08-27 The Board Of Regents Of The University Of Texas System Vectors for controlled gene expression
WO1998037091A2 (en) * 1997-02-20 1998-08-27 Board Of Regents, The University Of Texas System Modified retinoblastoma tumor suppressor proteins
WO1999002190A1 (en) * 1997-07-11 1999-01-21 Genzyme Corporation Organ-specific targeting of cationic amphiphile/retinoblastoma encoding dna complexes for gene therapy

Also Published As

Publication number Publication date
AU6415494A (en) 1994-10-11
KR100326136B1 (en) 2002-11-23
KR960700752A (en) 1996-02-24
US5496731A (en) 1996-03-05
JPH08508166A (en) 1996-09-03
CA2159085A1 (en) 1994-09-29
US5912236A (en) 1999-06-15
IL109092A (en) 2005-09-25
TW379227B (en) 2000-01-11
EP0693874A1 (en) 1996-01-31
EP0693874A4 (en) 1997-06-25
AU697446B2 (en) 1998-10-08
JP3739787B2 (en) 2006-01-25
IL109092A0 (en) 1994-06-24

Similar Documents

Publication Publication Date Title
US5912236A (en) Broad-spectrum tumor suppressor genes gene products and methods for tumor suppressor gene therapy
US6468985B1 (en) Retinoblastoma protein-interacting zinc finger proteins
US7777005B2 (en) Compositions, kits, and methods relating to the human FEZ1 gene, a novel tumor suppressor gene
US5869040A (en) Gene therapy methods and compositions
US7033750B2 (en) Recombinant P53 adenovirus methods and compositions
JPH09509310A (en) Methods and compositions for inhibiting NEU-mediated transformation
JP2001502538A (en) Mini E1A gene and gene product
JP2000507815A (en) CFTR gene regulator
US6323335B1 (en) Retinoblastoma protein-interacting zinc finger proteins
AU736210B2 (en) Brca1 compositions and methods for the diagnosis and treatment of breast cancer
HUT74413A (en) A novel tumor suppressor gene
WO1998001460A9 (en) Brca1 compositions and methods for the diagnosis and treatment of breast cancer
JP2002503108A (en) Method for suppressing cancer cell growth using vector expressing pRb2 / p130
JP2001512325A (en) Modified retinoblastoma tumor suppressor protein
US20100317598A1 (en) Isolated BRCA1 Peptides and Method of Use
EP1297126A1 (en) Nucleotide and amino acid sequences of a cellular inhibitor of the dna replication in the epstein-barr herpes virus
MXPA99000406A (en) Brca1 compositions and methods for the diagnosis and treatment of breast cancer
HUT76558A (en) Prostate tumor suppressor gene located on human chromosome

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA CN CZ FI JP KR NO RU US

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE

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

Ref document number: 2159085

Country of ref document: CA

Ref document number: 1019950704162

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 1994911697

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1994911697

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1994911697

Country of ref document: EP