MXPA99007571A - Combined tumor suppressor gene therapy and chemotherapy in the treatment of neoplasms - Google Patents

Combined tumor suppressor gene therapy and chemotherapy in the treatment of neoplasms

Info

Publication number
MXPA99007571A
MXPA99007571A MXPA/A/1999/007571A MX9907571A MXPA99007571A MX PA99007571 A MXPA99007571 A MX PA99007571A MX 9907571 A MX9907571 A MX 9907571A MX PA99007571 A MXPA99007571 A MX PA99007571A
Authority
MX
Mexico
Prior art keywords
cells
protein
nucleic acid
tumor
tumor suppressor
Prior art date
Application number
MXPA/A/1999/007571A
Other languages
Spanish (es)
Inventor
Loretta Nielsen
Jo Ann Horowitz
Daniel C Maneval
G William Demers
Mary Ellen Rybak
Gene Resnick
Original Assignee
Canji Inc
G William Demers
Jo Ann Horowitz
Daniel C Maneval
Loretta Nielsen
Gene Resnick
Mary Ellen Rybak
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 Canji Inc, G William Demers, Jo Ann Horowitz, Daniel C Maneval, Loretta Nielsen, Gene Resnick, Mary Ellen Rybak filed Critical Canji Inc
Publication of MXPA99007571A publication Critical patent/MXPA99007571A/en

Links

Abstract

In one embodiment, this invention provides methods of treating mammalian cancer or hyperproliferative cells, said method comprising contacting said cells with a tumor suppressor protein or tumor suppressor nucleic acid and also contacting said cell with at least one adjunctive anti-cancer agent. The invention also provides for a pharmacological composition comprising a tumor suppressor protein or a tumor suppressor nucleic acid and at least one adjunctive anti-cancer agent, and a kit for the treatment of mammalian cancer or hyperproliferative cells.

Description

COMBINED THERAPY AND CHEMOTHERAPY OF THE TUMOR SUPPRESSOR GENE, IN THE TREATMENT OF NEOPLASMS FIELD OF THE INVENTION The present invention describes novel methods for treating subjects suffering from hyperproliferative diseases such as tumors or metastatic diseases. In particular, this invention provides methods for inhibiting the hyperproliferation of cells, more specifically neoplastic cells, comprising the combined use of a tumor suppressor gene or gene product with an added anticancer agent. BACKGROUND OF THE INVENTION Chromosomal abnormalities are often associated with genetic disorders, degenerative diseases and cancer. In particular, deletion or multiplication of copies of whole chromosomes or chromosomal segments, and higher-level amplifications of specific regions of the genome are common phenomena in cancer. See for example Smith (1991) Breast Cancer Res. Task t. , 18: Suppl. 1: 5-14; van de Víler (1991) Became. Beefiest. Minutes 1072: 33-50, Sato (1990) Cancer Res. , 50: 7184-7189. In fact, the amplification of the sequences of DNA containing proto-oncogenes and deletion of DNA sequences contain tumor suppressor genes, each of which is frequently characteristic of tumorigenesis. Dutrillaux (1990) Cancer Genet. Cytogenet 94: 203-217. Mutation of the p53 gene is the most common genetic alteration in human cancers (Bartek (1991) Oncogene 6: 1699-1703, Hollstein (1991) Science, 253: 49-53). In addition, the introduction of the wild-type p53 into cancer cells in mammals lacking the endogenous p53 wild-type protein suppresses the neoplastic phenotype of those cells (see for example US Pat. No. 5,532,220). Of the many available chemotherapeutics, paclitaxel, available on the market under the name of Taxol® (number of the NSC collection: 125973) has generated interest for its efficacy in clinical trials against refractory drug tumors, including tumors in glands ovarian and mammary (Hawkins (1992) Oncology, 6: 17-23, Hor itz (1992) Trends Pharmacol, Sci.13: 134-146, Rowinsky (1990) J. "Na tl.Can.Inst. 82: 1247- 1259) Recent studies on the interaction of paclitaxel and therapy with tumor suppressor genes show that reduced levels of a tumor suppressor (ie p53) correlated with an increased phase of G2 / M arrest micronucleation, as well as independent apoptosis, induced by paclitaxel. survivors with intact p53 progressed through a mitosis and transiently accumulated in the subsequent Gl phase, coincident with higher levels of p53 and p2? ciP1-wafl proteins (Wahl (1996) Nature Med. 2: 72-79 ). Similarly, Ha kins (1996) Canc. Res. 56: 892-898, showed that deactivation of p53 improved sensitivity to certain antimitotic agents including paclitaxel. The authors suggested that p53 may play a role in DNA repair, thus allowing cells to progress more easily through the S phase even in the presence of drugs. These studies suggest in this way that a therapy with tumor suppressor genes and drug therapy with antimitotic agents (especially a paclitaxel therapy) act for cross-effects purposes. SUMMARY OF THE INVENTION This invention provides methods for treating hyperproliferative cells in mammals. The invention is based in part on the surprising discovery that adjunctive anticancer agents in combination with a therapy with the tumor suppressor gene (e.g., p53) provide an enhanced effect in the inhibition of neoplastic cell proliferation and others that they have a deficient tumor suppressor activity. Thus, in one embodiment, this invention provides methods for treating cancer or hyperproliferative cells by contacting the cells with a tumor suppressor protein or tumor suppressor nucleic acid or at least one adjunctive anti-cancer agent. In some embodiments, methods include the combined administration of the protein or nucleic acid, tumor suppressor, and the adjunctive anti-cancer agent, with at least one chemotherapeutic agent. For example, a tumor suppressor nucleic acid (e.g., a nucleic acid encoding p53) can be administered with an adjunctive anti-cancer agent (e.g., paclitaxel) and a DNA-damaging agent such as cisplatin, carboplatin, navelbine (vinoralbine tartrate) ). Cancer or hyperproliferative cells are often neoplastic cells. When the cells are present in a tumor, the method inhibits the growth of the latter and thus provides a method to treat a cancer. These cancers include, but are not limited to, ovarian cancer, pancreatic cancer, non-small cell lung cancer, small cell lung cancer, hepatocarcinoma, melanoma, retinoblastoma, breast tumor, colo-rectal carcinoma, leukemia, lymphoma. , brain tumor, cervical carcinoma, sarcoma, prostate tumor, bladder tumor, reticuloendothelial tissue tumor, Wilm's tumor, astrocytoma, glioblastoma, neuroblastoma, ovarian carcinoma, osteosarcoma, kidney cancer and cancer in the head and neck. A preferred adjunctive anti-cancer agent is paclitaxel or a paclitaxel derivative, while a preferred tumor suppressor nucleic acid is a nucleic acid encoding a tumor suppressor protein selected from the group consisting of the p53 protein and its analogues as well as a retinal blastoma protein (RB). A preferred tumor suppressor nucleic acid in particular encodes a wild-type protein p53 and a particularly preferred retinoblastoma protein is a pllORB or a p56RB. The tumor suppressor nucleic acid is preferably delivered to a target cell by a vector. These vector viruses have been modified by recombinant DNA technology to allow expression of the tumor suppressor nucleic acid in the target cell. These vectors can be derived from vectors of non-viral origin (eg, plasmids) or viral (eg, adenovirus, adeno-associated virus, retrovirus, herpes virus, and vaccinia virus). In the preferred practice of the invention, the vector is a recombinantly modified adenoviral vector. The non-viral vectors are preferably subjected to complex formation with agents to facilitate the entry of DNA through the cell membrane. Examples of such Non-viral vector complexes include the formulation with polycationic agents that facilitate DNA condensation and lipid-based delivery systems. An example of a lipid-based delivery system would include delivery based on nucleic acid liposomes. Particularly suitable adenoviral vectors (e.g., delivery of a nucleic acid encoding a wild type p53 protein (comprising a partial or total deletion of a protein IX DNA.) In one embodiment, deletion of the protein IX gene sequence is extends from about 3500 bp from the terms 5 'to approximately 4000 bp from the viral terms 5' The vector may also comprise a deletion of a non-essential DNA sequence in the early region 3 of the adenovirus and / or in the early region of adenovirus 4 and in one embodiment the deletion is the sequence of DNA and Ela and / or Elb. A particularly preferred recombinant adenoviral vector for delivery of a p53 cDNA comprises the major late promoter of the adenovirus type 2 or the promoter of Human VMC, as well as the tripartite lido of adenovirus, cDNA A preferred adenoviral vector of this type is ACN53.The preferred paclitaxel or the paclitaxel derivatives preferred they include their own paclitaxel and / or Taxotere®, with paclitaxel (Taxol®) being preferred to a greater degree.
Another preferred adjunctive anti-cancer product is Epothilone. In a particularly preferred embodiment the A / C / N / 53 tumor suppressor and the adjunctive anticancer agent is paclitaxel. The tumor suppressor protein or the tumor suppressor nucleic acid can be dispersed in a pharmacologically acceptable excipient. Similarly, the adjunctive anti-cancer (e.g., paclitaxel or a paclitaxel derivative) can be dispersed in a pharmacologically acceptable excipient. The tumor suppressor protein or the nucleic acid with the same characteristics and paclitaxel or the paclitaxel derivative can both be dispersed in a single composition (comprising a multiple excipient or excipients). The tumor suppressor material (protein or nucleic acid) and / or the adjunctive anticancer can be administered intraarterially, intravenously (for example, injected), intraperitoneally and / or intratumorally, together or sequentially. Preferred sites of administration include the intrahepatic artery, the intraperitoneal route, or when it is convenient to treat the cells in the head (e.g., neurological cells) in the carotid system of the arteries. The tumor suppressor protein or nucleic acid can be administered in a single dose or in a multiplicity of treatments, for example, each one separated by at least 6 hours, with greater preference in at least three separate treatments in approximately 24 hours. In another preferred embodiment, the tumor suppressor protein or the tumor suppressor nucleic acid is administered (with or without an adjunctive anti-cancer agent) in a total dose ranging from 1 x 109 to about 1 x 1014 or about 1 x109 to about 7.5 x 1015, preferably of the order of 1 x 1011 to 7.5 x 1013 of adenovirus particles in a treatment regimen selected from the group consisting of: The total dose in a single dose, the total dose administered daily for 5 hours. days, the total dose administered daily for 15 days and the total dose administered daily for 30 days. The dose can also be administered continuously for 1 to 30 days. Paclitaxel or the paclitaxel derivative is administered in a total dose ranging from 75 to 350 mg / m2 for a period of 1 hour, 3 hours, 6 hours or 24 hours in a treatment regimen selected from the group consisting of: a single dose, in the form of a total dose administered daily in each day of day 1 and day 2, with the total dose administered daily during days 1, and 3, with a daily dosage for 15 days, with a daily dosage for 30 days, with a continuous infusion for 15 days, with a continuous infusion for 30 days. A preferred dose is 100 to 250 mg / m2 for 24 hours- Alternatively, paclitaxel or its derivative may be administered weekly in an amount of 60 mg / m2. This method of administration may be repeated for 2 or more cycles (more preferably for 3 cycles) and the two cycles or the largest number of cycles may be spaced for 3 or 4 weeks. In some preferred forms, a daily dose in the range of 7.5 x 109 to about 7.5 x 10 15, preferably of the order 1 x 10 12 to about 7.5 x 10 3 of adenovirus particles every day for up to 30 days can be administered daily. days (for example, a 2-day regimen or 2 to 5 days up to 14 days or 30 days with the same dose being administered each day.) Multiple regimens can be repeated in recurring cycles of 21 to 28 days. intra-arterial (for example, the intra-hepatic artery), the intratumoral and intra-peritonial route When the tumor suppressor nucleic acid (eg, p53) is administered in an adenoviral vector with an adjunctive anti-cancer agent (e.g. paclitaxel) and an agent that damages DNA (eg, cisplatin, carboplatin, or navelbine), the adenoviral acid vector is administered for 5 to 14 days at about 7.5 x 102 to about 7.5 x 1013 particles adenovirals per day. When the adenoviral vector and paclitaxel are administered with carboplatin, the dose is typically 7.5 x 1013 adenoviral particles per day. For example, a daily dose of the order of 7.5 x 10 12 adenoviral particles can be used for administration to the lung. The invention also provides kits for the treatment of breast cancer or hyperproliferative cells. These kits include a protein or a nucleic acid, tumor suppressor, as described herein (more preferably a protein or nucleic acid-wild-type, p53 (eg, in a viral or non-viral vector), or a protein or nucleic acid of retinoblastoma (RB), as well as an adjunctive anticancer agent described herein (for example, paclitaxel or a paclitaxel derivative) and / or optionally any of the other chemotherapeutic agents described herein.The kit, as another option may include further instructions describing the administration of either the tumor suppressor protein or nucleic acid and the adjunctive anti-cancer agent (and optionally another chemotherapeutic agent) to inhibit the growth or proliferation of the cancer or of the hyperproliferative cells. / C / N / 53 and paclitaxel In another embodiment this invention provides pharmacological compositions comprising a tumor suppressor protein or a tumor suppressor nucleic acid, as well as an adjunctive anticancer agent. In various embodiments, the pharmacological composition may optionally include any of the other chemotherapeutic compounds described herein. A particularly preferred composition includes a p53 nucleic acid (e.g., A / C / N / 53) and paclitaxel. The nucleic acid or protein, of the tumor suppressor type, and the myotherapeutic agent (eg, paclitaxel) may be present in different excipients or may be contained in a single excipient, as described herein. When there are multiple excipients, these excipients can be mixed together or stored separately (for example, in microcapsules). In yet another embodiment, this invention provides a composition comprising a breast cancer or a hyperproliferative cell in which such a cell contains a tumor suppressor nucleic acid of the exogenous type, or a tumor suppressor protein, of the same characteristic. The cell may additionally include an adjunctive anticancer agent such as paclitaxel or a paclitaxel derivative. The tumor suppressor nucleic acid or the tumor suppressor protein, of the exogenous type, can be any one or more of the nucleic acids and / or proteins, tumor suppressors described here. Similarly, the cell may be any one or more of the hyperproliferative and / or cancerous cells described herein. In yet another embodiment, this invention provides a method for treating a metastatic cell. The method involves contacting the cell with a tumor suppressor nucleic acid or with a tumor suppressor polypeptide. Suitable tumor suppressor nucleic acids and polypeptides include any of those tumor suppressor nucleic acids and / or polypeptides disclosed herein. The method may further include bringing the cell in contact with any of the adjunctive anticancer agents disclosed herein. In a particularly preferred embodiment, the method involves the topical administration of the nucleic acid and / or tumor suppressor polypeptide in a surgical wound. In another embodiment, this invention provides a particularly preferred dosage regimen. Thus, in one embodiment, this invention provides a method for treating mammary cells, which method involves administering to the cells a total dose of a tumor suppressor protein or of a tumor suppressor nucleic acid, this total dose being administered in a multiplicity of administrations of incremental doses of such protein or nucleic acid, tumor suppressor. The Preferred multiple administrations are separated from each other by an interval of at least 6 hours. A preferred administration consists of a minimum of three separate treatments for about 24 hours. In another embodiment, this invention provides a method for treating a mammary cell. The method involves administering to the cell a total dose of a protein or nucleic acid, tumor suppressor, the total dose being administered here in a multiplicity of administrations of incremental doses of protein or nucleic acid, tumor suppressor. Administrations can be spaced for a minimum of about six hours. The method may involve the administration of a minimum of three incremental doses and the doses may be administered daily. In one embodiment, the method can comprise at least three separate treatments within a period of at least 24 hours. In another embodiment, the method can involve the administration to the tumor of the tumor suppressor nucleic acid in a total dose of about 1 x 109 to 7.5 x 10 -15, or about 1 x 10 to 7.5 x adenovirus particles in a treatment regimen selected from the group consisting of the following: the total dose in a single administration, the total dose administered daily in a course of 5 days, the total dose administered daily for 15 days and the total dose administered every day for 30 days. The method may further comprise administering paclitaxel or a derivative thereof in a total dose ranging from about 75 mg / m2 to about 350 mg / m2 for a period of 24 hours within a treatment regimen selected from the group consisting of: the administration in a single dose, in a dose administered daily on days 1 and 2, in a dose administered daily on days 1, 2 and 3, a daily dosage for 15 days, a daily dosage for 30 days, a continuous infusion for 15 days. days or a daily continuous infusion for 30 days. These treatment regimens can be repeated for two or more cycles and these two or more cycles can be spaced for 3 or 4 weeks. The cells thus treated include the neoplastic cells comprising a cancer selected from the group consisting of ovarian cancer, mesiotelioma, pancreatic cancer, non-small cell lung cancer, small cell lung cancer, hepatocellular carcinoma, melanoma, retinoblastoma, breast tumor. , colorectal carcinoma, leukemia, lymphoma, brain tumor, cervical carcinoma, sarcoma, prostate tumor, bladder tumor, tumor in the reticuloendothelial tissues, ilm tumor, astrocytoma, gliblastoma, neuroblastoma, osteosarcoma, renal cancer and cancer of the head and neck. The results of the treatment turn preferably around the inhibition of the proliferation of a tumor as established by measuring the volume of the tumor. The invention also provides a pharmacological composition comprising a tumor suppressor protein or a tumor suppressor nucleic acid and at least one adjunctive anti-cancer agent. The adjunctive anti-cancer agent can be paclitaxel or a paclitaxel derivative. The tumor suppressor protein or nucleic acid can be selected from the group consisting of a nucleic acid encoding a wild type p53 protein, a nucleic acid encoding a retinoblastoma protein (RB), a p53 protein of type wild and a retinoblastoma protein (RB). The retinoblastoma protein can be pllORB or a p56RB. The nucleic acid may be contained in a recombinant adenoviral vector. The nucleic acid may be contained in a recombinant adenoviral vector comprising a partial or total deletion of a DNA IX protein and comprising a nucleic acid encoding a p53 protein. In one embodiment, the deletion of the protein IX gene sequence can range from about 3500 bp for the 5 'viral terms to about 4000 bp from the 5' viral terms. DNA deletion may include the sequence designated Ela and Elb. The recombinant adenoviral rector may also comprise the type of adenovirus 2 in the form of its promoter late major or the human CMV promoter, the tripartite leader cDNA adenovirus 2 type and a human p53 cDNA. In a preferred embodiment, the vector is A / C / N / 53. The composition may be paclitaxel, a paclitaxel derivative or a paclitaxel analog. The invention further provides a composition comprising a mammary or hyperproliferative cancer cell, said cell containing an exogenous type tumor suppressor nucleic acid or a tumor suppressor protein as well as an adjunctive anticancer agent. The tumor suppressor nucleic acid can be a nucleic acid encoding a tumor suppressor protein selected from the group consisting of the wild type protein p53 and a retinoblastoma protein (RB). The retinoblastoma protein may be pllORB, or p56RB. The cells may be present in a mammal. The cells can be neoplastic cells and the neoplastic cells can comprise a cancer selected from the group consisting of an ovarian cancer, pancreatic cancer, a non-small cell lung cancer, small cell lung cancer, hepatocarcinoma, melanoma, retinoblastoma, tumor of breast, colorectal carcinoma, leukemia, lymphoma, brain tumor, cervical carcinoma, sarcoma, prostate tumor, bladder tumor, tumor of reticuloendothelial tissues, Wilm's tumor, astrocytoma, glioblastoma, neuroblastoma, osteosarcoma, kidney cancer and head and neck cancer. The invention provides a method for treating a metastatic cell, said method comprising contacting the cell with a tumor suppressor nucleic acid or tumor suppressor polypeptides, as well as an adjunctive anti-cancer agent. The contacting may consist of the topical administration of a tumor suppressor nucleic acid to a surgical wound. The method may further include the administration with an adjuvant of a chemotherapeutic agent, and this chemotherapeutic agent may be cisplatin, carboplatin or navelbine. DEFINITIONS The term "adjunctive anticancer agent" refers to an agent that has at least any of the following activities: the ability to modulate a microtubule formation or action, the ability to inhibit the transferase activity of the polyprenyl protein, the ability to of inhibiting angiogenesis or the ability to inhibit endocrine activity. In more detail, the adjunctive anticancer agents useful in the invention are written below. As used herein the adjunctive anti-cancer agents according to the invention do not include compounds with an activity that damages DNA. The "tumor suppressor genes" are the nucleic acids for which the mutations with loss of function are oncogenic. Thus the absence, mutation or disruption of the normal expression of a tumor suppressor gene in an otherwise healthy cell increases the probability or the results of the cell reaching a neoplastic state. Conversely, when a gene or protein, functional and tumor suppressor type, is present in a cell, its presence suppresses the tumorigenicity, malignancy or hyperproliferative phenotype of the host cell. Examples of suppressive nucleic acids "of tumors within this definition, limitative character, the pllO, p56, p53, and other tumor suppressors described herein and in the currently pending application USSN 08 / 328,673, filed on October 25, 1994. Tumor suppressor nucleic acids include tumor suppressor genes or nucleic acids derived therefrom (e.g., cDNAs, cRNAs, mRNAs and subsequences thereof encoding respective active fragments of the tumor suppressor polypeptide), same as the vectors comprising these sequences A "polypeptide or protein, of the tumor suppressor type" refers to a polypeptide that, being present in a cell, reduces the tumorigenicity, malignancy or hyperproliferative phenotype of the cell. The term "viral particle" refers to an intact virion.
Infectious adenoviruses are typically determined by a spectrophotometric detection of DNA, as described by Huyghe (1995) Human Gene Ther. 6: 1403-1416. The term "neoplasia" or "neoplastic" is intended to describe a cell that grows and / or divides at a rate beyond the normal limits of growth for that cell type. The term "tumorigenic" or "tumorigenicity" is intended to mean having the ability to form tumors or the ability to cause the formation of tumors. The phrase "treating a cell" refers to the inhibition or improvement of one or several pathological features of a diseased cell. When used in reference to a cancer cell that is neoplastic (eg, a cancer cell in a mammal lacking an endogenous, wild-type tumor suppressor protein), the phrase "treating a cell" refers to mitigation or the elimination of the neoplastic phenotype. Typically such treatment results in the definition (a reduction or also the suspension of growth and / or proliferation) of the cell compared to the same cell under the same conditions except for the treatment (for example, an adjunctive anticancer agent and / or a tumor suppressor nucleic acid or polypeptide of this class).
Such inhibition may include death of the cell (e.g., apoptosis). These terms, when used with reference to a tumor, mean the inhibition of the proliferation growth of the tumor mass (for example, as it is measured volumetrically). This inhibition can be mediated by the reduction in the growth rate and / or in the rate of proliferation and / or in the death of the cells comprising the tumor mass. Inhibition of growth or inhibition of proliferation may be accompanied by an alteration in the cellular phenotype (for example, the restoration of the characteristic morphology of healthy cells, the restoration of contact inhibition, the loss of the invasive phenotype, the inhibition of anchored independent growth, etc.) - For the purposes of this text, a diseased cell will have one or several pathological features. These features in a diseased cell may include, among other things, a defective expression of one or more tumor suppressor proteins. The defective expression can be characterized by a complete loss of one or more functional tumor suppressor proteins or a reduction in the level of expression of one or more functional tumor suppressor proteins. Such cells are often neoplastic and / or tumorigenic. The term "systemic administration" refers to the administration of a composition or a drug, such as the recombinant adenoviral vectors of the invention or the anticancer or chemotherapeutic compounds, adjunctives, described herein, in such a way that the introduction of the composition or drug into the circulatory system is generated. The term "regional administration" refers to the administration of a composition or drug within a specific anatomical space, such as for example an intraperitoneal, intrathecal, subdural or some other specific organ and the like. Thus, for example, regional administration includes the administration of the composition or drug within the hepatic artery for regional administration to the liver. The term "local administration" refers to the administration of a composition or drug in a limited or circumscribed anatomical space, for example intratumoral injection into a mass of a tumor, subcutaneous injections, intramuscular injections and the like. Any person skilled in the art would understand that local administration or regional administration could also generate the entry of the composition or drug into the circulatory system. The term "reduced tumorigenicity" is used herein to refer to the conversion of hyperproliferative (eg, neoplastic) cells to a less proliferative condition. In the case of tumor cells the term "reduced tumorigenicity" is used to identify tumor cells that have become less tumorigenic or non-tumorigenic or cells lacking tumors whose ability to become tumor cells has been reduced or eliminated. Cells with reduced tumorigenicity do not form tumors in vivo or have a longer delay of weeks to months before the onset of tumor growth in vivo. Cells with reduced tumorigenicity can also result in slower growth of a three-dimensional tumor mass compared to the same type of cells that have a completely inactivated or non-functional tumor suppressor gene, which grows in the same physiological environment ( example a tissue, the age of an organism, the sex of an organism, the time within a menstrual cycle, etc.). As used herein, an "active fragment" of a gene or polypeptide includes a minor portion or smaller portions or smaller subsequences of the gene or nucleic acid derived therefrom (eg, cDNA) that has the ability to encode for proteins that possess a tumor suppressor activity. Similarly, an active fragment of a polypeptide refers to a subsequence of a polypeptide that possesses a tumor suppressor protein. An example of an active fragment is p56RB as described in the pending application of the United States of America USSN 07 / 328,673, filed on October 25, 1994.
The term "malignancy" is intended to describe a tumorigenic cell that has the ability to metastasize. The term "nucleic acids" as used herein may refer to DNA or RNA. Nucleic acids can also include modified nucleotides that allow a correct reading by a polymerase and that do not alter the expression of a polypeptide encoded by that nucleic acid. The phrase "nucleotide sequence" includes both sense and antisense cords either as individual cords or in duplex cords. The phrase "DNA sequence" refers to a single or double-stranded DNA molecule that consists of the nucleotide, adenosine, thymidine, cytosine and guanosine bases. The phrase "sequence of nucleic acids encoding ..." refers to a nucleic acid that directs the expression of a specific protein or peptide. Nucleic acid sequences include both the sequence of strands of DNA that is transcribed into RNA and the sequence of RNA that is translated into protein. Nucleic acid sequences include both full-length nucleic acid sequences and those that do not have their full length, derived from long sequences. full. It is further understood that the sequence includes the degenerative codons of the sequence or of the native sequences that can be introduced to provide a codon preference in a specific host cell. The phrase "expression cassette" refers to nucleotide sequences that are capable of affecting the expression of a structural gene in hosts compatible with such sequences. Such cassettes include at least the promoters and optionally also the transcription termination signals. Also, as we have described here, certain additional factors necessary or useful to effect the expression may be used. The term "operably linked" as used in this text refers to the link of a promoter upstream of a DNA sequence such that the promoter mediates the transcription of the DNA sequence. "Isolated" or "virtually pure", which term is used to refer to the nucleic acid sequences encoding the tumor suppressor protein or polypeptide or its fragments, refers to isolated nucleic acids that do not code for the protein or the peptides rather than the protein or polypeptide, tumor suppressor, or fragments thereof. The term "recombinant" refers to the DNA that it has been isolated from its native source or is endogenous and has been modified either chemically or enzymatically to suppress flanking nucleotides that occur naturally or that provide flanking nucleotides that do not occur naturally. The flanking nucleotides are those nucleotides that are found either upstream or downstream of the sequence or nucleotide subsequence, described. A "vector" comprises a nucleic acid that is capable of infecting, transfecting, translating transiently or permanently into a cell. It will be recognized that a vector can be a naked nucleic acid or also a nucleic acid complexed with a protein or a lipid. The vector optionally comprises nucleic acids and / or proteins, of viral or bacterial type, and / or membranes (for example, a cell membrane, a viral lipid envelope, etc.) of the same type. It is recognized that many times the vectors include an expression cassette which places the nucleic acid of interest under the control of a promoter. Vectors include, but are not limited to, replicons (eg, plasmids, bacteriophages) to which DNA fragments can bind to become replicated. Thus, vectors include, without limitation, RNAs, autonomous self-replicating circular DNAs (plasmids) and include both expression and expressionless plasmids.
When a microorganism or a recombinant-type cell culture is described as being host for an "expression vector" the term includes both extrachromosomal circular DNA and DNA that has been incorporated into the host chromosome or host chromosomes. When a vector is maintained by a host cell, the vector can be stably replicated by the cells during mitosis as an autonomous structure or incorporated into the host genome. The term "effective amount" is intended to identify that amount of the vector or drug that achieves a positive result in controlling the growth and / or proliferation of a cell. "U.C.I." is the translation of the abbreviation in English "C.I.U" and refers to the "infectious cell units". The U.I.C. it is calculated by measuring the positive cells in the viral hexon protein (e.g., 293 cells) after an infection period of 48 hours. (Huyghe (1995) Human Gene Ther 6: 1403-1416). The abbreviation "m.d.i." as used herein it refers to "multiplicity of infection" and is equivalent to the U.I.C. per cell. The term "paclitaxel" as used in this text refers to the drug that is known in a market as Taxol®. Taxol® inhibits the replication of cells Eukaryotic cells improving the polymerization of the tubulin portions in stabilized microtubule packages that are unable to reorganize themselves into the structures appropriate for mitosis. The term "contacting a cell" or "contacting a cell" when referring to contacting a drug and / or a nucleic acid is used in this text to refer to the contacting in such a way that the drug and / or the nucleic acid are internalized in the cell. In this context, contacting a cell with a nucleic acid is equivalent to transferring a cell to a nucleic acid. When the drug is lipophilic or the nucleic acid forms complex with a lipid (for example, a cationic lipid) the simple contact will result in a transport (active, passive and / or diffusive) to the interior of the cell. Alternatively, the drug and / or nucleic acid can be actively transported into the cell, as such or in combination with a carrier composition. Thus, for example when the nucleic acid is present in an infective vector (eg, an adenovirus) the vector can mediate the absorption of the nucleic acid in the cell. The nucleic acid can be complexed to form agents that specifically interact with extracellular receptors to facilitate delivery of the nucleic acid to the cell. The examples include the ligation / polycation / DNA complexes as described in U.S. Patent Nos. 5, 166,320 and 5,635,383. In addition, viral delivery can be improved by a recombinant modification of the button fields or viral genome fibers to incorporate those portions pointing towards the cell. The constructions designated herein, "A / C / N / 53", pllORB, p56RB, refer to the constructions so designated in the pending application of the United States serial number USSN 08 / 328,673, filed on October 25, 1994 , International Application WO 95/11984. A "conservative substitution" when a protein is described, refers to a change in the amino acid composition of the protein that basically does not alter the activity of the protein. Thus "conservatively modified variations" of a particular amino acid sequence refers to substitutions of those amino acids that are not critical for protein activity or the substitution of amino acids by others that possess similar properties (eg, acid, basic, charged) positively or negatively, polar or non-polar, etc.) in such a way that the substitution is from amino acids until critical basically does not alter the activity. Conservative substitution tables that provide functionally similar amino acids are well known in the art.
For example, the following six groups each contain amino acids that constitute conservative substitutions for others: 1) Alanine (A), Serine (S), Threonine (T), - 2) Aspartic acid (D), Glucanic acid (E); 3) Asparagine (N), Glutamine (Q); 4) Arginine (R), Lysine (K); 5) Isoleucine (I), Leucine (L), Methionine (M), Valine (V), - and 6) Phenylalanine (F), Tyrosine (Y), Tripofan (W). See also Creighton (1984) Proteins W.H. Freeman and Company. In addition, substitutions, suspensions or individual additions that alter, increase or suspend a single amino acid or a small percentage of amino acids in an encoded sequence also constitute "conservatively modified variants". BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 illustrates the inhibition in vitro of ovarian SK-OV-3 tumor cells by different concentrations of p53 (A / C / N / 53) and / or Taxol®. Figure 2 provides an isobologram analysis for the experiments illustrated in Figure 1.
A synergism was observed between Taxol® and p53 (A / C / N / 53) when the cells had been previously treated with Taxol® 24 hours before treatment with p53.
Figures 3a, 3b and 3c illustrate the efficacy of p53 Ad against xeno-injections of human breast cancer in nude mice. The mice were given a total dose of 2.2 x 109 U.I.C. of adenovirus (A / C / N / 53 or Ad) per mouse, divided into 10 injections for days 0-4 and 7-11. The mice were treated with p53 Ad, beta-gal Ad or only the vehicle. Figure 3a illustrates the results with the MDA-MB-231 tumors. Figure 3b illustrates the results with tumors MDA-MB-468 (-468) and Figure 3c illustrates the results with the tumors MDA-MB-435 (-435). Figures 4a and 4b provide the dose response curves with p53 Ad (A / C / N / 53) for tumors MDA-MB-231 (-231) (Figure 4a) and for tumors MDA-MB-468 ( Figure 4b). Mice were dosed 1 x 107-1 x 109 U.I.C. of p53 Ad (A / C / N / 53), quantity divided into 10 doses administered peritumorally on days 0-4 and 7-11. Percent inhibitions were calculated, on average, by comparing tumor volumes with each dose of Ad p53 with tumors treated with buffers on days 14/15, 18 21, 24, 28, 30/32 and 35 (MDA-tumors). MB-468 only on day 35). Tumors -231 showed an average of 22.5 ± 1.2 mm3 at day 0 while tumors -468 showed an average of 33.1 ± 1.8 mm3 at day 0. Figure 5 provides a comparison of the efficacy of the therapeutic agent when administered as a alone bolus or as divided doses. Tumors / MDA-MB-231) were dosed with a total of 2.2 x 108 U.I.C. of p53 Ad per week, given during weeks 1 and 3. Figure 6 illustrates the efficacy of multiple cycles of a low dose of Ad p53 against large, well established tumors. A total of 1.32 x 109 U.I.C. from p53 Ad for 6 weeks to xenografts of MDA-MB-468 (P = platform in the growth rate of the control tumor; E = end of dosing). Figures 7a, 7b and 7c illustrate the inhibition in vi vo of MDA-MB-468 tumors in nude mice who administered 1 x 109 U.I.C. of p53 Ad (A / C / N / 53) as an injection in a single bolus (Figure 7a) or divided into three injections (Figure 7b) or 5 injections (Figure 7c). Figure 8 illustrates the ability of dexamethasone in low dose to suppress the inhibition of tumor growth mediated by? K cells in "scid" type mice. The tumors MDA-MB-231 were dosed with a total of 2 x 109 U.I.C. of beta-gal Ad (1.1 x 1011 viral particles) divided into 10 injections given on days 14-18 and 21-25. The subcutaneous granules of dexamethasone (or, if applicable, placebo) released 83.3 μg of steroid per day. Figure 9. Comparison of a combination of p53 and cisplatin therapy in normal and tumor cells. DETAILED DESCRIPTION This invention provides new methods for inhibiting the growth and / or proliferation of cells, more particularly the growth and proliferation of cancer cells. In one embodiment, the methods involve placing the cells in contact with a tumor suppressor nucleic acid or a tumor suppressor protein and with an adjunctive anti-cancer agent. Typically the protein or nucleic acid, tumor suppressor, used here, will be of the same species as that tumor suppressor protein that is lacking. Thus, when the cells lack an endogenous p53 activity, a p53 protein or a p53 nucleic acid will be used. It was a surprising discovery of the invention that, contrary to the results described in previous studies (see for example Wahl et al. (1996) Nature Med., 2 (1): 72-79, and Hawkinss et al. (1996) Canc. Res. 56: 892-898), the treatment of cells in mammals lacking or deficient in endogenous and wild-type tumor suppressor protein (ie many neoplastic cells) both with an adjunctive anti-cancer agent ( for example paclitaxel (Taxol®)) and a tumor suppressor gene or polypeptide (e.g., p53) results in the inhibition of cell proliferation and / or growth to a greater degree than that observed with either chemical or with tumor suppressor construction alone. He has also It has been a discovery of this invention that pretreatment with adjunctive anticancer agents dramatically increases the antiproliferative effect of a tumor suppressor nucleic acid. Without being bound by any particular theory, we believe that a possible means by which an adjunctive anticancer agent can contribute to this enhanced effect is that of increasing the transfection efficiency of different gene therapy vectors (e.g., adenovirus vectors); or increase the expression levels of the tumor suppressor gene; either stabilize the microtubules to aid in the transport of the intracellular viruses, or also to provide the improved effect thanks to the interaction of different intracellular mechanisms (for example, signaling paths, apoptic paths and paths that allow cycling of the cells Thus in one embodiment this invention provides methods for inhibiting diseased cells in mammals lacking a tumor suppressor protein of wild type and endogenous cells, or lacking such a protein, by bringing such cells into contact with an agent adjunctive anticancer and with a tumor suppressor nucleic acid and / or with a polypeptide of the same type When cells are present in a tumor, the method inhibits the growth of a tumor and thus offers a method for treating the tumor.
Cancer. Particularly preferred nucleic acid or tumor suppressor polypeptides include p53, RB, h-NUC (see for example, Chen (1995) Cell Growth Differ., 6: 199-210) or their active fragments (e.g., pllORB , p56RB), while particularly preferred anticancer agents (compounds) include paclitaxel and compounds with paclitaxel-like activity such as paclitaxel derivatives (eg, analogs). It has also been a discovery of the present invention that the contacting of cells with a nucleic acid and / or a tumor suppressor peptide can inhibit metastatic cells. This inhibition can take the form of inhibiting the formation, growth, migration or reproduction of metastatic cells. In one embodiment, inhibition by inhibition (i.e., reduction and / or elimination) in the appearance of remote neoplasms of the primary tumor may be characterized. Thus, this invention provides methods for treating (mitigate or eliminate) the progress of a metastatic disease. The methods involve contacting the metastatic cells with a nucleic acid and / or polypeptide, tumor suppressor. In a particularly preferred embodiment, this method may involve contacting the cells at a site where a surgical wound is located (after removal or decrease the size of a tumor mass) with a tumor suppressor nucleic acid and / or a tumor suppressor polypeptide in combination with an adjunctive anticancer agent. The cells can be contacted additionally with an adjunctive anticancer agent as described in that text. In yet another embodiment, this invention provides advantageous treatment regimens in which tumor suppressor genes and gene products are used. In part these treatment regimes are based on the surprising discovery that nucleic acids and / or polypeptides, of the tumor suppressor type, are more effective in inhibiting a cell or tumor growth when the material is delivered rather in administrations. multiple and not so much in a single bolus. The order in which tumor suppressor agents and adjunctive anticancer agents are administered is not critical to the invention. Thus it is possible to administer the composition or the compositions in a simultaneous or sequential manner. For example, in one embodiment, the pretreatment of a cell with at least one adjunctive anti-cancer agent (only in combination with a chemotherapeutic agent) increases the efficacy of a tumor suppressor and nucleic acid, subsequently administered. In one embodiment, it is administered the chemotherapeutic agent before the adjunctive anticancer agent and the tumor suppressor and / or nucleic acid and / or polypeptide. In another embodiment, the adjunctive anti-cancer agent (alone or in combination with a chemotherapeutic agent) is administered simultaneously with the tumor suppressor acid and / or polypeptide. In another embodiment, the adjunctive anti-cancer agent is administered after administering the tumor suppressor and / or polypeptide agent.
The antitumor effect of administering the composition and methods of the present invention also includes a non-specific, anti-tumor effect, called "observer effect", (see for example, Zhan (1996) Methastasis Cancer Rev. 15: 385-401 and Okada (1996) Gene Ther 3: 957-96). In addition, the immune system can also be manipulated to selectively (or repress) the humoral arm or cell of the immune system, i.e., modulate the B cell and / or the T cell (e.g. a cytotoxic lymphocyte (CTL international abbreviation) or a response of infiltrating tumor lymphocyte (international abbreviation TIL)). Thus, for example, an increase in TIL responses is observed when an adenovirus expressing p-53 is administered to humans. Specifically, an increase in TIL is observed (phenotypically the T: CD3 + and CD4 + helper cells) when administered by the intra-hepatic arterial route an idenovirus expressing p-53 for the treatment of a metastatic hepatic carcinoma, as described in more detail below. It is recognized that the methods of this invention are not restricted to the use of a single adjunctive anti-cancer agent or even the use of a single individual chemotherapeutic. Thus, this invention provides methods for inhibiting diseased cells in mammals lacking an endogenous tumor suppressor protein, or a tumor comprising such cells, by contacting the cells or the tumor with a tumor suppressor nucleic acid and one or several adjunctive anticancer agents, as described herein. I. Adjuvant Anticancer Agents A) Agents Affecting Microtubules As explained above, in one embodiment this invention provides methods for inhibiting diseased cells lacking an endogenous tumor suppressor protein by contacting the cells with a tumor suppressor protein or tumor suppressor nucleic acid and an adjunctive anticancer agent, such as an agent that affects microtubules (e.g. paclitaxel, a derivative of paclitaxel or a compound similar to paclitaxel). As used in this text, an agent that affects microtubules is a compound that interferes with cellular mitosis, that is, it has an antimitotic effect, by affecting the formation and / or action of a microtubule. Such agents can be, for example, microtubule stabilizing agents or agents that interrupt the formation thereof. Agents affecting the microtubules useful in the present invention are well known to those skilled in the art and include, but are not limited to, allocolchicine (NSC 406042), Halicondrin B (609395), colchicine (NSC 757), derivatives of colchicine (eg, NSC 33410), dolastatin 10 (NSC376128), maytansine (NSC 153858), rhizoxin (NSC 332598), paclitaxel (Taxol '", NSC 125973), Taxol® derivatives (eg, NSC 608832), thiocolchicine (NSC 361792), trityl -cysteine (NSC 83265), vinblastine sulfate (NSC 49842), vincristine sulfate (NSC 361792), epothilone A, epothilone, and discodermolide (see Service, (1996) Sci ence, 274: 2009) estramustine, nocodazole, MAP4, and the like. Examples of such agents are also described in the scientific and patent literature, see for example, Bulinski (1997) "Cell. Sci. 110: 3055-3064; Panda (1997) Proc. Na tl. Acad. Sci. USA 94: 10560-10564; Muhltradt (1997) Cancer Res. 57: 334-3346; Nicolaou (1997) Nature 387: 268-272; Vasquez (1997) Mol. Biol. Cell. 8: 973-985; Panda (1996) J. Bi ol. Chem. 271: 29807-29812. Particularly preferred agents are compounds with an activity similar to that of paclitaxel. They include, without limitation, paclitaxel and paclitaxel derivatives (paclitaxel-like compounds) and the like. Paclitaxel and its derivatives are available in the market. In addition, methods for making paclitaxel, paclitaxel derivatives and their analogues are well known among experts. (see, for example, the US Patent Numbers ,569,729; 5,565,478; 5,530,020; 5,527,924; 5,508,447; 5,489,589; 5,488,116; 5,484,809; 5,478,854; 5,478,736; 5,475,120; 5,468,769; 5,461,169; 5,440,057; 5,422,364; 5,411,984; 5,405,972; and 5,296,506). Additional agents that affect microtubules can be evaluated with the use of one of many assays known in the art, such as a semi-automated assay that measures the tubulin polymerizing activity of paclitaxel analogs in combination with a cellular assay intended to measure the potential of these compounds to block cells in mitosis (see Lopes (1997) Cancer Chemother, Pharmacol., 41: 37-47).
In general terms, the activity of a test compound is determined by contacting a cell with that compound and then determining if the cell cycle is interrupted or not, in particular due to the inhibition of a mitotic event. Such inhibition can be mediated by interruption of the mitotic apparatus, for example the interruption of the formation, of normal axis. Cells in which mitosis is interrupted can be characterized by altered morphology (eg, microtubule compaction, increased number of microsomes, etc.). In a preferred embodiment, the compounds are filtered in vi tro with a possible tubulin polymerizing activity. In a preferred embodiment, the compounds are filtered against cultured WR21 cells (derived from line 69-2 of "ap-ras" type rats) for the inhibition of proliferation and / or for detecting an altered cell morphology, in particular in terms of a microtubule compaction. The filtration in vi vo of the positive test compounds can then be performed with the use of nude mice carrying the WR21 tumor cells. Porter (1995) Lab. Anim. Sci. , 45 (2): 145-150, describes the detailed protocols for this filtration method. Other methods of filtering or classifying Compounds for determining a desired activity are well known to those skilled in the art. Typically, such tests involve those tests designed to fix the inhibition of assembly and / or disassembly of the microtubules. The assays for the assembly of the microtubules are described, for example, in: Gaskin et al. (1974) J. Mol ec. Biol. , 89: 737-758. Also US Pat. No. 5569,720 provides in vi tro and in vi ve assays for compounds with an activity similar to that of paclitaxel. B) Polyprenyl protein transferase inhibitors In some other form of embodiments of the invention provides the combined use of nucleic acids and / or polypeptides, tumor suppressors, and polyprenyl-protein transferase inhibitors. Particularly preferred polyprenyl protein transferase inhibitors include, but are not limited to, the farnesyl protein transferase inhibitors (FPT international abbreviation), the transferase inhibitors of geranylgeranyl-protein and other monoterpene protein transierases. Examples of compounds that are polyprenyl protein transferase inhibitors are well known in the scientific and patent literature, see for example, Zhang (1997) "Biol. Chem., 272: 10232-10239; Nj oroge (1997)" 7. Med. Chem., 40: 4290-4301; Mallams (1997) Bioorg, Med. Chem., 5: 93-99. Exemplary which are farnesyl protein transferase inhibitors are presented below: The FPT inhibitor, identified as "FPT39", as described in International Application WO 97/23478, filed December 19, 1996, wherein the FPT39 is the compound designated "39.0", see page 95 of International Application WO 97/23478.
FPT39 Compound: As described below, when FPT39 is used in a combination therapy with the adenovirus expressing p53 of the invention against prostate tumor cells and mammary tumor cells, the combination was more effective in killing the tumor cells than the effect achieved with either the agents alone. The oligopeptides (in most cases the tetrapeptides, but also the pentapeptides which includes the formula Cys-Xaal-Xaa2 -Xaa3: EPA 461.489; EPA 520.823; Peptide-mimetic compounds, especially the mimetics of Cys-Xaa-Xaa-Xaa: and EPA memories 535,730, EPA 618,221; WO 94/09766; WO 97/10138; WO 97/07966; US 5,326,773, US 5,420,245; WO 96/20396; US ,439,918; as well as International Memory WO 95/20396. The farnesylated pepti-mimetic compounds, specifically the farnesylated mimetic Cys-Xaa-Xaa-Xaa: the Great Britain Patent GB-A2.276, 618. Other mimetic peptide compounds: the Memoirs: US 5,352,705, WO 94/00419; WO 97/00497; WO 95/09000, WO 95/09001; WO 91/12612; WO 95/25086; EPA 675,112, and FR-A 2, 718, 149. Tricyclic ring benzocycloheptapyridines: WO 95/10514; WO 95/10515; WO 95/10516; WO 96/30363; WO 96/30018; WO 96/30017; WO 96/30362; WO 96/31111; WO 96/31478; WO 96/31477; WO 96/31505; International Patent Application number PCT / US96 / 19603, WO 97/23478; US Patent Application Serial No. 08 / 728,104; US Patent Application Serial No. 08 / 712,989; US Patent Application Serial No. 08 / 713,326; US Patent Application Serial No. 08 / 713,908; US Patent Application Serial No. 08 / 713,705; US Patent Application number of series 08 / 713,703; US Patent Application Serial No. 08 / 710,225; US Patent Application Serial No. 08 / 711,925; US Patent Application Serial No. 08 / 712,924; US Patent Application Serial No. 08 / 713,323; and U.S. Patent Application Serial No. 08 / 713,297. The farnesyl derivatives: EPA 534,546; WO 94/19357; WO 95/08546, EPA 537,007; and WO 95/13059. Natural products and their derivatives: WO 94/18157; US 5,430,055; GB-A 2,261,373, GB-A 2,261,374, GB-A 2,261,375; US 5,420,334, US 5,436,263. Other components: WO 94/26723; WO 95/08542; US 5,420,157; WO 95/21815; and WO 96/31501. C) Anti-angiogenic components The proteins or nucleic acids, tumor suppressors, according to this invention can also be administered in combination with anti-angiogenic compounds. Preferred antiangiogenic compositions inhibit the formation of blood vessel proliferation, more preferably the formation and / or proliferation of blood vessels to tumors. Suitable anti-angiogenic compositions include, but are not limited to, Galardin (GM6001, Glycomed, Inc., Alameda, CA), as inhibitors of endothelial response (eg agents such as interferon-alpha, TNP-470, and vascular endothelial growth factor inhibitors), agents that induce cell matrix disintegration (eg, Vitaxin (human antibody LM-609 , from Ixsys Co., San Diego, CA, Metastat, CollaGenex, Newtown, PA, and Marimastat BB2516, from British Biotech), as well as agents that act directly on vessel growth (eg, CM- 101, which is derived from pxotoxin from antigen to Group A streptococcus and which binds to new blood vessels to induce an intense inflammatory response in the host, as well as Thalidomide.) Several classes of steroids have also been reported as substances that exert an anti-angiogenic activity- In particular, several reports have indicated that medroxyprogesterone acetate (MPA), a synthetic progesterone, has vigorously inhibited neovascularization in the assay or on the cornea in the rabbit (Oikawa (1988) Cancer Let t. , 43:85). This is a probiofarm of 5FU, 5'-deoxy-5-fluorouridine (5'DFUR), it can also be characterized as an antiangiogenic compound, since 5'DFUR is converted to 5-FU by thymidine phosphorylase activity of PD-ECGF / TP. The 5 'DFUR can be selectively active for detect tumor cells positive to PD-ECGF / TP with an autopotential of angiogenesis. Recent clinical investigations have shown that 5 'DFUR is probably effective for tumors positive for PD-ECGF / TP. It was shown that there was dramatic improvement in the antitumor effect of 5 'DFUR in those cells transfected with PD-ECGF-TP as compared to wild-type, non-transfected cells (Haraguchi (1993) Cancer Res., 53: 5680- 5682). In addition, the combined 5'-DFUR + MPA compounds are also effective antiangiogenic agents (Yayoi (1994) In t J Oncol -, 5: 27-32). The combination of 5 'DFUR + MPA could be categorized as a combination of two angiogenesis inhibitors with different spectra, an inhibitor of endothelial growth factor and a protease inhibitor. In addition, the experiments carried out in vi vo with the use of mammary carcinomas in the rat induced by DMBA, that is, the 5 'DFUR exhibited a combined effect with AGM-1470 (Yamamoto (1995) Oncol. Reports, 2: 793-796 ). Another group of anti-angiogenic compounds for use in this invention include polysaccharides capable of interfering with the function of growth factors with ligation to heparin, which promote angiogenesis (e.g. pentosan polysulfate).
Other modulators of angiogenesis include platelet factor IV and AGM-1470. Still others are derivatives of the collagenase inhibitor from natural sources such as vitamin D3 analogues, fumigaline, herbimycin A, and isoflavones. D) Endocrine therapy Endocrine therapy, which has already been established as well as a representative cytostatic treatment, can cause the cells to settle down depending on the hormones and which can reduce the number of tumor cells in vi, and is also able to inhibit the growth of tumors in patients with hormone-dependent tumors. It is expected that such therapies will increase the effect of tumor suppressors on the treatment of hypoproliferative cells. Thus, in another embodiment, this invention provides for example the combined use of a nucleic acid and / or polypeptide, tumor suppressor, with an anti-estrogen, anti-androgen, or anti-progesterone. Endocrine therapeutics are well known to those skilled in the art and include, but are not limited to, tamoxifen, toremifene (see, for example, US Pat. No. 4,696,949), flutamide, megace and lupron, see also, for example, Memoirs WO 91 / 00732, WO 93/10741, WO 96/26201, and Gauthier et al. (1997) J. Med.
Chem. , 40: 2117-2122). E) Supplies of Adjunct Anticancer Agents: Pharmaceutical Compositions Pharmaceutical Compositions The adjunctive anticancer agents used in the methods of the invention are typically combined with a pharmaceutically acceptable carrier (excipient) to form a pharmacological composition. The pharmaceutical composition of the invention may comprise one or more of the anticancer agents adjunct with a tumor suppressor gene or polypeptide, for example, p53 or RB, or without them. The pharmaceutically acceptable carriers can contain a physiologically acceptable compound which acts, for example, to stabilize the composition or increase or decrease the absorption of the agent and / or pharmaceutical composition. Physiologically acceptable compounds may include for example carbohydrates, such as glucose, sucrose or dextrans, antioxidants, ascorbic acid or glutathione, chelating agents, low molecular weight proteins, compositions that reduce the release or hydrolysis of adjunctive anticancer agents or excipients or others. stabilizers and / or tampons. Also detergents can be used to stabilize the composition or increase or decrease the absorption of the pharmaceutical composition (see below, for exemplary detergents). Other physiologically acceptable compounds include wetting agents, emulsifying agents, dispersing agents or preservation agents that are particularly useful for preventing the growth or action of microorganisms. Various preservation agents are well known and they include, for example, phenol and ascorbic acid. One skilled in the art would appreciate that the choice of a pharmaceutically acceptable carrier, including a physiologically acceptable compound, depends for example on the route of administration of the adjunctive anticancer agent and on the physiochemical characteristics of the adjunctive anti-cancer agent in particular. The compositions for administration will commonly comprise a solution of the adjunctive anticancer agent dissolved in a pharmaceutically acceptable carrier, preferably an aqueous carrier for adjuvant, water soluble anticancer agents. A variety of carriers can be used, for example, buffered salt water and the like. These solutions are sterile and are generally free of unwanted matter. These compositions can be sterilized by sterilization techniques conventional, well-known. The compositions may contain pharmaceutically acceptable excipients, as required to approximate physiological conditions such as pH adjusting agents and buffers, toxicity adjusting agents and the like, for example sodium acetate, sodium chloride, potassium chloride, chloride of calcium, sodium lactate and the like. The concentration of adjunctive cancer agent in these formulations can vary widely and will be selected primarily on the basis of fluid volumes, viscosities, body weight and the like in accordance with the specific mode of administration selected and the needs of the patient. Rutes of Sumini stru * The adjunctive anticancer agents used in the methods of the invention are useful and can be delivered alone or as pharmaceutical compositions (with a tumor suppressor, for example p53, or without it) by any means known in the art. technical, for example, systemic, regional, or local; by intraarterial, intratumoral, intravenous (IV), parenteral administration, in the intrapleural cavity, by topical, oral, or local route, as subcutaneous, intratracheal material (for example, by aerosol) or by the transmucosal route (by example, buccal, bladder, vaginal, uterine, rectal or nasal mucosa), and also intratumoral (that is, with transdermal application or local injection). Particularly preferred modes of administration include intra-arterial injections, especially when it is desirable to achieve a "regional effect", for example, by focusing on a specific organ (e.g., brain, liver, spleen, lungs). For example, injection into the intrahepatic artery is preferred when the so-called regional antitumor effect in the liver is desired; or also the injection into the intra-carotid artery, when it is desired to supply a composition to the brain (for example, for the treatment of brain tumors), a carotid artery or an artery of the carotid system of the arteries (for example the occipital artery, the auricular artery, the temporal artery, the cerebral artery, the maxillary artery, etc.). Paclitaxel and certain paclitaxel derivatives are only marginally soluble in aqueous solutions. In a preferred embodiment, these compositions are delivered directly to the tumor site (e.g., by injection, channelization, or direct application during a surgical procedure) or are solubilized in an acceptable excipient. The methods for administering paclitaxel and its derivatives are well known among the branch experts (see for example U.S. Patent Nos. 5,583,153, 5,565,478, 5,496,804, 45,484,809.) Other derivatives of paclitaxel are water soluble analogs and / or pro-drugs (see U.S. Patent Nos. 5,411,984 and 5,422,364) and are conveniently administered by any of a variety of different methods described above The pharmaceutical compositions according to this invention are particularly useful for topical administration, for example in surgical wounds to treat incipient tumors, neoplastic and metastatic cells and their precursors. Compositions for parenteral administration, such as intravenous administration or that which enters the body cavity or the lumen of an organ, are useful. Trame regimens The pharmaceutical compositions can be administered in a variety of unit dosage forms. agreement with e The method of administration. For example, unit dosage forms suitable for oral administration include powders, tablets, pills, capsules, and lozenges. It is recognized that adjunctive anti-cancer compounds (for example paclitaxel and related compounds described), when administered orally should be protected from digestion. This is typically accomplished either by forming a complex of the adjunctive anticancer agent with a composition to render it resistant to acid and enzymatic hydrolysis or by packaging the adjunctive anti-cancer agent in an effectively resistant carrier., as a liposome. Means for protecting digestion compounds are well known in the art (see for example U.S. Patent 5,391,377 which discloses lipid compositions for oral delivery of therapeutic agents). Doses for typical chemotherapeutics are well known among branch experts. Furthermore, it can be said that such dosages are typically of a recommendation nature and they can be adjusted according to the particular therapeutic context, patient tolerance, and so on. Thus, for example, a typical dosage of pharmaceutical composition (for example paclitaxel) for intravenous (IV) administration would be of the order of 135 mg / m2 administered for a period of 1 to 24 hours (typically at 1, 3 or 6, and more preferably every 3 hours) and more preferably, it is repeated every three weeks for 3 to 6 cycles. To reduce the frequency and severity of hypersensitivity reactions, patients can also receive about 20 mg of dexamethasone (Decadron, and others) orally for about 12 hours and 6 hours before, and about 50 mg of diphenhydramine (Benadryl, and others) plus about 300 mg of cimetidine (Tagamet ") or 50 mg of rantidma (Zantac), and IV 30 to 60 minutes before paclitaxel treatment, considerably higher doses (eg, up to about 350 mg / m2 per day) can be used particularly when administering the drug to a separate site and not in the bloodstream, as for example in a body cavity or in the lumen of an organ.Similarly higher doses are possible by any selected route, for example topical administration.Effective methods for preparing parenterally administrable compositions are known or obvious for those skilled in the art and are described in greater detail in publications such as Remington's Pharmaceutical Science, 15th ed., Mack Publishing Company, Easton, Pennsylvani a (1980) and in the North American Patents numbers: 5,583,153, 5,565,478, 5,496,804, and 5,484,809. Typical doses, for example for intraperitoneal administration, will be from 20 to 150 mg / m2 each week, or around 250 mg / m2 every 3 weeks. The compositions containing the adjunctive anticancer agents can be administered for therapeutic treatments. In the applications The compositions are administered to a patient suffering from a disease characterized by the hypoproliferation of one or more cell types in an amount sufficient to cure or at least partially arrest the disease and / or its complications. An amount adequate to achieve the above is defined as a "therapeutically effective dose". The effective amounts for this use will depend on the severity of the disease and the general state of the patient's health. Single or multiple administrations of the compositions can be administered according to the dosage and frequency required and tolerated by the patient. In all cases the composition should provide a sufficient amount of the adjunctive anti-cancer agents according to the present invention to treat the patient with all effectiveness. II. Suppressive genes of tumors and gene products A) Suppressors of known, preferred tumors. As explained above, in one embodiment this invention provides methods for inhibiting the growth and / or proliferation of cells by contacting the cells with a tumor suppressor nucleic acid and an adjunctive anticancer agent (e.g. paclitaxel, a paclitaxel derivative or a paclitaxel-like compound). The tumor suppressor genes are well known among branch experts and include, but are not limited to, RB, p53, APC, FHIT (see, for example, Siprashvill (1997) Proc. Na ti, Acad. Sci. USA 94: 13771 -13776), BRACA1 and BRCA2, VHL, WT, DCC, FAP, NF, MEN, E-cadherin, nm23, MMCI, and PTC. The RB gene or retinoblastoma is the prototypic tumor suppressor and has been perfectly characterized (see for example, Bookstein (1990) Sci ence 247: 712-715, Benedict (1980) Cancer Inves t., 8: 535-540, Riley (1990) Ann. Rev. Cell. Bi ol -, 10-1-29, and Wienberg (1992) Sci en 254: 1139 - 1146. Perhaps the best characterized tumor suppressor is the p53 that has been implicated in many neoblastomas. as well as in the genetic predisposition for the development of various tumors in families with the Li-Fraumeni syndrome (see for example, Wills (1994) Hum. Gene Therap., 5-1079-1088, U.S. Patent No. 5,532,220, WO 95/289048, and Harris (1996) J "Nat. Canc. Ins.88 (20): 1442), with which they describe donor expression and the use of p53 in gene therapy.) Other tumor suppressors include WT (ie, WT1 in llpl3) which is a gene characteristic of Wilms tumor (see Cali et al. (1990) Cell, 60:60: 509-520, Gessler (1990) Na ture 343: 774-778, and Rose et al. (1990) Cell, 60: 495-508). The tumor suppressor gene called FHIT, which is the abbreviation of the English term "Fragüe Histidine Triad", was found in a region on chromosome 3 (3pl4.2, also reported in 3p21), which is known to be particularly propitious. to translocations, breaks, and gaps and is believed to lead to cancers in the esophagus, stomach, and colon (see, eg, Ohta et al. (1996) Cell, 84: 587-597, GenBank Accession No: U469227). The tumor suppressor genes DCC (18q21) and FAP are associated with carcinoma in the colon (see for example, Hedrick et al. (1994) Genes Dev., 8 (10): 1174-1183, Gen Accession Bank No: X76132 for DCC, and Wienberg (1992) Science, 254: 1138-1146 for FAP). NF tumor suppressors (NFl in 17qll and NF2 in 22ql2) are associated with neurological tumors (eg, neurofibromatosis for NFl, see for example, Caivthon et al (1990) Cell, 62: 193-201, Viskochil et al. (1990) Cell, 62: 187-192, Wallace et al (1990) Science, 249: 181-186, and Xug et al. (1990) Cell, 62: 599-608; and meningioma and schwannoma for NF2). The MEN tumor suppressor is associated with tumors of the multiple endocrine neoplasia syndrome (see for example, Wienberg Science, 254: 1139-1146, and Marshall (1991) Cell, 64: 313-326). The VHL tumor suppressor is associated with von Hippel's disease Landau (Latíf (1993) Sci ence, 260: 1317-1320, GenBank Accession No: L15409). The widely published BRCA1 and BRCA2 genes are associated with breast cancer (see, for example, Skolnick (1994) Sci ence, 265: 66-71, GenBank Accession No: U14680 for BRCA1, and Teng (1996) Na ture Gene t. , 13: 241-244, GenBank Accession No: U43746). In addition, the E-cadherin gene is associated with the invasive phenotype of prostate cancer (Umbas (1992) Cancer Res., 52: 5104-5109, Bussemakers (1992) Cancer Res., 52: 2916-2999, GenBank Accession No : 272397). The NM23 gene is associated with tumor metastasis (Dooley (1994), Gene t., 93 (1): 63-66, Genbank Accession No: X75598). Other tumor suppressors include DPC4 (identified in 18q21) associated with pancreatic cancer, hMLHl (3p) and hMSH2 (2p) associated with colon cancers, and CDKN2 (pl6) and (9p) associated with melansma, and pancreatic and esophageal cancers . Finally the human PTC gene (a drosophil homolog of the drosophila dispatched gene (ptc)) is associated with the carcinoma syndrome in the basal nerve cells (NBCCS) and with carcinomas in the somatic basal cells (see for example, Hahn et al. (1996) Cel l, 85: 841-851). This list of tumor suppressor genes is neither exhaustive nor does it intend to have a limiting character; it is simply presented to illustrate the wide variety of tumor suppressors known. B) Identification and classification of suppressors of previously unknown tumors. Methods of identifying or assaying tumor suppressor genes are well known among those skilled in the art. Typically hyperproliferative cells are selected for whose aspects in terms of their gene loss or mutation thereof, is associated (correlated) with the hyperproliferative state. The most closed or severe test for a gene to qualify as a tumor suppressor gene (international abbreviation TSG) is its ability to suppress the tumorigenic phenotype of a tumor or cells derived from a tumor. Preferably, the tumor suppressor nucleic acid is introduced into tumor cells as a cDNA cloned into an appropriate expression vector, or an individual chromosome harboring a candidate gene, tumor suppressor, is introduced into tumor cells by the transfer technique of microcells. Alternatively, the product of the tumor suppressor gene (eg, a tumor suppressor polypeptide) is introduced into the cell or into the cells and the rate of proliferation of the cells is measured (eg, counting the cells or measuring the volume of the tumor, etc.). A complete or partial inhibition of proliferation, (as it can be the decrease in the proliferating rate), the inhibition of contacts, the loss of the invasive phenotype, a differentiation of cells as well as apoptosis, all constitute indicators of suppression of the tumorigenic phenotype (a reduced susceptibility to the neoplastic state). Methods for classifying or selecting tumors in order to identify altered or underexpressed nucleic acids are well known among those skilled in the art. Such methods include, without limitation, subtractive hybridization (see for example, Hampson (1992) Nucl ei c Aci ds Res. 20: 2899), a comparative genomic hybridization (international abbreviation CGH, see for example, WO 93/18186, Kallioniemi (1992) Sci ence, 258: 818), and the monitoring of expressions with the use of high-density arrays of nucleic acid probes (see for example, Lockhart (1996) Na ture Bio technology, 14 (13): 1675-1680). C) Preparation of p53 and other tumor suppressors. As indicated above, this invention involves contacting a cell, for example in vi tro, in a physiological solution (for example in blood), with a tissue organ or with an organism with a tumor suppressor nucleic acid. or gene product suppressors of tumors as a polypeptide. The nucleic acid or polypeptide, tumor suppressor, can be a nucleic acid or a polypeptide of any known tumor suppressor including, but not limited to: RB, p53, h-NUC (Chen (1995) supra), APC, FHIT, BRACA1, BRCA2, VHL, WT, DDC, FAP, NF, MEN, E-cadherin, nm23, MMACI, and PTC as described above. In a preferred embodiment, the tumor suppressor is a RB nucleic acid or a polypeptide or else a p53 nucleic acid or a polypeptide or one or more active fragments thereof. In a highly preferred embodiment, the tumor suppressor nucleic acid p53 or RB is present in an expression cassette under the control of a promoter that expresses the tumor suppressor gene or in cDNA when it is located in the target cell (ie say, the tumor cell). Methods for constructing such expression cassettes and / or vectors encoding tumor suppressor genes are well known to those skilled in the art, as described below. 1. Preparation of tumor suppressor nucleic acids. DNA encoding tumor suppressor proteins or protein subsequences of the same type, according to the present invention, can be prepared by any suitable method, including for example the cloning and restriction of certain appropriate sequences or direct chemical syntheses (for example with the use of information on sequences that already exists, as noted above), through methods such as for example Narang phosphotriester method (1979 Meth. Enzymol., 68: 90-99; the phosphodiester method of Brown et al., Me. Enzymol., 68: 109-151 (1979); Beaucage's diet ilphosphoramidite method et al., Tetra Lett., 22: 1859-1862 (1981), and the solid support method according to U.S. Patent No. 4,458,066.The chemical synthesis produces a single-stranded oligonucleotide.This product can be converted into double cord by hybridization with a complementary sequence or through a polymerization with a DNA polymerase using the single cord as a template or matrix.An expert would recognize that while the synthesis is limited s DNA chemistry to sequences of approximately 100 bases, it is possible to obtain longer sequences by ligature ligation of shorter sequences. As an alternative, subsequences can be cloned and the appropriate subsequences can be dissociated, that is to unfold, with the use of the appropriate restriction enzymes. Then the fragments can be ligated to produce the desired sequence of DNA. In one embodiment, the tumor suppressor nucleic acids according to this invention can be cloned with the use of DNA amplification methods such as the polymerase chain reaction (international PCR abbreviation). For example, the sequence or subsequence of the nucleic acid is amplified by PCR using a "primer", that is, a sense primer containing a specific restriction site (for example, Ndel) and an antisense primer containing another site of restriction (for example, HindIII). This will produce a nucleic acid encoding the tumor suppressor sequence or subsequence and having terminal restriction sites. This nucleic acid can then be easily ligated into a vector containing a nucleic acid encoding the second molecule and possessing the corresponding appropriate restriction sites. Suitable PCR primers can be determined by one skilled in the art with the use of published sequence information for any known tumor suppressor gene in particular, cDNA or protein. Appropriate restriction sites can also be added to the nucleic acid encoding the tumor suppressor protein or the protein subsequence of the same type by site-directed mutagenesis. site. The plasmid containing the tumor suppressor sequences or subsequences is dissociated with the appropriate restriction endonuclease and then ligated into the vector encoding the second molecule according to conventional methods. As indicated above, the sequences of the nucleic acids of many tumor suppressor genes are known. Thus we find, for example, the nucleic acid sequence of p53 in Lamb et al. , (1986) Mol. Cell Biol -, 6: 1379-1385, (GenBank Accession No: M13111). Similarly, the nucleic acid sequence of RB is described by Lee et al., (1987) Na ture, 329: 642-645.
(GenBank Accession No: M28419). The sequence of nucleic acids belonging to other tumor suppressors are available as indicated above in Section II (a). Taking advantage of the available information on the sequences, an expert of ordinary skill in the art will be able to clone the tumor suppressor genes into vectors suitable for use in this invention. Particularly preferred for use in the methods according to the present invention are p53 and RB tumor suppressors. The methods for cloning p53 and RB in suitable vectors for the expression of the respective proteins are well known among experts. suppressors of tumors or for gene therapy therapy applications. Thus, for example, the cloning and use of p53 have been described in detail by Wills (1994) supra; in U.S. Patent No. 5,532,220, in currently pending U.S. Patent Application USSN 08 / 328,673, filed October 25, 1994, as well as in International Report WO 95/11984. Typically, the expression cassette is constructed with the tumor suppressor cDNA operatively linked to a promoter, most preferably a strong promoter. (for example, the late major promoter of Ad2 (Ad2 MLP), or the promoter of the immediate early human cytomegalovirus (CMV) gene). In a particularly preferred embodiment, the promoter is followed by the tripartite leader cDNA and the cDNA tumor suppressor followed by a polyadenylation site (e.g., the polyadenylation site Elb) (see for example the currently pending US Patent Application). USSN 08 / 328,673, International Report WO 95/11984 and Wills (1994) supra). It will be noted that different tissue-specific promoters are also suitable. Thus, for example, a tyrosinase promoter can be used to target expression towards melanomas (see for example, Siders (1996) Cancer Res., 56: 5638-5646). In a particularly preferred embodiment it is expressed the tumor suppressor cDNA in a suitable vector to achieve gene therapy as described below. 2. Preparation of a tumor suppressor protein. a) Nde novo chemical syntheses. "With the use of known tumor suppressor polypeptide sequences, tumor suppressor proteins or subsequences thereof can be synthesized with the use of conventional chemical peptide synthesis techniques. When the desired subsequences are relatively short (for example when you want to have a particular antigenic determinant) you can synthesize the molecule as a single contiguous polypeptide.When you want to obtain larger molecules, you can synthesize the subsequences separately (in a or several units) to then create a fusion by condensation of the amino terminal of one molecule with the carboxyl terminal of the other molecule to form a peptide bond The solid phase syntheses in which the amino acid in C-terminal of the sequence to an insoluble support followed by the sequential addition of the rest of the amino acids in the sequence constitutes the preferred method for the chemical synthesis of the polypeptides of this invention. The techniques for these in-phase synthesis solid are described by Barany and Merrifield, Soli d-Phase Peptide Synthesi; pp. 3-284 in The Peptides: Analysi s, Synthesi s, Biology. Vol. 2: Special Methods in Peptide Synthesi, Part a. , Merrifield, et al. , J. Am. '- "Chem. Soc., 85: 2149-2156 (1963), and Stewart et al., Solid Phase Peptide Synthesi, 2nd ed. Pierce Chem. Co., Rockford, III. (1984). In one embodiment, the tumor suppressor proteins or their subsequences are synthesized with the use of recombinant DNA methodology.This generally involves the creation of a DNA sequence encoding the fusion protein, placing the DNA in an expression cassette under the control of a particular promoter, expressing the protein in a host, isolating the expressed protein and, if required, renaturing the protein.The methods for cloning the tumor suppressor nucleic acids are described above. a vector in particular.The nucleic acid sequences encoding the tumor suppressor proteins or the subsequences of such proteins can then be expressed in a variety of host cells, including E. coli, other h. bacterial uvts, ferments 5 and different higher eukaryotic cells such as COS, CHO and HeLa cell lines, as well as those myeloma cell lines. Since tumor suppressor proteins are typically found in eukaryotes, a eukaryotic host is preferred. The gene of the recombinant protein will be operably linked to suitable expression control sequences for each host. For the E. coli, this includes a promoter such as T7, trp or lambda promoters, a ribosome linker site and preferably a transcription termination signal. For eukaryotic cells the control sequences will include a promoter and preferably an enhancement agent derived from the immunoglobulin genes, SV40, cytomegalovirus, etc., as well as a polyadenylation sequence and they can include sequences of donor and splice acceptor. The plasmids of the invention can be transferred into the selected host cell by well-known methods such as calcium chloride transformation for E. coli and treatment with calcium phosphate or eletroporation for cells in mammals. Cells transformed by plasmids can be selected for resistance to antibiotics conferred by genes contained in plasmids such as amp, gpt, neo and hy genes. Once expressed, they can be purified recombinant tumor suppressor proteins, according to the conventional procedures of the art, including precipitation with ammonium sulfate, affinity columns, column chromatography, gel electrophoresis and the like (see in general terms, R. Scopes, Pro tein Purifi Cation, Springer-Verlag, NY (1982), Deutscher, Methods in Enzymology Vol. 182: Guide to Protein Purification, Academic Press, Inc., NY (1990)). Virtually pure compositions of a homogeneity of at least about 90 to 95% are preferred and homogeneities of 98% to 99% or more are most preferred. Once partially purified or even to the state of homogeneity, as desired, such polypeptides can then be used (eg, as immunogens for the production of antibodies). One skilled in the art would recognize that after chemical synthesis, biological expression or purification, this protein or such tumor suppressor proteins may possess a conformation virtually different from that corresponding to the original conformations of the constituent polypeptides. In this case, it may be necessary to denature and reduce the polypeptide to then cause this polypeptide to be refolded in the preferred conformation. Methods to reduce and denature the proteins and to induce this refolding are well known among those skilled in the art (see, Debinski (1993) -J. Biol. Chem., 268: 14065-14070; Kreitman (1993) Bioconj ug. Chem. , 4: 581-585; and Buchner (1992) Anal. Bi ochem. , 205: 263-270). Debinski (1993) supra, for example, describes the denaturation and reduction of body proteins of inclusion in the DTE of guanidine. After refolding the protein in a redox buffer containing oxidized glutathione and L-arginine. One skilled in the art will realize that many conservative variants of the nucleic acid and polypeptide sequences described herein provide functionally identical products. For example, due to the degeneracy of the genetic code, "silent substitutions" (this term identifies substitutions of a nucleic acid sequence that do not result in an alteration within a coded polypeptide) constitute an implicit characteristic of each sequence. of nucleic acid, which code for an amino acid. Similarly, "conservative amino acid substitutions" in one or several amino acids within an amino acid sequence are replaced by different amino acids with highly similar properties (see definitions section, supra), which are also they identify without problem as being highly similar to a given amino acid sequence or to a revealed sequence of nucleic acids, which codes for an amino acid. These conservatively substituted variants of each explicitly described sequence constitute a characteristic feature of the present invention. An expert would recognize that it is possible to make modifications in tumor suppressor proteins without decreasing their biological activity. Some modifications can be made to facilitate the cloning, expression or comparison of the target molecule in a fusion protein. Such modifications are well known to those skilled in the art and include for example an added methionine at the amino terminus to provide an initiation site, or additional amino acids (eg, poly His) placed at either terminal to create located restriction sites. conveniently or termination codons or purification sequences. Modifications in nucleic acids and polypeptides can be evaluated through routine classification techniques in assays suitable for the desired characteristic. For example, changes in the immunological character of a polypeptide can be detected by an appropriate immunological assay. Modifications of other properties, such as in the hybridization of nucleic acids to a target nucleic acid, the redox or thermal stability of a protein, hydrophobicity, susceptibility to proteolysis, or the tendency to globalize, are all tested in accordance with the conventional techniques. D) Delivery of tumor suppressors to target cells. The tumor suppressors used in the methods of this invention can be introduced into the cells, either as a protein, or as a nucleic acid. When the tumor suppressor is provided with a protein, a product is delivered with tumor suppressor gene expression (eg, a p53 or RB polypeptide or a fragment thereof having a tumor suppressor activity) to the cell objective using conventional methods for protein delivery (see discussion below). As an alternative when the tumor suppressor is a tumor suppressor nucleic acid (for example a gene, a cDNA, an mRNA, etc.) the nucleic acid is introduced into the cell with the use of conventional methods of delivering nucleic acids to cells. These methods typically involve the delivery methods of gene therapy in vi v or ex vi vo tal and as described below. Particularly preferred methods of delivering p53 or RB include the delivery of lipids or liposomes and / or the use of retroviral or adenoviral vectors. 1. Gene therapy in vivo. In a more preferred embodiment, tumor suppressor nucleic acids (e.g., cDNA or cDNAs encoding the tumor suppressor protein) are cloned into gene therapy vectors that are competent to transfect cells (other than human or other mammalian cells) ) in vi tro and / or in vi vo. Several approaches have been used to introduce nucleic acids into cells in vi ve, ex vivo and in vi tro. They include the delivery of lipid-based genes or liposomes (Memoirs WO 96/18372; WO 93/24640; Mannino (1988) BioTechniques 6 (7): 682-691; Rose, Patent North American number 5,279,833; WO 91/06309; and Felgner (1987) Proc. Na tl. Acad. Sci. USA 84: 7413-7414), and replication-defective retroviral vectors harboring a therapeutic polynucleotide sequence as part of the retroviral genome (see, for example, Miller (1990) Mol. Cell. Biol., 10: 4239; Kolberg ( 1992) J. NIH Res., 4:43, and Cornetta (1991), Hum. Gene Ther., 2: 215). For a review of procedures corresponding to gene therapies, see for example, Zhang (1996) C ncer Met s tasis Rev. 15: 385-401; Anderson, (1992) Sci ence, 256: 808-813; Nabel (1993) TIBTECH, 11: 211-127; Mitani (1993) TIBTECH, 11: 162-166; Mulligan (1993) Sci ence, 926-932; Dillon (1993) TIBTECH, 11: 167-175; Miller (1992) Na ture, 357: 455-460; Van Brunt (1988) Bi or technology, 6 (10): 1149-1154; Vigne (1995) J-estorative Neurology and Neurosci ence, 8: 35-36; Kremer (1995) Bri ti sh Medi cal Bulletin, 51 (1): 31-44; Haddada (1995) in Curren t Topi cs in Mírcrobiology and Immunology, Doerfler and Bóhm (eds) Springer-Verlag, Heidelberg Germany; and Yu (1994) Gene Therapy, 1: 13-26. Vectors useful in the practice of the present invention are typically derived from viral genomes. The vectors that can be used include DNA and RNA viruses, enveloped or unwrapped, recombinantly modified, which are preferably selected from the following species: baculoviridiae, parvoviridiae, picornoviridiae, herpesveridiae, poxviridiae, adenoviridiae, or picornnaviridiae. Chimeric vectors may also be used which advantageously exploit each in the properties of the related vectors (see for example, Feng (1997) Na ture Bi o technol ogy, 15: 866-870). Such viral genomes can be modified by recombinant DNA techniques in order to include the tumor suppressor gene and can be engineered to have replication deficiency, conditional repiration, or replication competence. In the preferred practice of the present invention, the vectors are replication deficient or have a conditional capacity of replication. Preferred vectors are derived from the adenoviral, viral, adeno and retroviral genomes. In the most preferred practice of the present invention, the vectors have replication incompetence and are derived from the human adenovirus genome. They use viral vectors with conditional replication to achieve selective expression in certain particular cell types while at the same time avoiding adverse infection with a broad spectrum. Examples of vectors with conditional replicator capacity are described by Bischoff et al. , (1996) Sci ence, 274: 373-376; Pennisi, E. (1996) Sci ence, 274: 342-343; Russell, S.J. (1994) Euro. J. of Cancer, 30A (8): 1165-1171. Additionally, the viral genome can be modified to include certain inducible promoters that achieve replication or transgene expression only under certain conditions. Certain examples of such inducible promoters are known in the scientific literature (see, for example, Yoshida and Hamada (1997) Biochem. Biophys.
Res. Comm. , 230: 426-430; I ida, et al., (1996) J. Virol., 70 (9): 6054-6059; Hwang, et al., (1997) J. Virol., 71 (9): 7128-7131; Lee, et al., (1997) Mol. Cell. Biol. 17 (9): 5097-5105; and Dreher, et al., (1997) J. Biol. Chem., 272 (46): 29364-29371. The transgene may also be under control of some tissue-specific promoter region, thus allowing expression of the transgene only in certain cell types. The widely used retroviral vectors include those based on the leukemia virus in rats, that is, murine type (MuLV), the leukemia virus in the gibbon monkey (GaLV), the simian immunodeficiency virus (SIV). , the human immunodeficiency virus (HIV), commonly known in the Spanish language with the abbreviation HIV, and its combinations. See, for example, Buscher (1992) J ". Virol., 66 (5) ^ 2731-2739; Johann (1992) J. Virol. , 66 (5): 1635-1640; Sommerfelt (1990) Virol., 176: 58-59; Wilson (1989) J. Vi rol., 63: 2374-2378; Miller (1991) J ". Virol., 65: 2220-2224; Wong-Staal, et al., PCT / US94 / 05700, and Rosenburg and Fauci (1993) in Fundamental Immunology, third edition Paul (ed) Raven Press, Ltd., New York and the references contained therein, and Yu (1994) supra). The vectors are optionally pseudotyped to extend the vector host range to those cells that are not infected by the retrovirus corresponding to the vector. Vesicular stomatitis virus-enveloped glycoprotein (VSV-G) has been used to construct HIV vectors pseudotyped by VSV-G, which can infect the cells of hematopoietic strains (Naldini et al., (1996) Science, 272: 263, and Akkina (1996) \ Virol-, 70: 2581). Vectors based on the adeno-associated virus (AAV) are also used to transduce cells with target nucleic acids, for example in the in vitro production of nucleic acids and peptides and in gene therapy procedures in vivo and ex vivo. See, Okada (1996) Gene Ther., 3: 957-964; West (1987) Virology, 160: 38-47; Cárter (1989) North American Patent number 4,797,368; Carter et al., WO 93/24641 (1993); Kotin (1994) Human Gene Therapy, 5: 793-801; Muzyczka (1994) ", Clin. Invst., 94: 1351, to obtain an overview of AAV vectors The construction of recombinant AAV vectors is described in a number of publications, including in the US Patent on behalf of Lebkowski, number 5,173,414; Tratschin (1985) Mol. Cell Biol., 5 (11): 3251-3260; Tratschin (1984) Mol Cell. Biol., 4: 2072-2081; Hermonat (1984) Proc. Natl. Acad Sci. USA, 81: 6466-6470, McLaughlin (1988) and Samulski (1989) ". Virol., 63: 3822-3828, with each other. The lines Cells that can be transformed by rAAV include those described in Lebkowski (1988) Mol. Cell. Biol., 8: 3988-3996. Other suitable viral vectors include the herpes virus and the vaccine virus. In a particularly preferred embodiment, the tumor suppressor gene is expressed in an adenoviral vector suitable for gene therapy. The use of adenoviral vectors in vivo and for gene therapy is described extensively in the literature of the patents and the scientific literature, see for example, Hermens (1997) J. Neurosci. Methods. , Jan., 71 (l): 85-98; Zeiger (1996) Surgery, 120: 921-925; Channon (1996) Cardiovasc. Res., 32: 962-972; Huang (1996) Gene Ther., 3: 980-987; Zepeda (1996) Gene Ther., 3: 973-979; Yang (1996) Hum. Mol. Ge-net-, 5: 1703-1712; Caruso (1996) Proc. Natl. Acad. Sci. USA, 93: 11302-11306; Rothmann (1996) Gene Ther., 3: 919-926; Haecker (1996) Hum. Gene Ther., 7.-1907-1914. The use of adenoviral vectors is described in detail in International Report WO 96/25507. Particularly preferred adenoviral vectors have been described by Wills (1993) supra; as well as in the pending US Patent Application USSN 98 / 328,673, as well as in the International Report WO 95/11984. Adenoviral vectors in particular Preferred include a deletion of some or all of the genes of protein IX. In a form of loyalty, the adenoviral vectors include deletions of the Ela and / or Elb sequences. In a more preferred embodiment the adenoviral construct is a construct encoding p53 such as, for example, A / C / N / 53 or A / M / N / 53 (see for example, USSN 98 / 328,673 and WO 95 / 11984. Equally preferred are vectors derived from the type of human adenovirus 2 or type 5. Those vectors are preferably deficient in replicating capacity due to modifications or deletions in the Ela and / or Elb coding regions. Other modifications in the viral genome to achieve particular expression characteristics or to allow repetitive administration, or a lower immune response, deserve the preference. More preferred are the recombinant adenoviral vectors having complete or partial deletions of the E4 coding region, and which optionally retain E4 ORF6. The coding sequence E3 can be deleted but preferably stored. Specifically, it is preferred that the E3 promoter operator region be modified to increase the expression of E3 in order to achieve a more favorable immunological profile for the therapeutic vectors. To a greater degree, adenoviral human type 5 vectors containing a DNA sequence coding for p53 under control of the cytomegalovirus promoter region and the tripartite leader sequence possessing E3 under the control of the CMV promoter and the deletion of the E4 coding regions, while regions E4 0RF6 and ORF 6/7 are saved. In the most preferred practice of the invention, exemplified here, the vector is ACN53. In a way . of particularly preferred embodiment, the tumor suppressor gene is p53 or RB. As explained above, the cloning and use of p53 are described in detail by Wills (1994) supra; as well as in the pending US Patent Application USSN 08 / 328,673, filed October 25, 1994, and also in International Report WO 95/11984. 2. Ex vivo gene therapy. In one embodiment, the methods according to this invention are used to inhibit hyperproliferative (e.g., neoplastic) cells in a subject (e.g., a mammal including, but not limited to, the rat, the mouse, bovine species, porcine, equine, canine, feline, largomorphic or human). The pathological hyperproliferative cells are characteristic of diseased states including, without limitation, Grave's disease, psoriasis, benign prostatic hypertrophy, Li-Fraumeni syndrome, breast cancer, sarcomas, bladder cancer, colon cancer, lung cancer, different leukemias and lymphomas and other neoplasms. The particular application of the methods of this invention in particular, provides an auxiliary to take advantage of an adequate sample of pathological hyperproliferative cells. Thus, for example, the hyperproliferative cells that contaminate the hematopoieic precursors during the reconstitution of the bone marrow can be eliminated by the application of the methods of this invention. Typically, these methods involve obtaining a sample of the subject's organism. The sample is typically a preparation of heterogeneous cells containing both phenotypically normal and pathogenic (hyperproliferative) cells. The sample is contacted with the tumor suppressor nucleic acids or proteins of the same type and the adjunctive anticancer agent, according to the methods of this invention. The tumor suppressor gene can be delivered, for example, in a viral vector, such as a retroviral vector or an adenoviral vector. The treatment reduces the proliferation of pathogenic cells to provide a sample that contains a higher proportion of normal than pathogenic cells, which can be reintroduced into the organism of the subject. Transformation of ex vivo cells for diagnostic, research or gene-based therapy purposes (for example through the re-infusion of transformed cells into the host organism) is well known to those skilled in the art. . In a preferred embodiment, the cells of the subject's organism are isolated, transfected with the tumor suppressor gene or cDNA of this invention and this material is reintroduced into the body of the subject (for example a patient). Several types of cells suitable for ex vivo transformation are well known to those skilled in the art. Particularly preferred cells are progenitor cells or driving cells (see for example, Freshney (1994) Cul ture of Animal Cells, a Manual of Basic Technique, third edition, Wiley-Liss, New York, and references cited therein. work for a discussion on how to isolate and grow the cells of patients). The transformed cells are cultured by means well known in the art. See also Kuchler (1977) Biochemi cal Methods in Cell Cul ture an Virology, Kuchler, R.J., Dowden, Hutchinson and Ross, Inc., and Atlas (1993) CRC Handbook of Microbiology cal Media (Parks edition) CRC press, Boca Ratón, Fl. The cellular systems in mammals will often be in the form of single layers (monolayers) of cells although suspensions of mammalian cells are also used. Alternatively cells can be derived from those stored in a bank of cells (such as a blood bank). Illustrative examples of mammalian cell lines include the HEC-lB cell line, the VERO and Hela cells, the Chinese hamster ovarian cell lines (CHO), the cell lines of W138, BHK, Cos-7 or well MDCK (see for example, Freshney, supra). In a particularly preferred embodiment, the driving cells are used in ex vivo procedures for cell transformation and gene therapy. The advantage of using propellant cells is that they can be differentiated into other types of cells in vi tro or that they can be introduced into a mammal (such as the donor of the cells) where they will be grafted into the bone marrow. Methods for differentiating promoter cells (e.g., CD34 + progenitor cells) in vi tro are known for introducing them into clinically important types of immune cells that utilize cytokines such as GM-CSF, IFN-gamma and TNF-alpha (see example, Inaba (1992) J ". Exp. Med., 176: 1693-1702; Szabolca (1995) 154: 5851-5861). Instead of using the driving cells, T cells or B cells are also used in some embodiments in the ex vivo procedures. Various techniques are known for isolating T and B cells. The expression of surface markers facilitates the identification and purification of such cells. Methods of identifying and isolating cells include the FACS system, incubation in flasks with fixed antibodies that bind to the particular cell type, and washing with magnetic spheres. The driving cells are isolated for transduction and differentiation with the use of known methods. For example, in mice, cells from the bone marrow are isolated by sacrificing the mouse and cutting the bones of the legs with scissors. The booster cells are isolated from the cells of the bone marrow, washing the cells of the bone marrow with antibodies that bind unwanted cells such as CD4 + and CD8 + (T cells) CD45 + (panB cells), GR-1 (granulocytes), and the Iad (cells that present differentiated antigens). For an example of this protocol see for example, Inaba (1992) supra. In humans, aspirations of the bone marrow are made from the upper edges of the iliac bone, for example under general anesthesia in the operating room. The aspirations of the bone marrow cover an amount of the order of 1,000 ml and the material of the posterior iliac bones and the upper ridges is collected. When the total number of cells collected is less than about 2 x 108 / kg, a second aspiration is performed using the sternum and the anterior ridges of the iliac bone in addition to the posterior ridges. During the operation, packed and irradiated red blood cells are administered to replace the volume of the marrow collected by the aspiration. Human hematopoietic progenitor and progenitor cells are characterized by the presence of an antigen on the CD34 surface membrane. This antigen is used for purification purposes, for example in affinity columns that bind CD34. Once the bone marrow is harvested, the mononuclear cells are separated from the other components by centrifugation with a ficol gradient. This action can be carried out by means of the so-called semi-automated method using a cell separator (for example, a Baxter Fenwal CS3000 + device or a Terumo machine). Low-density cells, which are mostly made up of mononuclear cells, are collected and then incubated in plastic bottles at a temperature of 37 ° C. for about an hour and a half. Adherent cells (monocytes, macrophages and B cells) are discarded. The non-adherent cells are then harvested and incubated with a monoclonal anti-CD34 antibody (for example, in 9C5 murine antibody) at 4 ° C, for a time of 30 minutes, with gentle rotation. The final concentration for the anti-CD34 antibody is preferably of the order of 10 μg / ml. After two washes, paramagnetic micro spheres are added (for example, the Dyna Beads spheres, supplied by Baxter Immunotherapy Group, Santa Ana, California) coated with sheep anti-mouse IgG (Fc) antibody to the cell suspension, at a ratio of approximately two cells per sphere. After another incubation period of about 30 minutes at a level of the order of 4 ° C, the aroseted cells are collected with the magnetic spheres, applying a magnet. Chemopapain (Baxter Immunotherapy Group, Santa Ana, California) can be added until a final concentration of 200 U / ml is reached in order to release the spheres of CD34 + cells. As an alternative and preferably, an isolation procedure based on an affinity column, which creates a binding to CD34, or antibodies bound to CD34 (see, for example, Ho) can be used. (1995) Ste-ii Cell s, 13 (suppl 3): 100-105 and Brenner (1993) Journal of Hematotherapy, 2: 7-17).
In another embodiment, hematopoietic stem cells can be isolated from fetal cord blood. Yu (1995) Proc. Natl. Acad. Sci. USA, 92: 699-703 describes a preferred method of transducing CD34 + cells from the blood of the human fetal cord using retroviral vectors. 3. Suppressor and Tumor Express Nucleic Acid Administration; Vectors and Expression Cassettes. Administration routes. Expression cassettes and vectors (e.g., retroviruses, adenoviruses, liposomes, etc.) containing nucleic acids that suppress and express tumors, of therapeutic type, according to the present invention, can be administered directly to the organism to transduce the cells in vi. The administration is carried out through any of the routes normally used to introduce a molecule in ultimate or definitive contact with the cells of the blood or tissues, for example by the systemic, regional or local route, as has been treated in detail above, for the administration of adjunctive anticancer agents. The "packaged" nucleic acids (at least one coding sequence, tumor suppressor with a promoter) are administered in any suitable manner, preferably with pharmaceutically carriers acceptable, as have also been discussed above. Suitable methods for administering such packaged nucleic acids are available and they are well known to those skilled in the art and although more than one route can be used to administer a specific composition, often a particular route can provide a more immediate reaction and more effective than another. For example, administration of a recombinant adenovirus vector engineered to express a tumor suppressor gene can elicit an immune response, specifically, an antibody response, against the adenoviral vector. Some patients may have pre-existing antibodies or anti-adenoviral antibodies. Thus, in some circumstances, a regional or local administration, instead of a systemic one, of the adenoviral vector that expresses and suppresses tumors, is optimal and highly effective. For example, and as discussed below, ovarian cancer limited to the abdominal cavity constitutes a clinical scenario in which regional p53 gene therapy, i.e., intraperitoneal (IP) administration, should be considered as a preferred treatment plan. The administration of recombinant adenoviruses via the IP route also results in a infection of the peritoneal lining and absorption of the adenoviral vector in the systemic circulation (another means of regional administration may also result in the introduction of the adenoviral vector into the systemic circulation). The extent of this effect may depend on the concentration and / or the total amount of viral particles administered by the IP route. If it is desired to have the systemic effect, preference may be given to a higher concentration for several consecutive days. Local administration of the suppressive and tumor expressing adenoviral vector, according to the present invention, is also preferred under certain circumstances, for example, when the patient has pre-existing anti-adenoviral reactive antibodies. Such "local administration" can be for example by intratumoral injection, if it is internal, or in the form of a mucosal application if it is external. Alternatively, a "local administration" effect can be created by targeting the adenoviral vector to the tumor using, for example, certain tumor-specific antigen-recognizing reagents (such as antibodies) on liposomes or on the adenovirus itself. Formulations Pharmaceutically acceptable carriers are determined in part by the specific composition that is being administered as well as by the particular method used to administer the composition. Accordingly, there is a wide variety of suitable formulations of the pharmaceutical compositions according to the present invention. Formulations suitable for allowing oral administration of pharmaceutical compositions comprising suppressor and tumor expressing nucleic acids, may consist of (a) liquid solutions, such as an effective amount of packed nucleic acid, suspended in diluents, such as water, salt water or PEG 400; (b) capsules, sachets "sachets" or tablets each containing a predetermined amount of the active ingredient, such as liquids, solids, granules or gelatin; (c) suspensions in an appropriate liquid; and (d) suitable emulsions. The tablet-like forms may include one or more specimens of: lactose, sucrose, mannitol, sorbitol, calcium phosphates, corn starch, potato starch, tragacanth, microcrystalline cellulose, acacia, gelatin, colloidal silicon dioxide, croscarmellose sodium , talc, magnesium stearate, stearic acid, and other excipients, colorants, fillers, binders, diluents, buffering agents, wetting agents, preserving agents, seasoning agents, dyes, disintegrating agents and carriers pharmaceutically compatible The forms of lozenges may comprise the active ingredient in some flavor, usually sucrose and acacia or tragacanth as well as lozenges comprising the active ingredient in an inert base, such as gelatin and glycerin or sucrose and acacia emulsions, gels and the like which also contain of the active ingredient, the carriers known in the art. Packaged nucleic acids, alone or in combination with other suitable components, can be converted into aerosol formulations (ie, they can be "nebulized"), to be administered via inhalation. Aerosol formulations can be placed within acceptable blowing agents under pressure, such as dichlorodifluoromethane, propane, nitrogen and the like. Appropriate formulations for rectal administration include for example suppositories consisting of the nucleic acid packaged with a suppository base. Suitable suppository bases include natural or synthetic triglycerides or paraffin hydrocarbons. Above it is also possible to use rectal gelatin capsules consisting of a combination of the nucleic acid packaged with a base, which includes for example liquid triglycerides, glycols of polyethylene and paraffin hydrocarbons. Formulations suitable for parenteral administration such as, for example, intra-articularly (in joints), intravenously, intramuscularly, intradermally, intraperitoneally and subcutaneously, include sterile and isotonic solutions for injection, of aqueous and non-aqueous type, which may contain antioxidants, buffers, bacteriostats and dissolved materials that make the formulation isotonic with the blood of the contemplated container, as well as the sterile, aqueous and non-aqueous suspensions, which may include suspending agents, solubilizers, thickening agents, stabilizers and preservation agents. In the practice of the present invention, the compositions can be administered, for example, by intravenous infusion, orally, topically, intraperitoneally, intravesicularly or intrathecally. Parenteral administration and intravenous administration are the preferred methods of administration. Packaged nucleic acid formulations can be presented in sealed containers or containers of single dose or multiple doses, such as ampoules and bottles. The formulations according to the invention can be prepared as injectable solutions and suspensions, starting from sterile powders, granules and tablets of the class previously described. The exact composition of the formulation, the concentration of the reagents and the nucleic acid in the formulation, its pH, buffers and other parameters will vary according to the mode and site of administration (eg, systemic, regional or local administration). ) and the needs related to aspects of storage, handling, shipping and shelf life of the particular pharmaceutical composition. The optimization of these parameters depending on the particular need for the formulation, can be achieved through routine methods and any of the ingredients and parameters for the known injectable formulations can be used. An example of a suitable formulation is, for example, a recombinant p53 wild-type expressing adenovirus vector, according to the present invention (rAd5 / p53) at a concentration of the order of 7.5 × 10 11 up to 7.5 × 10 10 particles per ml. , sodium phosphate monohydrate at 0.42 mg / ml, dibasic anhydride sodium phosphate in an amount of 2.48 mg / ml, sodium chloride with sodium phosphate monohydrate at 5.8 mg / ml, sucrose at 20.0 mg / ml , the magnesium chloride hexahydrate at 0.40 mg / ml, formulation that is typically stored in dosages of 1.0 ml. Cells transduced by nucleic acid packaged as described above within the context of an ex vivo therapy, they can also be administered intravenously or parenterally, as described above. The dose administered to a patient, within the context of the present invention, must be sufficient to achieve the beneficial therapeutic response in the patient over time. The dose will be determined based on the efficacy of the specific vector employed and according to the condition of the patient as well as according to the body weight or surface area of the patient to be treated. The size of the dose will also be determined by the existence, nature and extent of the various adverse side effects that accompany the administration of a particular vector, or according to the type of cell transduced in a given patient. By establishing the effective amount of the vector to be administered in the treatment, the physician evaluates the circulating plasma levels of the vector, the toxicities of the vector, the progression of the disease and the production of anti-vector antibodies. The typical dose for a nucleic acid depends to a large extent on the route of administration and the delivery system of genes. According to the delivery method, the dosage can easily fluctuate between 1 μg and 100 mg or more. In general, the equivalent dose of a naked nucleic acid of a vector is from about 1 μg to 100 μg for a typical 70 kilogram patient, and doses of vectors including a viral particle are calculated to provide an equivalent amount of therapeutic nucleic acid. For administration, the transduced cells according to the present invention can be applied, at a rate determined by the LD50 of the vector, or according to the type of the transduced cell and according to the lateral effects of the vector or the cell type at different concentrations, as it is applied to the mass and the patient's general state of health. The administration can be carried out through individual or divided doses, as described below. In a preferred embodiment, blood samples are obtained before the infusion and stored for analysis. Vital signs and oxygen saturation are closely monitored by pulse oximetry. Blood samples are preferably obtained 5 minutes and 1 hour after the infusion and they are saved for further analysis. In ex vi ve therapy, leucoferesis, transduction and reinfusion can be repeated every 2 to 3 months. After the first treatment, infusions can be made on the basis of a traveling patient, according to the opinion of the doctor. If the reinfusion is administered as a walking patient, the participant is monitored at least 4 hours and preferably at 8 hours after the therapy. As described above, the adenoviral constructs can be administered systemically (for example intravenously) regionally (for example intraperitoneally) or locally (for example by intratumoral, peritumoral or intracystic injection, for example in the case of gallbladder cancer). ). Particularly preferred modes of administration include intraarterial injection, more preferably injection into the intrahepatic artery (for example for the treatment of liver tumors), or when it is convenient to deliver a composition to a brain tumor, it is applied to a carotid artery. or to an artery within the carotid system of arteries (for example the occipital artery, the atrial artery, the temporal artery, the cerebral artery, the maxillary artery, etc.). The supply for treatment of lung cancer can be effected for example by the use of a bronchoscope. Typically such administration occurs in an aqueous, pharmacologically acceptable buffer, as described above. However, in a particularly preferred embodiment, the adenoviral constructs or cassettes with expression of tumor suppressors in lipid formulation, more particularly in complex with liposomes for lipid and nucleic acid complexes (for example as described by Debs and Zhu (1993) International Application WO 93/24640; Mannino (1988) supra; Rose, U.S. Patent No. 5,279,833; Brigham (1991) International Application WO 91/06309; and Felgner (1987) supra) or in an encapsulated manner in liposomes more preferably in immunoliposomes directed to specific tumor markers. It will be noted that these lipid formulations can also be administered topically, systemically, or through an aerosol. 4. The improvement in the delivery of tumor suppressors. The administration of tumor suppressors can be improved by the use of one or more "supply enhancing agents". A "delivery enhancing agent" refers to any agent that improves the delivery of a therapeutic gene, such as a tumor suppressor gene to a cancerous tissue or organ. This improved supply can be effected through different mechanisms. One such mechanism may be the disruption of the glycosaminoglycan protective layer on the epithelial surface of an organ or tissue (eg the vesicle). Examples of such agents Supply enhancers are detergents, alcohols, glycols, surfactants, bile salts, heparin antagonists, cyclooxygenase inhibitors, hypertonic salt solutions and acetates. Alcohols include, for example, aliphatic alcohols such as ethanol, N-propanol, isopropanol, butyl alcohol and acetyl alcohol. Glycols include glycerin, propylene glycol, polyethylene glycol and other low molecular weight glycols, glycerol and thioglycerol. In addition, acetates such as acetic acid, gluconol acetate and sodium acetate are examples of supply enhancing agents. Hypertonic salt solutions such as 1M NaCl are also examples of supply enhancing agents. Examples of surfactants are sodium dodecyl sulfate (SDS international abbreviation) and lysolecithin, polysorbate 80, nonylphenoxypolyoxyethylene, lysophosphatidylcholine, polyethylene glycol 400, polysorbate 80, polyexyethylene ethers, ether-based surfactants of polyglycol and DMSO. Bile salts such as taurocholate, sodium tauro-deoxycholate, deoxycholate, chenodeoxycholate, glycocholic acid, glycokenedeoxycholic acid and other astringents such as silver nitrate can also be used. Heparin antagonists can also be used, like quaternary amines, prolamine sulfate. The use of cyclooxygenase inhibitors, such as sodium salicylate, salicylic acid, and non-steroidal anti-inflammatory drugs (NSAIDS), such as indomethacin, naproxen, and diclofenac, can also be used. Detergents include anionic, cationic, zwitterionic, and nonionic detergents. Exemplary detergents include, but are not limited to taurocholate, deoxycholate, taurodeoxycholate, cetylpyridium, benalkonium chloride, ZWITTERGENTF-3 -14 detergent, and CHAPS hydrate. (3 - [(3-colamidopropyl) dimethylammoniol] -1-propanesulfonate, Aldrich), Big CHAP (as described in US Patent Application Serial No. 08 / 889,355, filed July 8, 1997, and, according to International Application WO). 97/25072, of July 17, 1997), the "Deoxi Big CHAP . { ibi d), the detergent TRlTON®-X- 100, C12E8, the Octyl-B-D-Glucopyranoside, the detergent PLURONIC-F68, the detergent TWEEN 20, and the detergent TWEEN 80 (CALBIOCHEM Biochemicals). In one embodiment, the supply enhancing agent is included in buffer in which the delivery system of the recombinant adenoviral vector is formulated. The delivery enhancing agent can be administered before the recombinant virus or simultaneously with the virus.
In some forms of execution the agent is provided enhancer of delivery with the virus in advance as a virus preparation is mixed with the formulation of the delivery enhancing agent before being administered to the patient. In other embodiments, the delivery enhancing agent and the virus are provided in a single bottle to the person in charge of the administration. In the case of a pharmaceutical composition comprising a tumor suppressor gene contained in a recombinant adenoviral vector delivery system formulated in a buffer that further comprises a delivery enhancing agent, the pharmaceutical composition is prefer administered in a period of time from about 5 hours. minutes to 3 hours, and prefer between 10 minutes and 120 minutes, and more prefer between about 15 minutes and 90 minutes. In another embodiment, the delivery enhancing agent can be administered before administering the delivery system of the recombinant adenoviral vector containing the tumor suppressor gene. The previous administration of the supply enhancing agent can be in a time frame of 30 seconds to 1 hour, and prefer between 1 minute and 10 minutes, and more preferably between approximately 1 minute and 5 minutes before the administration system of the adenoviral vector containing the tumor suppressor gene is administered. The concentration of the delivery enhancing agent will depend on a number of factors known to one of ordinary skill in the art such as the specific delivery enhancing agent used, the buffer, the pH, the target tissue or organ as well as the mode of administration. The concentration of the improving agent of supply will be in the range of 1% to 50% (v / v), preferably between 10% and 40% (v / v) and more preferably between 15% and 30% (v / v). Preferably, the detergent concentration in the final formulation administered to the patient is of the order of 0.5 - 2X the critical micellization concentration (CMC). A preferred concentration of Big CHAP is of the order of 2 to 20 mM, more preferably between 3.5 and 7 mM. The buffer containing the delivery enhancing agent can be any pharmaceutical buffer, such as, for example, phosphate-buffered salt water, or sodium phosphate with sodium sulfate, Tris buffer, glycine buffer, sterilized water and others. tampons known to those skilled in the art, such as those described by Good et al. , (1966) Biochemi stry, 5: 467. The pH of the buffer within the pharmaceutical composition comprising the tumor suppressor gene contained in the adenoviral vector delivery system may be in the range of 6.4 to 8.4, preferably between 7 and 7.5, and more preferably between 7.2 and 7.4. A preferred formulation for administering a recombinant adenovirus is of the order of 109-1011 PN / ml of virus, about 2 to 10 M of Big CHAP or about 0.1 to 1.0 mM of detergent TRITON * -X-100, in buffered salt water with phosphate (PBS), plus an approximate amount of 2 to 3% of sucrose (weight / volume) and about 1 to 3 mM of MgCl2, and a pH of the order of 6.4 to 8.4. The use of the best supply agents is described in detail in the North American application currently pending USSN OS / 119, 627, which was deposited on January 7, 1997. In order to facilitate improved gene transfer for nucleic acid formulations comprising commercial preparations of Big-CHAP, the concentration of Big CHAP will vary based on your commercial source. When the Big CHAP product is obtained from the company CALBIOCHEM, it is preferable that the concentration is in a range of 2 to 10 millimolars. More preferred is 4 to 8 millimolars. And the most preferred level is of the order of 7 millimolars. When the Big CHAP of Sigma is obtained, it is preferred that the concentration of the same be in a range of 15 to 35 millimolars. And more preferred is the margin of 20 to 30 millimolars. While the most preferred level corresponds to 25 millimolars. In another embodiment of the invention, supply enhancement agents are provided, which Formula 1 possesses: Xi- in which n is an integer from 2 to 8, Xi is a group of cholic acid or a deoxycholic acid group, and X and X3 are each independently selected from the group consisting of a group of colic acids, a group of acids deoxycholics and a group of saccharins. At least one of X2 and X3 is a group of saccharides. The saccharide group can be selected from the group consisting of the groups of pentose monosaccharides, hexose monosaccharides, pentose-pentose disaccharides, hexose hexose disaccharides, pentose hexose disaccharides and hexose-pentose disaccharides. In a preferred embodiment, the compounds of the present invention possess Formula II: wherein Xi and X2 are selected from the group consisting of a group of colic acids and a group of deoxycholic acids and X-3 is a group of saccharides. Preferably these compounds are used in a range of the order of 0.002 to 2 mg / ml, more preferably 0.02 to 2 mg / ml, and more preferably 0.2 to 2 mg / ml in the formulations of the invention. Most preferred is the order level of 2 mg / ml. Salt water buffered with phosphate (PBS) is the preferred solubilizing agent for these compounds. However, an expert of ordinary skill in the art will recognize that certain additional excipients and additives may be desirable to achieve solubility characteristics of such agents for different pharmaceutical formulations. For example, the addition of well-known solubilizing agents such as detergents, fatty acid esters and surfactants can be carried out in appropriate concentrations to facilitate the solubilization of the compounds in the different solvents used. When the solvent is PBS, a preferred solubilizing agent is Tween 80 in a concentration of the order of about 0.15%. 5. Administration of tumor suppressor proteins. The tumor suppressor proteins (polypeptides) can be delivered directly to the tumor site by injection or they can also be administered systemically, as described above. In a preferred embodiment, the tumor suppressor proteins are combined with a pharmaceutically acceptable carrier (excipient) to form a pharmacological composition, as described above. The tumor suppressor polypeptide will be administered in a therapeutically effective dose. Thus, the compositions will be administered in an amount sufficient to cure or at least partially arrest the disease and / or its complications. The effective amounts for this use will depend on the severity of the disease and the general state of health of the patient. It will be recognized that tumor suppressor polypeptides, when administered orally, should be protected from digestion. This is typically accomplished either by forming a complex of the polypeptide with a composition to render it resistant to acidic or enzymatic hydrolysis, or by packaging the polypeptide in a carrier of correct strength, such as a liposome as described above. polypeptides for oral delivery are well known in the art (see for example U.S. Patent 5,391,377 which discloses lipid compositions for oral delivery of therapeutic agents). III. Combination Pharmacists The tumor suppressor and the adjunctive anticancer agent can be administered individually, either with the nucleic acid or the polypeptide, tumor suppressor, administered before the adjunctive anticancer agent (pretreatment with tumor suppressor) or it is also possible to administer the adjunctive anticancer before of the nucleic acid and / or polypeptide, tumor suppressor (pretreatment with the anti-cancer drug). Of course, the nucleic acid and / or polypeptide, tumor suppressor, as well as the adjunctive anti-cancer agent can be administered simultaneously. In one embodiment, the nucleic acid and / or polypeptide, tumor suppressor, as well as the adjunctive anti-cancer agent are administered as a single pharmacological composition. In this embodiment the nucleic acid and / or the polypeptide, tumor suppressor, and the adjunctive anti-cancer agent can be suspended or solubilized in a single homogenous delivery vehicle. As an alternative it is pointed out that nucleic acid and / or polypeptide, tumor suppressor, as well as the adjunctive anticancer agent can be suspended or solubilized each in different delivery vehicles which, in turn, are suspended (discharged) in a single excipient , either at the time of administration or continuously. Thus, for example, an adjunctive anticancer agent can be solubilized in a polar solvent (for example paclitaxel in ethanol) and a complex of the tumor suppressor nucleic acid can be formed with a lipid which is then stored together in a suspension or, as alternative, are combined at the time of administration. Several are described above supply vehicles, excipients of suitable character, and so on. IV. Treatment regime: combined and individual therapy. A) Treatment regimen with tumor suppressors. It has been a discovery of this invention that nucleic acids or polypeptides, tumor suppressors, more particularly tumor suppressor nucleic acids, show greater efficacy in the inhibition of tumor growths when administered in multiple doses rather than in a single dose. single dose. Thus, this invention provides a treatment regimen for a tumor suppressor gene or polypeptide, comprising multiple administrations of the nucleic acid or polypeptide, tumor suppressor. The tumor suppressor protein or tumor suppressor nucleic acid can be administered (with or without the adjunctive anti-cancer agent) in a total dose ranging from about 1 x 109 to about 1 x 1014, about 1 x 199 to about 7.5 x 10 15, preferably in an order of 1 x 10 11 to about 7.5 x 10 13 adenovirus particles, in a treatment regimen selected from the group consisting of: the total dose in a single dose, the total dose divided over the course of days, either administered daily for 5 days, the total dose divided over a period of 15 days, or administered daily for 15 days, and the total dose divided by 30 days, or administered daily for 30 days. This method of administration can be repeated for two or more cycles (more preferably for three cycles) and the two or more cycles can be spaced by three or four weeks The treatment may consist of a single-dose cycle or the dosage cycles may range from about 2 to about 12, and more preferably from about 2 to about 6 cycles. A particularly preferred treatment regimen includes the total dose distributed over a period of 5 days and administered daily, but it is also that in which the total dose is distributed over a period of 15 days and is administered daily, as well as the system in which it is administered. The total dose is distributed in 30 days and is administered daily. In some preferred embodiments, a daily dose in the range of 7.5 x 10 9, to about 7.5 x 10 15, preferably of the order of 1 x 10 12 to about 7.5 x 10 13, of adenovirus particles may be administered, each day up to a total of 30 days (for example, a regimen of 2 days, 2 to 5 days, 7 days, 14 days, or 30 days with the same dose being administered every day). The multiple regimen can be repeated in recurring cycles of 21 to 28 days. In some embodiments, different routes of administration will result in the use of different preferred dosage ranges. For example, for an intra-hepatic arterial supply, a preferred range will typically be from 7.5 x 109 to about 1 x 1015, more preferably from about 1 x 1011 to about 7.5 x 1013 adenovirus particles per day, for a period of 5 to 14 days. These regimens may also include the administration of adjunctive anticancer agents, such as FUDR or 5'-deoxy-5-fluorouridine (5 'DFUR), or the hydrochloride of irinotecan (CPT-11); 7-ethyl-10- [4- (1-piperidino) -1-piperidino] carbonyloxycamptothecin). For intratumoral delivery, a preferred range will typically be one that is between 7.5 x 109 and 1 x 1013, more preferably between 1 x 1011 and 7.5 x 10 12 adenovirus particles per day. For intraperitoneal delivery a preferred range will typically be one that is between 7.5 x 109 and 1 x 1015, more preferably between 1 x 1011 and 7.5 x 1013 adenovirus particles per day, for a period of 5 to 10 days. B) Treatment regimen with combination therapies. When the tumor suppressor is used in combination with an adjunctive anticancer agent, the tumor suppressor nucleic acid is administered in a total dose, as described above. In combination, the adjunctive anti-cancer agent is administered in a total dose depending on the agent used. For example, paclitaxel or a paclitaxel derivative is administered in a total dose ranging from 75 to 350 mg / m2 for 1 hour, 3 hours, 6 hours, or 24 hours in a treatment regimen selected from the group consisting of the material in a single dose, in a dose administered daily on days 1 and 2, in a dose administered daily on day 1, 2 and 3, a daily dosage for 15 days, a daily dose for 30 days, a daily continuous infusion during 15 days, and a daily continuous infusion for 30 days. A preferred dose is 100 to 250 mg / m2 for a period of 24 hours. Previous treatment with an adjunctive anti-cancer agent (for example paclitaxel) before performing the Treatment with a tumor suppressor nucleic acid improves the effectiveness of the tumor suppressor. Thus, in a particularly preferred embodiment, the cell, tissue or organism is treated with an adjunctive anticancer agent before the tumor suppressor nucleic acid. Treatment with the adjunctive anti-cancer agent, preferably precedes treatment with the tumor suppressor nucleic acid, for a period of approximately twenty-four hours although longer or shorter periods are acceptable. The pretreatment is particularly effective when the adjunctive anti-cancer agent is a paclitaxel-like compound, more preferably paclitaxel, or a paclitaxel derivative (eg Taxol or Taxotere). Particularly preferred tumor suppressors are RB and p53, among which p53 is most preferred, in particular p53 in an adenoviral vector (for example A / C / N / 53). V. Treatment and prophylaxis of metastasis. As Examples 2 and 3 are illustrated, therapy with replacement of tumor suppressor genes (e.g., p53) as has been demonstrated, has an effective effect against human tumor cells in vi tro xeno-inj erts of human tumors in immunocompromised hosts and human lung tumors (in vivo). Surgical reduction or removal of primary tumors in patients often results in a return of tumor growth at the primary site and a metastasis of the tumor from that site due to the "nests" of microscopic size of tumor cells, which do not They were removed by the surgeon. Alternatively, to safely establish that the entire tumor has been removed from a primary site, the patient You may undergo a disfiguring surgery in which a large amount of normal tissue is removed around the site of the primary tumor. In another embodiment, the invention provides methods for inhibiting the growth and / or proliferation of metastases (metastatic cells). The method generally involves the systemic or topical administration of a tumor suppressor, more preferably the topical administration of p53 or RB. A) Systemic treatment. As explained in Examples 2 and 3, systemic treatment (e.g., intravenous injection) of tumor suppressor vectors (e.g., A / C / N / 53) inhibited the progression of in vivo metastases. Thus, in one embodiment, this invention provides methods for inhibiting the progression of a metastatic disease by administering to an organism a nucleic acid and / or a tumor suppressor polypeptide, as described above. The tumor suppressor is preferably a tumor suppressor nucleic acid, more preferably a p53 tumor suppressor nucleic acid and more preferably a p53 nucleic acid in an adenoviral vector (eg A / C / N / 53). In another preferred embodiment, the tumor suppressor nucleic acid encapsulated in a liposome or complex with a lipid is provided (see, for example, Debs and Zhu (1993) WO 93/24640, Mannino and Gould-Fogerite (1988) BioTechniques, 6 (7): 682-691, Rose, Patent North American number 5,279,833; Brigham (1991) WO 91/06309; and Felgner et al. , (1987) Proc. Natl. Acad. Sci. USA, 84: 7413-7414.
B) Topical treatment. In another embodiment, topical application of the tumor suppressor protein or of the tumor suppressor nucleic acid, in combination with a surgical intervention, is preferred. In this embodiment, the tumor suppressor, preferably in the form of an infectious vector, is applied to the surface of the wound cavity once the tumor is removed. Infectious particles will carry p53 to any residual tumor cell at the site of the wound, inducing its apoptosis (programmed cell death). This treatment will impact the patient's long-term survival and / or reduce the amount of normal tissue surrounding the tumor site that needs to be removed during surgery. The tumor suppressor is preferably converted to one of the many formulations known to those skilled in the art, suitable for topical application. Thus, for example, an infectious preparation of the p53 human tumor suppressor gene (e.g., A / C / N) is suspended. / 53) in a suitable vehicle (e.g., a petroleum jelly or other cream or ointment) that is effective to spread over the surface of the wound cavity. Alternatively, the tumor suppressor can be prepared in an aerosol vehicle to be applied as a spray to the interior of the wound cavity. In other embodiments, the tumor suppressor can be prepared in degradable materials (resorbents), such as for example resorbent sponges which can be packaged in the wound cavity and which release the tumor suppressor protein or the vector in a weather.
Preferred embodiments for applying the recombinant adenoviral vectors to certain defined topical areas, such as for example the cornea, the gastro-mtestinal apparatus, tumor resection sites, use solid carriers to sustain a longer incubation time, facilitating the once the viral infection. The carriers can be gauze or ointments soaked with the recombinant adenovirus solution. The virus can be applied through the gauze support to the cornea to achieve improved effects of the transgenes. The drained gauze can also be applied prophylactically to certain areas of excised tumors, in order to avoid recurrence. Ointments can be applied topically to areas of the gastrointestinal tract or topically to areas of the pancreas to effect therapy with tumor suppressor genes. Exemplary carriers in the form of ointments include oil-based Puralube or water-soluble KJ-Jelly. In a method eg emplificativo, sterile gauze cushions (5 x 5 cm) or strips can be soaked for flow tests, removable, in a solution of an adenoviral vector (for example 1 x 109 PN / ml) until having a total humidity. The cushions or strips are placed in layers above the target tissue and incubated at 37 ° C for 30 minutes. One skilled in the art will recognize that it is also possible to include other tissues, gelatins or ointments that can absorb water or mix with it. In addition, other excipients that can improve the transfer of the genes as described above can be added. SAW . Combination treatments with other chemotherapeutics.
A) Suppressors of tumors administered in combination with multiple chemotherapeutic combinations. It will be noted that the methods according to the present invention are not limited to the combination of a tumor suppressor with a single adjunctive anticancer agent. While the methods typically involve contacting a cell with a tumor suppressor (e.g. p53) and an adjunctive anticancer agent such as paclitaxel, the methods of this invention likewise cover contacting the cell with a combination of a tumor suppressor gene or polypeptide and two, or three, or a multiplicity of adjunctive anticancer agents and optionally other chemotherapeutic drugs. In addition, an expert will recognize that it is possible to use a chemotherapeutic agent or chemotherapeutic agents with proteins or genes, tumor suppressors, in the absence of an adjunctive anticancer agent or adjunctive anticancer agents. Many chemotherapeutic drugs are well known in the scientific and patent literature; Exemplary drugs that can be used in the methods of the invention include, but are not limited to: DNA-damaging agents (including DNA alkylating agents) for example, cisplatin, carboplatin (see for example, Duffull (1997) Clin. Pharmacokinet. , 33: 161-183); Droz (1996) Ann. Oncol. , 7: 997-1003), navelbine (vinorelbine), Asaley, AZQ, BCNU, Busulfan, carboxyphthalate, CBDCA, CCNU, CHIP, chlorambucil, chlorozotocin, cisplatin, clomesone, cyanomorpholino-doxorubicin, cyclodison, cytoxan, dianhydrogalactitol, fluorodopan, hepsulfam, hicantone, melphalan, methyl NCCU, mitomicma C, mitozolamide, nitrogen mustard, PCNU, piperazine alkylator, piperazinedione, pipobroman, pofiromycin, spirohydantoin mustard, teroxirone, tetraplatin, thio-tepa, triethylene-ammine, nitrogen mustard of uracil, Yoshi-864); topoisomerase I inhibitors (eg, topotecan hydrochloride, iritecan hydrochloride (CPT-11), camptothecin, camptothecin Na salt, aminocamptothecin, CPT-11 and other camptothecin derivatives); topoisomerase II inhibitors (doxorubicin, including doxorubicin encapsulated in liposomes (see U.S. Patent Nos. 5,013,556 and 5,213,804) amonafide, m-AMSA, antiramyol derivatives, pyrazoloacridine, bisanthrene HCL, daunorubicin, deoxidoxorubicin, mitoxantrone, menogaril, daunomycin of N, N-dibenzyl, oxantrazole, rubidazone, VM-26 and VP-16), - the RNA / DNA antimetabolites (for example L-alanosine, 5-azacytidine, 5-fluorouracil, acivicin, aminopterin, aminopterin derivatives, an antifol, Baker's soluble antifol, dicloralil lausone, brequinar, ftafur (pro-drug), 5,6-dihydro-5-azacitidine-methotrexate, methotrexate derivatives, N- (phosphonoacetyl) -L-aspartate (PALA), pyrazufurine, and trimetrexate), - and, the DNA antimetabolites (e.g., 3-HP, 2'-deoxy-5-fluorouridine, 5-HP, alpha-TGDR, aphidicolin glycinate, ara-C, 5-aza- 2'-deoxycytidine, beta-TGDR, cyclocytidine, guanazole, hydroxyurea, inosine, glycodialdehyde, macbecin II, pyrazoloimidazole, thioguanine, and thiopurine). Tumor suppressor nucleic acid and / or polypeptide can also be administered in combination with chemotherapeutic agents such as vincristine, temozolimide (see for example, U.S. Patent No. 5,260,291), and toremifene (see for example, U.S. Patent No. 4,6996,949 for more information on toremifene). Preclinical studies performed in relevant animal models have shown that p53 adenovirus combined with cisplatin, carboplatin, navelbine, doxorubicin, 5-fluorouracil, methotrexate, or etoposide, has inhibited cell proliferation more effectively than chemotherapy alone for the treatment of tumors: neck and neck cancers SSC-9, neck and neck cancers SSC-15, cervical and neck cancers SSC-25, ovarian cancer SK-OV-3, prostate cancer DU-145, breast cancer MDA-MB-468 as well as breast cancer tumor cells MDA-MB-231. In another embodiment, improved anti-tumor efficacy is observed with the use of a combination of three drugs of the p53 gene (expressed, for example, in an adenovirus vector), an adjunctive anti-cancer agent (for example paclitaxel), and an agent DNA harmster (for example cisplatin). The combination of p53, paclitaxel and cisplatin, as has been demonstrated, is effective in an ovarian tumor model. These data corroborate the combination of a p53 gene-based therapy with chemotherapy in clinical trials. These other chemotherapeutic drugs can be used in combination with the nucleic acid and / or polypeptide, tumor suppressor, in the presence of an adjunctive anticancer agent or in the absence thereof. The invention also contemplates the use of a radiation therapy combined with any of the suppressors of tumors described above or in combination with the tumor suppressors described above, and in addition in combination with an adjunctive anticancer agent. It will also be noted that any of these chemotherapeutics can be used individually in combination with a nucleic acid or polypeptide, tumor suppressor, according to the methods of this invention-When the tumor suppressor nucleic acid (for example p53) is administered in a adenoviral vector with an adjunctive anticancer agent (for example paclitaxel) and a DNA damaging agent (such as cisplatin, carboplatin or navelbine), the adenoviral vector is typically administered for 5 to 14 days with an amount of about 7.5 x 10 12 at about 7.5 x 1013 adenoviral particles per day. For example, a daily dose of about 7. 5 10 13 adenoviral particles in combination with carboplatin. In one embodiment, a daily dose of the order of 7.5 x 10 12 adenoviral particles can be used to be administered to the lung. In another embodiment, p53 is administered with topotecan. Typically, the DNA damaging agent will be administered at the recommended dose, see for example Phi sycian 's Desk Ref rence, 51a (Medical Economics, Montvale, NJ 1997). For example, carboplatin is administered to achieve an area under the curve (abbreviation in English AUC) of the order of 6 to 7.5 mg / ml / min. Protease inhibitors.
Protease inhibitors. In still another embodiment, this invention provides the possibility of the combined use of nucleic acids and / or tumor suppressor type polypeptides with protease inhibitors. Particularly preferred protease inhibitors include, but are not limited to, inhibitors of collagenase, metal inhibitors or matrix proteinase (MMP), (see for example Chambers (1997) J. Na tl. Cancer Ins t 89: 1260-1270 ). In a preferred embodiment, the methods comprise administering, concurrently or sequentially, an effective amount of a protease inhibitor and an effective amount of a tumor suppressor polypeptide and / or nucleic acid. Examples of the compounds that are protease inhibitors are well known in the scientific and patent literature. Immunomodulators. The tumor suppressor-like proteins and nucleic acids according to this invention can be used in combination with immunomodulators, in which the latter regulate an immune response directed against hyperproliferative or cancerous cells (for example an immune response directed against an antigen specific to a tumor), upstream, or also downstream, an immune response directed against the tumor suppressor protein, the tumor suppressor nucleic acid, the tumor suppressor vector, tumors (for example an anti-adenoviral reaction) and / or a combined chemotherapeutic. Thus for example this invention provides the possibility of the sequential or concurrent, combined administration of an effective amount of a tumor suppressor nucleic acid and / or tumor suppressor polypeptides, with an effective amount of an immunomodulator. Immunomodulators include, but are not limited to, cytokines such as IL-2, IL-4, IL-10 (US 5,231, 012, Lalani (1997) Ann. All ergy As thma Immunol., 79: 469-483; Geissler (1996) Curr. Opinion He tol tol 3: 203-208, IL-12 (see for example Branson (1996) Human Gene Ther.1: 1995-2002) and gamma-interferon Immunomodulators that function as immunosuppressants can be used to mitigate an immune response targeted against the therapeutic (e.g., tumor suppressor protein or a nucleic acid of the same type or an adjunctive anticancer agent, etc.) Immunosuppressants are well known to those skilled in the art. Suitable immunosuppressants include, but are not limited to, cyclophosphamide, dexamethasone, cyclosporin, FK506 (tacrolimus) (Lochmuller (1996) Gene Therapy 3: 706-716), IL-10 and the like .. Antibodies against cell surface receptors that modulate the immune response, They can be used equally. For example, antibodies that block the binding of ligations to cellular receptors on B cells, T cells, NK cells, macrophages, and tumor cells can be used for this purpose. For examples of this strategy see for example Yang (1996) Gene Therapy 3: 412-420; Lei (1996) Human Gene Therapy 7: 2273-2279; Yang (1996) Science 275: 1862-1867. VII. The therapeutic cases. In another embodiment, this invention provides the possibility of using therapeutic kits. These kits include, but are not limited to, the tumor suppressor-like nucleic acid or polypeptide, or a pharmaceutical composition thereof. The kits may also include an adjunctive anti-cancer agent or a pharmaceutical composition thereof. The different compositions can be supplied in separate containers for individual administration or also to be combined before administration. As an alternative it is possible to supply the different compositions in a single container. The kits may also include various devices, buffers, reagents for assays and the like to bring the methods of this invention into practice. They may also contain the instructional materials kits to teach the use of the case in the different methods of this invention (for example in the treatment of tumors, in the prophylaxis and / or treatment of metastases and the like). The kit may optionally include 1 or more immunomodulators (eg, immunosuppressants). Particularly preferred immunomodulators include any of the immunomodulators described herein. VIII. Cells containing nucleic acids or polypeptides - of the tumor suppressor type, heterotriose, as well as other agents. The present invention also provides a prokaryotic or eukaryotic host cell, transfected or otherwise treated, for example an animal cell (e.g., a mammalian cell) containing a heterologous tumor suppressor nucleic acid and / or a polypeptide. tumor suppressor of the same type. The cell may optionally and additionally contain an adjunctive anticancer agent, for example paclitaxel or another agent that affects microtubules. Suitable prokaryotic cells include, without limitation, bacterial cells such as E cells. col i. Appropriate animal cells preferably include mammalian cells and most preferably human cells. The host cells include, without limitation, any cell of mammal and more preferably any neoplastic or tumor cell as any of the cells described herein. The transfected host cells described herein are useful as compositions for diagnostic or therapeutic purposes. When used pharmaceutically, they can be combined with different pharmaceutically acceptable carriers, as described ab for therapy with exotic genes. The cells can be administered therapeutically or prophylactically in effective amounts as described in detail ab In a diagnostic context the cells can be used for teaching purposes or other reference purposes, providing suitable models for the identification of the cells thus transfected and / or treated. IX. Efficacy and clinical therapy of p53 adenovirus genes. Adenovirus-mediated p53 gene therapy is currently undergoing phase I / II clinical trials in different countries. The pharmaceutical composition used in these clinical assays includes an adenovirus expressing p53, wild-type, as an example of the invention (rAd / p53) which consists of a replication-deficient type 5 adenovirus vector, which expresses the suppressor gene of human tumors under the control of the cytomegalovirus promoter ("rAd5 / p53") as described in this text (Wills (1994) supra). Regional administration. Ovarian cancer limited to the abdominal cavity is one of the clinical scenarios in which p53 gene therapy should be considered, regionally, ie intraperitoneally, as a preferred treatment plan. EXAMPLES The following examples are offered to illustrate, without limitation, the invention claimed. Example 1 Combination therapy with p53 and Taxol® The invention provides combined administration of a nucleic acid expressing a tumor suppressor polypeptide and paclitaxel in the treatment of neoplasms. The following example presents details about the ability of an adenovirus that expresses p53 according to the invention in combination with Taxol® to treat neoplasms and that this combination therapy was more effective in killing tumor cells than either agent alone. Combined therapy in vi tro. The cells were subjected to 1 of 3 treatment regimens: in treatment 1 the cells were pretreated with Taxol® 24 hours before being exposed to the p53 adenovirus construction material: A / C / N / 53. In treatment 2, the cells were pretreated with the p53 adenovirus construction and then the contacting with Taxol® was established. In treatment 3 the cells were contacted simultaneously with both Taxol® and adenovirus p53. Thus Ad p53 and Taxol® can be administered within the same twenty-four (24) hour period or simultaneously. Approximately 1.5 x 104 cells in a culture medium (SCC-9 and SCC-25 head and neck cell lines and SCC-25 in a 1: 1 mixture of DMEM + Ham's F12 medium with 0.4 μg / ml cortisol and 10% FBS and 1% non-essential amino acids, with prostate DU-145 cells and SK-OV-3 ovarian cells in the Eagles essential medium plus 10% FBS) were added to each well in a microtiter plate of 96 wells the material was cultivated for about 4 hours at 37 ° C and with 5% C02. The drug (Taxol®), p53 adenovirus or the appropriate vehicle or buffer was added to each well. Since paclitaxel is not soluble in water the drug was dissolved in ethyl alcohol before administration. The cells were then cultured overnight at 37 ° C and 5% C02. P53 adenovirus was administered in a 20 mM phosphate buffer of NaH2 P04, pH 8.0, 130 mM NaCl, 2 mM MgCl2, 2% of sucrose). Then cell death was quantified according to the method of Mosmann (1983) -J. Immunol. Meth. , 65: 55-63. In short terms, about 25 μl of 5 mg / ml of vital dye MTT [3- (4,5-dimethylthiazol-2-yl) -2,5-diphenyltetrazolium bromide] was added to each well and the material was allowed to incubate. for 3 to 4 hours at 37 ° C and with 5% C02. Then 100 μl of 10% SDS detergent was added to each well and allowed to incubate overnight at 37 ° C and 5% C02. The signal was quantified in each well using a microtiter plate reader from Molecular devices (molecular devices) (Thermo-Max). The particular cell lines used and the results obtained from this assay are listed in Table 1. Table 1: In Vitro evaluation of the adjunctive anticancer agent Taxol® combined with nucleic acid suppressor of tumors. Treatment In general p53 adenovirus was more effective when it was added later or simultaneously with Taxol® than when it was first added. These results suggest a synergistic interaction between A / C / N / 53 and Taxol®. Analysis in isobologram establishes synergistic effect. SK-OV-3 ovarian tumor cells (p53 zero) were treated with combinations of Taxol® and p53 adenovirus (A / C / N / 53) as illustrated in Table 2. Dosing was carried out in the manner described above. Cell death was quantified at day 3 using the MTT assay as described above. In addition, a curve of dose response for Ad p53 alone (using the doses listed in Table 2) (after exposure of the cells for 2 days to the drug) and a dose response curve for Taxol® was only carried out using the doses mentioned above (3-day cell exposure to the drug). Table 2. Subjected to treatments with combined Taxol® and Ad p53 (A / C / N / 53) - Figure 1 illustrates the inhibition of cell proliferation (as compared to the buffer control) as a function of the treatment. In general, the increasing doses of either Taxol® or p53 decreased the rate of cell proliferation, whereas the combination of p53 and Taxol® had a greater effect than that of any drug. Figure 2 illustrates an isobologram analysis of these data using the Isobolo method as reviewed by Berenbaum (1989) Pharmacol. Rev. 93-141. A synergism was observed between Taxol® and p53 (A / C / N / 53) when cells were treated with Taxol® 24 hours before treatment with p53 (A / C / N / 53). In Figure 2 the straight line (isobolo) for ED50 represents the effects on the cell proliferation that would be expected if it were merely additive or supplementary treatment with the 2 drugs. In fact, the effects observed fall on the lower left of the isobolic line, which indicates that lower concentrations were needed than expected for each drug and that a synergistic interaction occurred. Example 2 Gene therapy, mediated with p53 adenovirus against metastasis. The invention provides the possibility of administering the nucleic acid expressing a tumor suppressor polypeptide in the treatment of metastasis. The following gives detailed data on the ability of an adenovirus expressing p53 according to the invention to infect various tissues in the body and to treat metastases. SCID mice, females (homozygous mice for the SCID mutation lack both T and B cells due to an effect on recombination V (D) J) received injections with 5 x 10 6 MDA-MB-231 mammary carcinoma cells in their pads mammary fat. After the primary tumors were well established and they had time to metastasize to the lungs, the primary tumors were surgically removed (the day 11) . Mice were treated with A / C / N / 53 intravenously or with a control buffer on days 23, 30, 37, 44 (IqW) with a control buffer or with A / C / N / 53 (a p53 in a adenovirus) in 4 x 108 UIC by injection. On day 49, the lungs were harvested, fixed, stained and examined under a microscope. The results are illustrated below in Table 3. Table 3. Inhibition of lung metastasis with MDA-MB-231 using A / C / N / 53.
Treatment with A / C / N / 53 under the number of metastases in affected mice. In a second experiment, 231 tumors were applied to the mammary fat cushions of scid or scid-beige mice, peritumoral injections A / C / N / 53. A total dose of 2-4 x 109 U.I.C .; administered in 10 injections, the number of mice with lung metastasis decreased, for 80% in the scid mice and in 60% in the scid-beige mice. Likewise, the number of metastasis by mouse in those mice that did not have any lung tumor. As indicated above, intravenous dosing with A / C / N / 53 also demonstrated efficacy against lung metastases in scid mice. These data indicate that a cancer gene therapy with A / C / N / 53 may influence the severity of metastatic disease in addition to decreasing the burden of primary tumors. In another experiment, female scid mice were injected with 5 x 106 MDA-MB-231 mammary tumor cells per mouse in the mammary fat pad on day 0. Primary tumors (mammary) were surgically excised on day 18. Mice were treated with intravenous injections of buffer, Ad of beta-gal or Ad p53 (A / C / N / 53) on days 21, 24, 32, 39 and 36. The viral dose per injection was 4 x 108 UIC (A / C / N / 53) (PN / UIC = 23.3) and 9.3 x 109 Ad particles of beta-gal (PN (U. I C. = 55.6, 1.7 x 108 U. I C.) Lungs and liver were harvested on day 51 and fixed in formalin.The tissue sections for lung tumors and liver damage were evaluated.The organs larger than 2 mice under buffer and 2 mice under Ad-beta-gal were frozen by evaporation system in order to achieve cryo-sectioning and an analysis of the activity of the B-galactosidase enzyme was carried out.
Table 4. Inhibition of lung metastasis MDA-MB-231 with the use of A / C / N / 53.
* Number of undervalued metastasis. Multiple tumors grew together in these lungs. The number of metastases per lung in the low buffer group and the low Ad group of beta-gal was not significantly different (p = 0.268, see Table 4). Treatment with Ad p53 markedly decreased the number of metastases per lung when compared with either the buffer group or the Ad beta-gal group (p <0.001 and p <0.002, respectively). In addition to the reduction of number of metastases there was also a marked reduction in the size of lung metastases in the Ad p53 group. In the control groups, the tissue sections of most of the lungs were occupied by more than 50% by neoplastic tissue and the individual tumors could no longer be recognized in large areas of the lungs. In contrast, lung metastases were low and easily distinguishable as individual tumors in most of the corresponding group Ad p53. Tissue distribution by adenovirus. Liver tissues had the highest number of infected cells (around 50%) and beta-galactosidase activity was intense. The lung showed scattered patches of infected cells distributed evenly throughout the tissue. The intestines and the stomach had a periodic infection of the cells in the soft outer wall of the muscle that surrounded the organs. There was also a beta-galactosidase activity in the microvili (tiny hairy appendages) per lumen. The soft outer muscular wall that surrounded the uterus had a periodic infection of cells similar to that seen in the intestines. Most of the stromal cells in the ovary were infected. The spleen had a scattered activity of beta-galactosidase in the components of the In the soft muscle of the organ there were very few infected cells (< 1%) inside the main thickness of the striated cardiac muscle- Casinus there were no infected cells in the primary tumors within the mammary fat pad nor in the underlying striated muscle. There were no infected cells in the kidney. Liver pathology All livers, in general terms, had a normal condition at necropsy. There was no obvious necrosis in any kidney. However, the mice treated with adenoviruses did indeed have hepatocellular abnormalities (which were not present in the buffer group), which included high numbers of cells in mitosis, cell inclusions and changes in size and shape of the hepatocytes. Example 3 Gene therapy with p53 adenovirus-mediated genes against human breast cancer xenografts. The invention provides for the treatment of different cancers by the administration of a nucleic acid expressing a tumor suppressor polypeptide. The following example presents the details about the ability of a p53 expressing the adenovirus of the invention to treat human breast cancer. The introduction of wild-type p53 in tumors with p53 zero or p53 mutant offers a novel strategy for controlling the growth of tumors. Casey (1991) Oncogene 6: 1791-1797, introduced wild-type p53 in breast cancer cells in vi tro through a plasmid DNA vector. The number of colonies of MDA-MB-468 (P53mut) and T47D (P53raut) that arise after plasmid transfection was reduced by 50% by wild-type p53. It is also noted that none of the resulting colonies expressed the wild-type p53 transfectant. In contrast, the number of MCF-7 colonies (P53wt) was not affected. Negrini (1994 Cancer Res. 54: 1818-1824, carried out a similar study using MDA-MB-231 cells.) Colony formation was reduced by 50% thanks to transfection with a plasmid containing the wild type p53 and none of the resulting colonies expressed wild-type p53. Paradoxically in this study, similar results were observed with MCF-7 cells In the study described in this example p53 adenovirus, replication deficient, recombinant, suppressed was tested in the region (p53 Ad; (A / C / N / 53) Wills (1994) supra) against 3 human breast cancer cell lines expressing the mutant p53, MDA-MB-231, MDA-MB-468 , and MDA-MB-435. MDA-MB-231 cells carry a mutation of Arg-a-Lys in the codon 280 of the p53 gene (Bartek (1990) Oncogene 5: 893-899). MDA-MB-468 cells carry a mutation of Arg-a-His at codon 273 (Id.). MDA-MB-435 cells carry a Gly-a-Glu mutation at codon 266 of the p53 gene (Lesoon-Wood (1995) Hum. Gene Ther 6: 395-405). Previous studies have shown high levels of wild-type p53 expression in tumor cells from the human breast, ovary, lung, colon-rectum, liver, brain and bladder, after being infected with p53 Ad viin (Wills (1994) supra., Harris et al. (1996) Cancer Gene Therapy 3: 121-130) . Expression of p53 mediated with adenovirus ultimately resulted in changes in cell morphology and the induction of apoptosis in tumor cells from p53 zero or mutant p53. The infection of 468 breast cancer cells by p53 Ad at 10 multiplicities of infection (m.d.i.) caused an almost 100% inhibition of DNA synthesis at 72 hours after infection. In addition, infection with p53 Ad. in vi tro inhibited the proliferation of MDA-MB-468 and MDA-MB-231 cells with ED50 3 + 2 and 12 + 10 multiplicities of infection, respectively. The proliferation of 3 other lines of breast carcinoma of p53-mutant was also inhibited at low concentrations of p53 Ad. The ED50 values were 16 + 4 m.d.i. for SK-BR-3 cells, 3 ± 3 m.d.i. for T-47D cells, and 2 + _2 m.d.i. for BT-549 cells. Infection of MDA-MB-468 and MDA-MB-231 cells with 30 m.d.i. of an equivalent, recombinant adenovirus that expressed beta-galactosidase (beta-gal) from E. coli in place of p53, resulted in 67% of positive MDA-MB-468 beta-gal cells and 34 to 66% of MDA cells -MB-231 beta-gal positive. By correlating the percentage of positive beta-gal cells with the anti-proliferative effects of p53 in a large panel of tumor cells with their altered p53, Harris et al. (supra) showed a strong positive correlation between the degree of inhibition induced by p53 and the degree of transduction by adenovirus. In contrast, cell lines expressing normal levels of wild type p53 were affected to a minimum by p53 transduction, independently of the rate of adenovirus transduction. Proliferation of MCF7 and HBL-100 cells, two human mammary cell lines containing wild-type p53, was relatively unaffected by p53 Ad concentrations greater than or equal to 99 m.d.i. in vi tro. In other words, an inhibition of growth of the MCF-7 and HBL-100 cells required p53 pAd concentrations at least 8 to 33 times higher than the ED50 values for the class 231 and 468 cells, respectively. Using a similar recombinant p53 Ad, Katayose (1995) Clin Cancer Res. 1889-897, demonstrated increased expression of p53 protein, decreased cell proliferation, and increased apoptotic cell death in class -231 cells transduced in vi tro. This study extends these results in vi tro with cells of class -468 and -231 to xeno-injures of breast cancer in vi vo. The efficacy of adenovirus-mediated p53 gene therapy is evaluated in another breast cancer cell line (MDA-MB-435) that is resistant to adenovirus transduction in vi tro. Materials and methods Cell lines and adenovirus infections in vitro. Human breast cancer cell lines MDA-MB-231 and -435 were obtained from ATCC (Rockville, Maryland, United States of America). Cells -231 were cultured in DMEM (Life Technologies, Grand Island, NY) with 10% fetal calf serum (FCS; Hyclone, Logan, Utah) at levels of 37 ° C and 5% C02. -468 cells were cultured in Leibovitz L-15 medium (Life Technologies) containing 10% FCS at 37 ° C. Cells -435 were cultured in the Leibovitz L-15 medium with 15% FCS and 10 μg / ml insulin bovine (Sigma Chem. Co., St. Louis, Missouri) at 37 ° C. The construction and propagation of recombinant adenoviruses (rAd) expressing wild-type human p53 and beta-galactosidase (beta-gal) of E. coli where the expression of the transgenes by the promoter of the human megalovirus cyto is directed, have already been described previously (Wills (1994) supra). Adenoviruses were administered in a phosphate buffer (20 mM NaH2P04, pH 8.0, 130 mM NaCl, 2 mM MgCl2, 2% sucrose). The cell infection unit U.I.C- is defined as cellular infectious units. The concentration of the infectious viral particles was determined by measuring the 293 positive cells to viral hexon proteins after an infection period of 48 hours (Huyghe (1995) supra). For studies of infection in vi tro with p53 Ad cells were placed at a density of l-5xl04 cells per well (well) in 12 cavity tissue culture dishes (Becton Dickinson, Lincoln Park, New Jersey, United States of America). The cells were transduced with 0, 10 or 50 m.d.i. (multiplicity of infection = U. I.C./ cells) of p53 Ad and the material was cultured for 72 hours as previously described (Wills (1994) supra). For the studies of infection with beta- gal Ad, ie the beta-gal adenoviruses, the cells were applied in a density of lxlO5 cells per well. The cells were transduced with 0, 10, 50 or 100 m.d.i. of beta-gal Ad. After 48 hours, the cells were fixed with 0.2% glutaraldehyde (Sigma Chemical Co.) and then the material was washed 3 times with PBS (Life Technologies). The cells to be tested were then subjected to 1 ml of a solution of X-Gal [1.3 mM MgCl2, 15 mM NaCl, 44 mM Hepes buffer, with a pH of 7.4, 3 mM potassium ferricyanide and 1 mg / ml of X-Gal in N, N-dimethylformamide (final concentration of 10%)]. The X-Gal had been purchased from Boehringer Mannheim Corp., Indianapolis, Indiana. All other chemicals were purchased from Sigma. To determine the percentage of transduced cells, 5 fields were counted under a microscope from each culture well and the average percentage expressing beta-galactosidase was calculated for 3 wells with each multiplicity of infection (m.d.i.). In vivo adenovirus treatment Nude, female, and nude mice were purchased from Charles River Laboratories (Wilmington, Massachusetts, United States of America). All mice were kept in a facility with a HFV barrier and all procedures were performed on the animals according to the rules set forth in guide N.I.H. Guide for the Care and Use of Laboratory Animáis (Guide of N.I.H. for the care and Use of Laboratory Animals). The tumor cells were injected either subcutaneously or within the mammary fat pad. Inoculations of the cells were as follows: 5 x 10 cells of type -231 / mouse, lxlO7 cells MDA-MB-468 per mouse or 1 x 107 cells MDA-MB-435 per mouse. Tumors were allowed to grow in vivo for a period of 10 to 11 days before starting dosing except for a -468-type experiment in which the tumors grew for 33 days before starting treatment. The volume of the tumors was calculated, the product of the measurements in 3 dimensions. Tumor volumes for the different treatment groups, each day, were compared using the Student's test using the Statview II software (Abacus Concepts Berkeley, California). Inhibitions were calculated on a percentage basis for the groups dosed on days 0-4 and 7-11 using significant values (p <0.05) from day 14 at the end of the study. The specific effects of p53 were distinguished from the effects of the adenovirus vectors by subtracting the inhibition of tumor growth on average caused by Ad beta-gal from the inhibition of growth caused by Ad p53. All virus injections were peri / intra-tumoral. In general, two courses of five days each of tumor therapy (that is 5 injections) to each mouse, separated by a "rest period" of 2 days. In some cases this dosing regimen was extended for more than two weeks and / or the vehicle of the buffer was replaced for some virus, for certain injections. The growth curves of the tumors represent the average volume of the tumors - + s.e.m., that is, with the usual deviations. Histology and immunohistochemistry apoptag Tissue samples were fixed in 10% buffered formalin and the material was processed overnight in a Miles VIP tissue processor and the material was embedded in paraffin. Woven sections of 5 microns were cut with a microtome apparatus (microtome) from Leitz. The slices were stained with Harris' hematoxylin routinely and with an eosin stain (Luna et al., 1968), Manual of Hierarchies, Staining Methods of the Armed Forces Ins ti tu te of Pa thol ogy, New York: McGraw Hill Book Co.). Apoptosis detection kits, "Apoptag in situ" from Oncor (Gaithersburg, Maryland, United States of America) were purchased. Samples were tested according to the instructions contained in the kits. In short, the material was scattered, the rehydrated sections of the tissue with the enzyme Oncor Protein Digesting Enzyme, the material was incubated with TdT, and the disclosure was made using an avidin-peroxidase kit (rabbit IgG Sigma Chem. Co. EXTRA-3) and DAB (Vector Lab. KS4100). The slices were counterstained with methyl green. Assay with beta-galactosidase. The tumors were embedded in TBS (Triangle Biomedical Sciences, Durham, North Carolina, United States of America) and the material was frozen instantaneously in a bath of dry ice and 2-methylbutane. Frozen tissue sections (8 μm thick) were fixed in 0.5% gluteraldehyde at 4 ° C for 5 minutes and then assayed for beta-gal expression as described above. FACS Integri analysis. The cells were suspended by treatment with 0.02% EDTA, turned into granules and then washed twice with PBS. The cells were resuspended at a concentration of lx106 cells / ml and the material was incubated with primary antibodies (final concentration I: 250 / ml) at 4 ° C for 1 hour. Suspensions of the cells were washed 2 times with PBS to remove excess primary antibody. The cells were then incubated with the conjugated rabbit anti-mouse FITC-conjugated antibody (final concentration I: 250 / ml, Zymed) at 4 ° C for 1 hour. hour. The cells were washed as before with PBS and analyzed immediately. Fluorescence was measured with a FACS Vantage flow cytometer (Becton Dickinson, Mountain View, California, United States of America). The dispersion was simultaneously determined, that is, the lateral and forward scattering simultaneously and all the data were collected with a Hewlett Packard computer equipped with a FACS research software (Becton Dickenson). Potential primary antibodies were obtained to detect Integrin receptors from the following suppliers: anti-alfav (12084-018, Gibco BRL); anti-beta3 (550036, Becton Dickenson); anti-alfavbeta3 (MAP1976), Chemicon); anti-betai, (550034, Becton Dickenson); and anti-alphav beta5 (MAB 1961, Chemicon). Adenovirus Results Efficiency of transduction of and inhibition of growth by p53 in vi tro. Cells of type -231 and -468 were both transduced to a high extent in vi tro with a multiplicity of infection value (m.d.i.) of 10. Cells were rarely transduced -435, not even 100 m.d.i. for cells -231, 8% (10 m.d.i.), 46% (50 m.d.i.) and 62% (100 m.d.i.) of the cells were transduced by Ad beta-gal. For type 468 cells, transduced 78% (10 m.d.i.), 84% (50 m.d.i.) and 97% (100 m.d.i.) of the cells by Ad beta-gal. For 435 cells, 0.5% (10 m.d.i.), 1% (50 m.d.i.) and 1.3% (100 m.d.i.) were transduced by Ad beta-gal. The infection with 50 m.d.i. of Ad p53 resulted in an almost complete death of the cells in the cultures of 231 and 468 cells. In contrast Ad p53 had no detectable effect on the growth of the 435 cells. The efficacy of Ad p53 against xeno-injections of human breast cancer. Adenovirus-mediated p53 gene therapy was highly effective against xeno-injections of -231 and -468 (Figures 3a and 3b) in experiment with the 231 cells, a mouse in the Ad beta group remained tumor-free at the end of the study. -gal and 3 mice in the Ad p53 group and all tumors showed a regression during treatment with Ad p53. Inhibition of tumor growth in the group of -231 showed an average of 86% (p <0.01). The component of growth inhibition of life to p53 showed an average of 37% while the specific inhibition to adenovirus showed an average of 49% (p <0.01). Inhibition of tumor growth in -468 averaged 74% (p <0.001). A mouse in the Ad p53 group was free of tumors at the end of the study and all tumors regressed during Ad p53 treatment.
The component of inhibition of life growth at p53 showed an average of 45% (p <0.001) while the specific inhibition of adenovirus showed an average of 28% (p < 0.05). No side effects were observed in any experiment. The ED50 values for the inhibition of tumor growth in groups -231 and -468 were 3 x 108 of infectious units of cells (U.I.C.) and 2 x 108 of U.I.C. respectively (figure 4). Tumors in group -435 were almost totally resistant to treatment with Ad p53 (Figure 3c). The inhibition of growth in the tumor groups of 435 treated with adenovirus was not significant. Figure 5 shows a comparison of the efficiency of two dosing regimens with Ad p53 against tumors of -231. All mice received 5 peritumoral injections per week. All mice treated with the therapeutic agent (Ad p53) received a total of 2.2 x 109 U.I.C. mouse per week. One group received a single bolus injection containing the week-long dosage of the adenovirus. The remaining 4 injections for the week consisted of the buffer vehicle (group IX). The other treated group received the same dose of Ad subdivided into 5 injections per week (5X group). This dosage regimen was administered during weeks 1 and 3 (day 0-4, 14-18). The Growth inhibition showed an average of 74% for the 5X group (p <0.01) and only 44% for the group IX (p <0.05 for the first 3 weeks of the study and was not significant after day 21). ). The first cycle of p53 gene therapy was more effective than the second cycle. After the first therapeutic cycle, 4 mice remained in the group IX, 5 mice in the 5X group and 1 mouse in the vehicle control group. A mouse from the 5XS group had a recurrence with a very small tumor by day 21. No other "cures" were observed after the second cycle of therapy. Figure 6 shows an experiment using type 468 tumors that were initially 4 times larger than the type 468 tumors shown in Figure 3b, treated with a 10-fold lower dose of adenovirus. A total dose of 2.2 x 108 U.I.C. of Ad p53 per mouse per week. One group received a single bolus virus injection, followed by 4 injections of buffer per week (group IX). The other treated group received the same viral dose subdivided into 5 injections per week (group 5X). These dosing schedules were administered for 6 weeks. The total dose of virus administered for 6 weeks was approximately equal to half the dose used in Figure 3. This dose regimen resulted in a cytostatic effect in the volume of tumors in mice treated with Ad p53 (p <0.05). The treatments given earlier in the study were found to be more effective than those given during the following weeks. A mouse within the 5X group was found to be tumor free by day 21. However, when the inhibition in tumor growth was compared in all the mice, the dosing regimen IX (60%) was mild but not significantly more effective than the 5x regime (55%). One week after the end of the dosage, the growth rate of tumors in the 5X group began to increase. One month after the start of the study, the vehicle control tumors began to show necrosis and the growth remained at the same level. In vivo infectivity after repeated exposure to adenovirus. At the end of the studies shown in Figures 5 and 6, some tumors were injected with Ad beta-gal. These tumors were harvested 24 hours later and sections of frozen tissue to be tested were subjected to look for the expression of beta-galactosidase. Tumors treated with Ad p53 for 2 or 6 weeks were still transduced by Ad beta-gal although the transduction was the lowest in the tumors corresponding to group 468, treated for 6 months. weeks with Ad p53, 5 times a week. The sections from only one of the three tumors of group 468, injected into the 5X group, had cells expressing a beta-galactosidase. Induction of apoptosis in vivo by Ad p53 The xeno-injures of breast cancer MDA-MB-231 and MDA-MB-468 in nude mice were injected with 1-5 x 108 U.I.C. of Ad p53 or buffer 48 to 72 hours before harvesting. The induction of an apoptosis by Ad p53 was assayed using an immunohistochemistry of Apoptag in the sections of the tissues. The tumors injected with Ad p53 had areas of broad apoptosis throughout the needle section of the tumors injected intratumorally and around the outer edges of those tumors injected peritumorally. In contrast, tumors injected with buffer had only a few scattered apoptotic cells, as expected. The comparison of integrin expression in the cells of MDA-MB-231 and MDA-MB-435 A FACS analysis of integrin expression was carried out in the cells MDA-MB-231 and MDA-MB-435 a In order to determine if the low Ad transduction in the MDA-MB-435 cells was due to a deficiency in the alpha-integrins required for the internalization of Ad type 2, 3 and 4 (Wickham et al (1993) Cell, 73: 309-319; Wickham et al. (1994) J. Cell Biol., 127: 257-264; and Mathias et al. (1994) J. Virol. 68: 6811-6814). Both cell types expressed integrin portions of alphav beta3 alphav beta5 and betax integrin at approximately the same levels. The expression of alphav beta3 and beta3 classes were higher in MDA-MB-435 cells than in MDA-MB-231 cells. Discussion: When a total dose of 2.2 x 109 of U.I.C. of Ad p53 in 10 injections, inhibition of tumor growth was 74% for MDA-MB-468 tumors and 86% for MDA-MB-231 tumors but not significant for MDA-MB-435 tumors. In MDA-MB-468 tumors, 61% of the total response was specific to p53 whereas tumors MDA-MB-231 43% of the total response was specific to p53. The ability of Ad beta-gal to transduce the cells of MDA-MB-231, MDA-MB-468, and MDA-MB-435 in vi tro was generally a predictor of the results in vi vo. At the same virus concentrations, the -468 cells had a slightly higher transduction rate than the MDA-MB-231 cells while the MDA-MB-435 cells were resistant to adenovirus transduction. The results in the MDA-MB-435, in vi tro correlated with the very poor response in vi. Systemic treatment of nude mice that had tumors MDA-MB-435, with a liposome vector of p53, caused, as could be demonstrated, an inhibition in the growth of the tumors and in some cases a regression (Lesson-Wood et al (1995) Hum. Gene Ther., 6: 395-405). Treatment with the p53 liposome also reduced the number of lung metastases. These results demonstrated that the lack of tumor response MDA-MB-435 to a treatment with Ad p53 in this study was not due to the inability of p53 to inhibit the growth and metastasis of MDA-MB-435 tumors. Rather, these results suggest that it was the low transducer deficiency of the adenoviruses in the MDA-MB-435 cells that caused their lack of response to treatment with Ad p53. Alpha-alphav -integrins have been implicated as the cellular elements required for the efficient internalization of type 2, 3 and 4 adenoviruses (Wickham (1993) supra.; Wickham (1994) supra.; and Mathias (1994) supra. ) It is likely that alphav -integrins play the same role for Ad type 5. Wickham et al. (1994) supra. , observed a 5- to 10-fold greater internalization of recombinant type 5 adenovirus in cells transfected with alphav beta5 compared to cells lacking alphav expression or transfected with alphav beta3. The cells of group -293, of the human embryonic kidney used for the p53 production here used express integrins of alf v betai. but not alphav beta3 (Bodary (1990) J. "Biol. Chem. 265: 5938-5941.) It therefore seemed prudent to measure the expression in the -435 cells of the integrin subunits alphav betai, beta3 and alpha5. MDA-MB-231 and MDA-MB-435 cells expressed generally equivalent levels of the molecules corresponding to the integrin family.Therefore it can be said that the lack of Ad transduction in MDA-MB-435 cells should not be There is currently no literature on the identity of the cellular receptor required to bind Ad to the target cells, it is possible that the MDA-MB-435 cells are deficient in this receptor or that it is defective. another component required for viral ligation, internalization or gene expression.The continued efficacy of Ad p53 over multiple therapy cycles was examined in tumor models of type MDA-MB-231 and MDA-MB-468 It turns out that the efficacy decreased with continued dose, however this effect needs to be examined in greater detail. The prevailing theory that infection with adenovirus generates a rapid inflammatory and cytolytic response mediated by cytotoxic T cells in hosts with fully functional immune systems (reviewed by Wilson (1995) Na ture Med. 4: 887-889). This response of the T cells is stimulated by adenovirus antigens produced in host cells and presented in combination with portions of MHC on the surface of the cells. Neutralizing antibodies, specific to adenovirus-transduced cells, are later produced, within the immune response it is believed that they are responsible for the decreased ability to reinfect host cells with adenoviruses after the initial inoculations. The athymic nude mice used in these studies have a defective T-cell immune response to foreign antigens, however they are capable of generating an antibody response mediated by B cells (Boven (1991), The Nude Mouse in Oncology Research, Boston: CRC Press ). The production of neutralizing anti-adenovirus antibodies could explain the lower efficacy of a p53 adenovirus therapy (Ad p53) in the present studies, over time. Reduced immune function in nude mice and poor blood supply to interiors of tumor xeno-injections may explain the partial effectiveness of Ad p53 even after a 6-week dosing and the ability to infect a few tumor cells. with Ad beta-gal even after repeated injections with Ad p53.
In addition to breast cancer, a number of other cancers have been treated with recombinant adenoviruses expressing wild-type p53. These reports include models of cervical cancer (Hamada (1996) Cancer Res. 56: 3047-3054), prostate cancer (Eastham (1995) Cancer Res. 55: 5151-5155), head and neck cancer (Clayman (1995) Cancer Res. 55: 1-6), lung cancer ((Wills (1994) supra.), Ovarian cancer (13), glioblastoma (27, 28) and cancer in the colon-rectal tract (13, 29). Collectively, these data are corroborated by the clinical investigations that continue to be carried out and in which the effects of adenovirus-mediated p53 gene therapy are being evaluated. The present results demonstrate the ability of wild type p53 to restrict the growth of cancer cells in vivo in xenografts of breast cancer with expression of the p53 mutant. Current studies also confirm that it turns out that the adenovirus is an efficient delivery vehicle for the 53 when the target cells express the appropriate "receptor" or viral "receptors". Example 4 Further research on the treatment regimen to inhibit tumors. The invention provides a system for treating different cancers by administering an acid nucleic acid expressing a tumor suppressor polypeptide with the use of different dosage regimens. The following example details the increased efficacy of applying a divided dosage in the administration of a p53 expressing the adenovirus, according to the present invention. In order to investigate the effect of a single-dose regimen compared to divided doses administered over a period of time, "scid" type mice were treated with tumors MDA-MB-468 and MDA-MB-231 with a total dose per mouse of 1 x 109 IU Ad p53 (A / C / N / 53) administered in the form of a single bolus injection or divided the material into 3 or 5 injections administered once a day over the course of a week (as indicated by arrows in Figure 7). The results obtained with the MDA-MB-468 tumors were similar to those obtained with the MDA-MB-23 tumors and are illustrated in Figures 7a, 7b and 7c. In general terms, the divided dosages have inhibited tumor growth better than single bolus injections with a 5-injection dosage regimen and have a clear improvement compared to the 3-injection dosage regimen. Example 5 Dexamethasone suppresses growth inhibition tumor in association with the immune response of anti-adenovirus mediated with NK cells. It has been possible to demonstrate that a repeated administration of adenovirus vectors can induce an anti-adenovirus type immune response. To investigate whether the immunosuppressant properties of dexamethasone at low dose (Dex) can inhibit the anti-adenovirus immune response (e.g. the NK cell response), tumors were treated in "scid" type mice with the recombinant virus of the invention in the absence of dexamethasone and in the presence thereof. About 5 x 106 of MDA-MB-231 cells were injected into the mammary fat pads of female "scid" mice at day 0. On day 11 dexamethasone granules or placebo were implanted subcutaneously. The 5 mg granules were set up to release 83.3 μg dexamethasone per day, continuously, over a period of 60 days (Innovative Research of America, Sarasota, FL). All mice received a total of 10 peritumoral injections administered once a day, days 14-18 and 21-25 (0.1 ml per injection).
The total virus dose was 2 x 109 U. I C. / mouse Ad p53 (A / C / N / 53 or Ad of beta-galactosidase). The treatments were as indicated in Table 5. Table 5. Treatment of MDA-MB-231 tumors in mice lscid " Treatment with dexamethasone in low dose did not have a significant effect on the growth rate of MDA-MB-231 tumors in "scid" mice (p >; 0.05). No adverse side effects of dexamethasone were observed. Treatment of tumors with beta-galactosidase adenovirus caused significant inhibition of tumor growth with placebo control (p <0.001, day 21-30) but not in tumors treated with dexamethasone (p> 0.05, Figure 8) Tumors treated with placebo and Ad-beta-gal grew more slowly than tumors treated with placebo and dexamethasone (p <0.01, days 23-30). There was no significant, p53-specific inhibition of tumor growth in those tumors under placebo control (p> 0.05). In contrast, tumors treated with dexamethasone and Ad p53 in fact grew significantly more slowly than tumors treated with dexamethasone and Ad beta-gal (p <0.02, days 21-30) or placebo and Ad p53 ((p < 0.04, days 21-30) Thus, treatment with dexamethasone in low dose quenched the inhibition of tumor growth associated with the immune response of anti-adenovirus (for example the response to NK cells) in "scid" mice without effects The data also suggest that treatment with dexamethasone at a low dose may stimulate the expression of transgenes (for example p53) driven by the CMV promoter in recombinant adenoviruses, on the other hand it can be said that dexamethasone can increase the efficiency of the transduction of the adenovirus to increase the death of the tumor cells.The breast cancer model of type MDA-MB-231 was then used to evaluate the antitumor efficacy of Ad without p53 and with e in mice with different capacities to mount an immune response to foreign antigens. Naked mice were studied with non-functional T cells, "scid" mice with non-functional T and B cells but with high NK cells as well as "scid-beige" type mice with non-functional T, B and NK cells. To study the efficacy of rAd5 / p53 (described above) against xeno-inj ertos MDA-MD-231 was administered to nude mice a total dose of 2.2. x 109 of U.I.C. of Ad per mouse, divided into 10 injections on days 0 to 4 and 7 to 11. The SCID mice were given a total virus dose of 4x09 of UIC, divided into 10 doses administered on days 0-4 and 7-11 . The SCID-Beige mice were administered a total virus dose of 1.6xl09 of U.I.C., divided into 10 doses administered on days 0-4 and 7-11. All mice were treated with Ad p53, Ad type beta-gal or only with the vehicle. In nude mice (nonfunctional T cells) or "scid" mice (nonfunctional T and B cells, elevated NK cells) intratumoral dosing with the control Ad vector (without p53 insert) caused some growth inhibition of the tumors. The Ad expressing p53 (rAd5 / p53) showed a much improved antitumor efficacy compared to the Ad control. In contrast, in the "scid-beige" type mice (non-functional B, B and NK cells) the antitumor efficacy was all due to the expression of p53 without Ad vector component to inhibit tumor growth. These data demonstrate a previously unknown role for NK cells in the inhibition of tumor growth mediated by Ad. The data also suggest that the suppression of the immune system could abrogate some specific lateral effects of vectors, mediated, with NK cells. Example 6 Combined treatment of p53 adenovirus and chemotherapy The invention provides the combined administration of nucleic acid expressing a tumor suppressor polypeptide with chemotherapeutic agents in the treatment of neoplasms. The following example presents details on the ability of an adenovirus expressing p53 according to the invention in combination with different anticancer drugs, cisplatin, doxorubicin, 5-fluorouracil (5-FU), methotrexate and etoposide to treat neoplasms and that a combination therapy was more effective, ie had a synergistic result in killing the tumor cells than either agent alone. p53 administered with chemotherapeutic drugs in vi tro. Ci splatín, doxorubicina, 5 -f luorouracil (5-FU), meto trexa te and etoposida, in combination with p53. The effect of clinically relevant anticancer drugs was investigated in vi tro: cisplatin, doxorubicin, 5-fluorouracil (5-FU), methotrexate and etoposide, in combination with a tumor suppressor vector of the invention (A / C / N / 53 ). They submitted squamous cell carcinoma in head and neck SCC-9, squamous cell carcinoma in head and neck SCC-15, squamous cell carcinoma in head and neck SCC-25 and cells of prostate carcinoma DU-145 in 1 of 3 treatment regimens: in treatment 1 cells were previously treated with the anticancer chemotherapy 24 hours before exposure to a p53 adenovirus construct: A / C / N / 53. In treatment 2 the cells were pretreated with p53 adenovirus construction and then contacted with a chemotherapeutic agent with the anticancer chemotherapeutic. In treatment 3 the cells were simultaneously contacted with both the anticancer chemotherapeutic and the p53 adenovirus. All cell lines were obtained from ATCC (Rockville, MD). The tumor cells in the head and neck SCC-9, SCC-15, and SCC-25 (p53cero) were cultured in a 1: 1 mixture of DMEM and Ham F-12 (GIBCO / Life Technologies, Grand Island, NY) with a 10% fetal calf serum (FCS, Hyclone, Logan, Utah), 0.4 μg / ml hydrocortisone (Sigma Chem. Co., St. Louis, MO) and 1% amino acids Non-essential cells (GIBCO) at 37 ° C and 5% C02 were cultured human ovarian tumor cells SK-OV-3 .p53 ') and human prostate tumor cells DU-145 (p53mut in the Eagle MEM plus 10% FCS at 37 ° C and 5% C02 - Human mammary tumor cells MDA-MB-231 (p53mut) in DMEM (GE3C0) were cultured with 10% fetal calf serum (Hyclone) at 37 ° C and 5% C02. Human mammary tumor cells MDA-MB-468 (p53mut) were cultured in Leibovitz's L-15 medium (GEMBO) containing 10% FCS at 37 ° C without any C02 Mammary tumor cells MDA-MB-231 carry a Mutation of Arg-a-Lys at codon 280 of the p53 gene and expressing the p53 mutant (Bartek (1990) supra). Prostate tumor cells DU-145 carry two mutations of p53 or on different chromosomes, a mutation of Pro-a-Leu in codon 223 and a mutation Val-a-Phe in codon 274 (Isaacs (1991) Cancer Res. 51: 4716-4720). SK-OV-3 ovarian tumor cells are of the p53-0 type (Yaginuma (1992) Cancer Res. 52: 4196-4199). SCC-9 cells have a deletion between codons 274 and 285 which results in a mutation with frame shift; no immunoreactive p53 protein can be detected in the SCC-9 nuclei (Jung (1992) Cancer Res. 52: 6390-6393). The SCC-15 cells have an insertion of 5 base pairs between codons 224 and 225; they produce low levels of p53 mRNA but no detectable p53 protein (Min (1994) supra). SCC-25 cells have a loss of heterozygosity (LOH) on chromosome 17 and a deletion of 2 base pairs at codon 219 on the allele remainder; p53 mRNA is not detectable in SCC-25 cells and no immunoreactive p53 protein is observed in its nuclei (Caamano (1993) supra). Approximately 1.5 x 104 in culture medium (as described in Example 1) were added to each well in a 96-well microtiter plate and this material was cultured for about 4 hours at 37 ° and with 5% C02. The construction and propagation of wild-type human p53 and adenoviruses (Ad) of E. coli galactosidase (beta-gal) have been previously described (Wills (1994-) supra). The concentration of the infectious viral particles was determined by measuring the concentration of the 293 cells positive for the viral hexon protein after an infection period of 48 hours (Huyghe (1995) supra). The U.I.C. as cellular infectious units. Adenoviruses expressing p53 were administered in a phosphate buffer (20 mM NaH2 P04, pH 8.0, 130 mM NaCl, 2 mM MgCl2, 2% sucrose). The drug, the p53 adenovirus or the appropriate vehicle or buffer was added to each well. For studies in vitro with Ad p53, cells with a density of 1.5 x 104 cells per well were placed in a 96-well plate and cultured for 4 hours at 37 ° C and 5% C02. The drug, Ad p53 or the appropriate vehicle was added to each well and culture was continued. the cells during the night. The Ad p53 drug or the appropriate vehicle was then added to each well. Cell culture was continued for another 2 days. The death of cells was then quantified according to the MTT assay as described by Mosmann (1983) -J. Immuno 1. Meth. , 65: 55-63. Briefly, about 25 μl of 5 mg / ml of vital dye MTT ([3- (4,5-dimethylthiazolyl-2-yl) -2,5-diphenyltetrazolium bromide] was added to each well and allowed to incubate by 3 to 4 hours at 37 ° C and 5% C02, then 100 μl of 10% SDS detergent was added to each well and the material was allowed to incubate overnight at 37 ° C and 5% of C02 The signal in each well was quantified using a microtiter plate reader for molecular devices In all cases, the use of cisplatin (see table 6 for the summary results), doxorubicin (see Table 7 for a summary of results), 5-FU, methotrexate and etoposide, was more effective in the form of a combination therapy, to kill the tumor cells, than either agent alone. The combination of metotetraxate and Ad p53 was tested in a cell line. When the SCC-15 cells were treated with 0.7 μM of metotetraxate 24 hours before 5 m.d.i. of Ad p53, the combined antiproliferative effect of the two drugs it was only 5% higher than in the case of Ad p53 alone, although this difference was statistically significant (p <0.003). Pretreatment of DU-145 cells with 2.6 μM of etoposide 24 hours before 5 or 10 m.d.i. of Ad p53 resulted in a higher combined efficacy compared to any drug alone (p < 0.0001). When the SCC-15 cells were treated with 0.3 μM of etoposide 24 hours before 5 m.d.i. of Ad p53, the combined antiproliferative effect of the two drugs was only 5% higher than with Ad p53 alone, although this difference was also statistically significant (p <0.003). The combination of therapy with tumor suppressor genes and anti-neoplastic agents showed no antagonistic effects. In a second experiment, the effectiveness of the treatment of normal cells (MRC-9 cells) was compared with tumor cells (figure 9). In this experiment the growth to be tested was subjected as an incorporation of thymidine H instead of applying the MTT assay. Normal cells (MRC-9 cells from diploid fibroblasts) showed no more marked effects with the combination treatment. As expected. The effect of tumor suppressor alone was negligible in normal cells and highly significant in tumor cells. In contrast, the chemotherapeutic anticancer alone (cisplatin, doxorubicin, 5-FU, methotrexate and etoposide) was more effective in normal cells than in cancer cells (see figure 9). Table 6. Anti-proliferative effects of Ad p53 in combination with cisplatin. Greater combined efficiency? Table 7. Anti-proliferative effects of Ad p53 in combination with doxorubicin. Greater combined efficiency? The effects of doxorubicin and p53 on Human Hepatocellular carcinoma. The following example presents in detail the ability of an adenovirus expressing p53, according to the invention, in combination with doxorubicin to treat neoplasms and indicates that combination therapy has been more effective, ie the effect has been synergistic. to kill the tumor cells that the effect of any agent alone. The results demonstrate a synergistic interaction between the p53 expressing vector of the invention (ACN53) and doxorubicin. Doxorubicin (adriamycin) and p53 (ACN53, the recombinant adenovirus construct expressing the wild type human p53 transgene) were administered to the following cell lines: HLE, a human hepatocellular carcinoma cell line (Hsu (1993) Carcinogenesis 14: 987-992; Farshid (1992) J. Med.
I saw role. 38: 235-239; Dor (1975) Gann. 66: 385-392), with a mutated p53; HLF, a human hepatocellular carcinoma cell line with a mutated p53 (ibid); Hep 3B, a human hepatocellular carcinoma with p53 zero (Hasegawa (1995) In Vi tro Cell Dev Biol Anim. 31: 55-61); Hep G2 a human hepatocellular carcinoma with wild type p53 (ibid) and SK-HEP-1, an adenocarcinoma of human liver with wild-type p53 (Lee (1995) FEBS Lett 368: 348-352). The viability of the cells was measured with the live cell probe calcification AM (Probes molecule) (see for example Poole (1993) J. Cell Sci. 106: 685-691). The calcin of the AM substrate is split by cellular esterases to generate a fluorescent product. The cells were placed in 96-well plates (5 x 103 cells per well, allowed to adhere overnight, the material was treated in triplicate with dilutions of ACN53 and doxorubicin dilutions on day 0 in such a way that a dose response for doxorubicin treatment with each dose of ACN53.At day 3 the media was aspirated and the AM calcification in PBS was added to the cells.The fluorescent intensity of each well was determined using the fluorescent plate reader. Fluorescent value of the wells without cells was subtracted and the data expressed as the viability percentage (fluorescent intensity) compared to control wells - without treating. The ED50 values were used to generate projections in isobolograms to establish the interaction between ACN53 and doxorubicin. The isobologram analysis for each cell line showed a synergistic interaction between the p53 expressing vector of the invention (ACN53) and doxorubicin; this synergy was independent of the p53 status of the cell line. Note, on the other hand, that the ED50 value for ACN53 in the absence of doxorubicin is higher in the wild-type p53 cell lines than in the lines altered with p53. In another similar experiment, HLE cells were placed in 96-well plates (5 x 103 cells per well), allowed to adhere to the material overnight and then treated in triplicate with dilutions of ACN53 and doxorubicin dilutions in such a manner that a dose response curve was generated for the treatment with doxorubicin with each dose of ACN53. Three groups were used to test the effects in terms of the dosage order, on the interaction between ACN53 and doxorubicin.
The cells were incubated for 3 days after the first treatment. The medium was aspirated and the AM calcification in PBS was added to the cells. The fluorescence intensity of each well was determined using the fluorescent plate reader. The fluorescent value of the wells without cells was subtracted and the data expressed as the viability percentage (fluorescent intensity) as compared to the untreated control wells. The ED50 values were used to generate isobologram projections to evaluate the interaction between ACN53 and doxorubicin. Isobologram analysis for each dosing regimen showed a similar interaction, consistent with the synergy, between ACN53 and doxorubicin in the HLE cells, which was independent of the dosage order of the treatment. p53 with chemotherapeutic drugs in vivo The effect of clinically important anticancer drugs was further investigated in vivo: cisplatin, doxorubicin and 5-fluorouracil (5-FU) in combination with a tumor suppressor vector of the invention (A / C / N / 53). C mice were purchased. B .17 / lCR-scid from Taconic Farms (Germanotown, NY) or from Charles River Laboratories (Wilmington, M.) Nude mice were purchased in nu / nu from Charles River Laboratories.All mice were kept in a barrier facility to HFV and all animal procedures were carried out in accordance with the guidelines set forth in the NIH laboratory guide for the care and use of animals: "No. H Guide for the Care and Use of Labaratory You encourage. "Tumor volumes for different treatment groups, on each day, were compared using the Student's test using the Statview II software.
(Abacus Concepts, Berkeley, CA). Curves were constructed on the growth of the tumors to show the mean volume of the tumors +. of the "s.e.m." deviation.
There were usually 10 mice per group. Ovarian tumor model KS-OV-3: Intraperitoneal KS-OV-3 tumors were treated with intraperitoneal doses of vehicles, Ad p53, cisplatin or both drugs. The mice were given 6 injections of Ad p53 for a period of 2 weeks. The total dose of the virus was 1.5 x 109 U.I.C. (3.1 x 1010 viral particles). Efficacy of the cisplatin: Scid female mice were injected and with 5 x 106 cells of ovarian tumors SK-OV-3, by I.P., at day 0. The mice were dosed by the I.P. on days 6, 8, 10, 13, 15 and 17 (and Ad p53 only on day 17). Mice received 0.2 ml total volume (0.1 ml of cisplatin or cisplatin vehicle plus 0.1 ml of Ad or Ad p53 buffer). The dose of Ad p53 was 2.5 x 108 U. I. C. / mouse / day (5.2 x 109 viral particles). The dose of cisplatin was 2 mg / kg / day. Tumors were harvested and passed on day 20. Mice in a treatment group received 5 doses of cisplatin simultaneously with the first 5 doses of Ad p53. This dose of Ad p53 by the intraperitoneal route reduced the tumor load of the mice by only 17% by day 20 (p <; 0.01). However, when combined with cisplatin, Ad p53 caused a 38% decrease in tumor burden compared to cisplatin alone (p <0.0008). Mice treated with drug vehicles or Ad p53 alone had ascites, ie, accumulation of fluid with blood and nodules of invasive tumors in the diaphragm muscle. These symptoms were absent in mice treated with cisplatin alone or with cisplatin with Ad p53.
Efficacy of cisplatin with paclitaxel: Female scid mice were injected with 2.5 x 106 ovarian tumor cells SK-OV-3, via I.P. to day 0. The mice were dosed by I.P. on days 7, 9, 11, 16 and 18. The mice received a 0.3 ml total volume (0.1 ml cisplatin vehicle or cisplatin plus 0.1 ml paclitaxel vehicle or paclitaxel plus 0.1 ml Ad Buffer or Ad p53). The dose of Ad p53 was 2.5 x 108 of U. I. C. / mouse / day (5.2 x 109 viral particles). The dose of cisplatin was 0.5 mg / kg / day. The dose of paclitaxel was 1 mg / kg / day. Tumors were harvested and weighed on day 30; n = 7 = 10 mice per group. In this second study, ovarian tumors of type SK-OV-3 were treated with intraperitoneal doses of vehicles: Ad p53, cisplatin plus paclitaxel or all 3 drugs simultaneously. The combination of all 3 drugs reduced the tumor burden by 34% more than the combination of cisplatin plus paclitaxel, which shows the improved efficacy of the combination of the 3 drugs (p <0.0006). Prostate tumor model DU-45: Efficacy of the cisplatin: DU-45 intraperitoneal tumors were treated with intraperitoneal doses of vehicles, Ad p53, cisplatin or both drugs. Male scid mice were injected with 2.5 x 10 cells of DU-145, via I.P., on day 0. The mice were dosed by I.P. on days 7, 9, 11, 14 and 16. These mice received 0.2 ml of total volume (0.1 ml of cisplatin vehicle or cisplatin plus 0.1 ml of Ad or Ad p53 buffer). The dose of Ad p53 was 8.3 x 108 U. I. C. / mouse / day (2.9 x 1010 PN). The dose of cisplatin was 1 mg / kg / day. The tumors were harvested and weighed on day 22. The combination of Ad p53 and cisplatin had greatly improved the antitumor efficacy compared to the effect of any single drug (p <0.0004). Mammary tumor model MDA-MB-468; Efficacy of the cisplatin: MDA-MB-468 tumors established with vehicles were treated, with Ad p53, cisplatin or with both drugs. Female scid mice with 1 x 10 7 MDA-MB-468 cells were injected into the mammary fat pad, 11 days before the start of dosing at day 0. The dose of intraperitoneal cisplatin was 1 mg / kg / day. The intratumoral Ad p53 dose was 8.3 x 108 U. I. C. / mouse / day (2.9 x 1010 viral particles) administered simultaneously on days 0-4. Ad p53 had improved efficacy when combined with cisplatin (days 8-31, p <0.0004). Efficacy of doxorubin: In a second experiment, MDA-MB-468 tumors were treated with vehicles, Ad p53, doxorubicin or both drugs. Female mice, nude, were injected with 1 x 10 7 cells-MBDA-MB-468 subcutaneously 12 days before the start of dosing on day 0. The dose of intraperitoneal doxorubicin was 4 mg / kg / day on days 0, 2 , 7 and 9. The dose of intratumoral Ad p53 was 5 x 108 U. I. C. / mouse / day (1.03 x 1010 viral particles) on days 0-4 and 7-11. Ad p53 was more effective when administered in combination with doxorubicin (days 14-24-p <0.05). Head and neck tumor model SCC-15: Efficacy of 5-fluorouracil: Subcutaneous SCC-15 tumors were treated with vehicles, Ad p53, 5-fluorouracil (5-FU) or with both drugs. Scid mice were injected with 5 x 10 6 SCC-15 cells subcutaneously 7 days before the start of dosing on day 0. The intraperitoneal dose of 5-f luorouracil was 50 mg / kg / day in 40% hydroxypropellent-beta-cyclodextran (Cerestar Inc., Hammond, IN) administered by IP on days 0, 7 and 14 (once a week for 3 weeks). The dose of Ad p53 was 2 x 108 U. I. C. / mouse / day (4 x 109 viral particles) on days 0, 1, 7, 8, 14 and 15 (6 intratumoral injections per a period of 3 weeks). The 5-FU dose of 50 mg / kg was administered. The combination of Ad p53 and 5-FU resulted in greater antitumor activity than when any of the drugs were administered alone (p <; 0.04). p53 with FPT inhibitor The effect of a farnesyl protein transferase inhibitor in combination with a tumor suppressor vector designated as A / C / N / 53 was investigated in vi tro. The following examples present in detail the ability of a p53-expressing adenovirus according to the invention in combination with the FPT inhibitor designated "FPT39", as described in the international application WO 97/23478, filed on December 19, 1996, in which the FPT39 was designated as the compound "39.0"; see page 95 of said international application WO 97/23478, for treating neoplasms and that for the combination therapy against prostate tumor cells and mammary tumor cells, it was more effective in killing the tumor cells than any agent alone. Anti-proliferative efficacy of rAd5 / p53 and FPT39 against the ovarian tumor SK-OV-3. Methods: Human ovarian tumor cells SK-OV-3 (p53cero) were aliquoted into 96-well plates with a density of 250 cells per well.
Well in Eagle MEM plus 10% fetal bovine serum. The cells were then incubated at 37 CC and 5% C02 for 4 hours. FPT39 or drug vehicle was added to each well and cell culture is continued for 3 days. After 3 days the untreated cells were counted in some wells in order to calculate the amount of rAd5 / p53 that was missing to be added. Then the rAd5 / p53 or drug vehicle was added to each well and cell culture was continued for another 3 days. The proliferation of the cells was measured using the MTT assay. In brief terms, they added μl of 5 mg / ml vital dye MTT [3- (4,5-dimethylthiazol-2-yl) -2,5-diphenyltetrazolium bromide] to each well and the material was allowed to incubate for 3 to 4 hours at 37 ° C and with 5% C02. Then, 100 μl of 10% SDS detergent was added to each well and incubation was continued overnight. The fluorescence in each well was quantified using a microtiter plate reader from Molecular Devices. Cell proliferation data were analyzed using the statistical methodology called Thin Píate Spline (literal translation: Thin Plate Splice) of O'Connell and Wolfinger (1997) J. Comp. Graph. Sta t 6: 224-241. Results: rAd5 / p53 and FPT39 had an additional efficacy to inhibit cell growth. It was not shown in this experiment any presence of a synergism (p> 0.05) or antagonism (p> 0.05). Antiproliferative and synergistic efficacy of rAd5 / p53 and FPT39 (FPT inhibitor) against prostate tumor cells DU-145. Methods: Human prostate tumor cells DU-145 (p53mut.) Were treated with FPT39 or drug vehicle, and rAd5 / p53, and then the cultures of the cells were analyzed as described above for the human ovarian tumor cells SK -OV-3 The experiment was repeated twice. Result: .Experiment 1: rAd5 / p53 and FPT39 had an additional efficacy to inhibit cell growth. No presence or synergism (p> 0.05) or antagonism (p> 0.05) was demonstrated in this experiment. Experiment 2: rAd5 / p53 and FPT39 had a synergistic efficacy (P = 0.0192). These results demonstrate that rAd5 / p53 and FPT39 can interact and have a synergistic efficacy against the proliferation of prostate tumor cells. Anti-proliferative and synergistic efficacy of rAd5 / p53 and FPT39 (FPT inhibitor) against mammary tumor cells MDA-MB-231. Method: .Cell cells in human breast MDA-MB- 231 (p53mu.) Were treated with FPT39 or drug vehicle, and rAd5 / p53, and then the cell cultures were analyzed as described above for the human ovarian tumor cells SK-OV-3. The experiment was repeated twice. Result: Experiment 1: rAd5 / p53 and FPT39 had additional efficacy. No synergism was shown (p> 0.05) in this experiment. Experiment 2: rAd5 / p53 and FPT39 had additional efficacy on most of the response surfaces. However, the synergism was evident in the isolates greater than or equal to 70 (that is, less than 30% of the cells were killed, p = 0.0001). These results demonstrate that rAd5 / p53 can interact and have a synergistic efficacy against the proliferation of human breast cancer cells. E? 7 Profile of the Immune Response in Patients with Metastatic Hepatic Carcinomas Treated with the Vector Adenovirus Carrying p53 The invention provides the combined administration in vi ve of nucleic acid expressing p53 and other chemotherapeutic agents in the treatment of neoplasms. The following example presents a detailed summary of the capacity of the adenovirus that expresses p53 according to the invention to increase the levels of lymphocytes that kill tumors and that are found within human liver carcinoma. The purpose of this study was to characterize the genotypes and phenotypes of tumor infiltrating lymphocytes (TIL) in metastatic liver carcinomas from the colon that harbored p53 mutations (for discussions of these tumor infiltrating lymphocytes, TIL). , see for example Wang (1997) Mol. Med. 3: 716-731; Marrogi (1997) In t. J. Cancer 74: 492-501). A total of 16 patients were treated in a dose-escalation form of 109-1011 particles, by channeling into the hepatic artery with an adenovirus vector carrying the p53 wild-type gene. A total of 4 biopsies were obtained from each patient 3 to 7 days after administering the adenoviral vector. Immunohistochemical analyzes were carried out on frozen tissues obtained from normal liver and interface sites between tumor and host tissue. A computer-assisted image analysis was carried out to quantify the immunoreactivity for the following monoclonal antibodies: CD3, CD4, CD8, CD25, CD56, CD56, HLA-DR, IFN-gamma, TNF-alpha and IL-2. . An increase in the population of TIL (CD3 + Y CD4 +) lymphomas was observed with a maximum at the level of 7.5 x 1010 particles. TO At higher doses, a decrease in the CD3 + and CD4 + population was observed. An inverse correlation was observed for CD8 + cells. At the maximum dose (2.5 x 1011) an increase in the CD3 + CD4 + and CD8 + population was observed in the tumor compared to the normal level. These results demonstrate that the provision of high doses of adenovirus particles results in an increased population of TIL lymphocytes, ie of CD4 + and CD8 + - It is understood that the examples and embodiments described herein are for illustrative purposes only-- and that various modifications or changes in light of the foregoing will occur to those skilled in the art, modifications and changes that must be included within the spirit and scope of this application and the framework of the appended claims. All the publications, patents and patent applications cited here must be included in this text as a reference material, for all purposes.

Claims (18)

  1. CLAIMS 1. A method for treating cancer in mammals or hyperproliferative cells, which comprises placing the cancer cells or hyperproliferative cells in contact with a tumor suppressor nucleic acid and contacting the cells equally with an agent that affects the cells. microtubules or a polyprenyl-protein transferase inhibitor, wherein the tumor suppressor nucleic acid is a nucleic acid encoding a tumor suppressor protein comprising a wild type p53 protein or a retinoblastoma protein (RB).
  2. 2. The method according to claim 1 wherein the agent that affects the microtubules is paclitaxel, Taxol®, Taxotere® or a derivative of paclitaxel.
  3. 3. The method according to claim 1 wherein the polyperenyl protein transferase inhibitor is a farnesyl protein transferase inhibitor (FPT) or a transferase inhibitor of geranylgeranyl-protein.
  4. 4. The method according to claim 1 wherein the farnesyl protein transferase inhibitor (FPT) is FPT39, a mimetic compound of a farnesylated peptide, a tricyclic ring benzocycloheptapyridine or a farnesyl derivative.
  5. 5. The method according to claim 1 which further comprising putting the cell in contact with a chemotherapeutic agent.
  6. 6. The method according to claim 1 wherein the nucleic acid is delivered by a vector selected from the group consisting of a plasmid of naked DNA, a plasmid within a liposome, a plasmid formed in complex with a lipid, a viral vector, a AAV vector and a recombinant adenoviral vector.
  7. 7. The method according to claim 6 wherein the vector is A / C / N / 53. The method according to claim 1 wherein: the tumor suppressor protein or the tumor suppressor nucleic acid is administered in a total dose ranging from about 1 x 109 to about 7.5 x 10 15 adenovirus particles in a selected treatment regimen of the group consisting of: the total dose in a single dose, the total dose distributed between 5 days and administered daily, the total dose distributed between 15 days and administered daily, and the total dose distributed between 30 days and administered daily, and paclitaxel or the paclitaxel derivative is administered in a total dose ranging from 75 to 350 mg / m2 for 24 hours in a treatment regimen selected from the group consisting of an administration in a single dose, a dose administered daily on the day 1 and on day 2, a dose administered daily on day 1, day 2 and day 3, a daily dose for 15 days, a daily dose for 30 days, a daily continuous infusion for 15 days and a daily continuous infusion for 30 days. days . 9. A pharmacological composition comprising a tumor suppressor nucleic acid and a microtubule-affecting agent or a polyprenyl-protein transferase inhibitor, wherein the tumor suppressor nucleic acid is a nucleic acid encoding a protein suppressor of tumors comprising a wild type p53 protein or a retinoblastoma protein (RB). The pharmacological composition according to claim 9, wherein the microtubule-affecting agent paclitaxel, Taxol®, Taxotere® or a paclitaxel derivative And in that the polyprenyl protein transferase inhibitor is a farnesyl protein transferase inhibitor (FPT) or a transferase inhibitor of geranylgeranil-protein. The method according to claim 10 wherein the nucleic acid is delivered by a recombinant adenoviral vector comprising a partial or total deletion of a DNA IX protein and comprising a nucleic acid encoding a wild-type p53 protein. The method according to claim 11, wherein the deletion of the protein IX gene sequence extends from about 3500 bp from the 5 'viral terminals. up to approximately 4000 bp from the 5 'viral terminals. The method according to claim 12 further comprising the deletion of a non-essential DNA sequence in an early region 3 of the adenovirus. The method according to claim 11 which further comprises the deletion of a non-essential DNA sequence in the early region of adenovirus 4. 15. The method according to claim 11 further comprising a deletion of the DNA sequence designated as Ela and Elb. The method according to claim 10 wherein the recombinant adenoviral vector comprises the major late promoter of adenovirus type 2 or the human CMV promoter, the cDNA of the tripartite leader of type 2 adenovirus and a human p53 cDNA. 17. The method according to claim 16 wherein the vector is A / C / N / 53. 18. The method according to claim 2 wherein the agent that affects the microtubules is selected from the group composed of paclitaxel and Taxotere®-19. The method according to claim 18 in which the agent that affects the microtubules is Taxol®. The method according to claim 3, wherein the cells are first contacted with the tumor suppressor nucleic acid or the tumor suppressor protein and then contacted with paclitaxel or a derivative thereof. The method according to claim 3 wherein the cells are first contacted with paclitaxel or a paclitaxel derivative and with the tumor suppressor protein or the tumor suppressor nucleic acid. 23. The method according to claim 1 wherein the cells are neoplastic cells. The method according to claim 23, wherein the neoplastic cells comprise a cancer selected from the group consisting of: ovarian cancer, pancreatic cancer, non-small cell lung cancer, small cell lung cancer, hepatocarcinoma, melanoma, retinoblastoma , breast tumor, carcinoma in the colon-rectal tract, leukemia, lymphoma, brain tumor, cervical carcinoma, cervical carcinoma, sarcoma, prostate tumor, gallbladder tumor, reticuloendothelial tissue tumor, Wilm's tumor, astrocytoma, glioblastoma, neuroblastoma, osteosarcoma, kidney cancer and cancer of the head and neck. 25. The method according to claim 1 wherein the tumor suppressor protein or the tumor suppressor nucleic acid is dispersed in a pharmacologically acceptable excipient-26. The method according to claim 3 wherein the paclitaxel or the paclitaxel derivative is dispersed in a pharmacologically acceptable excipient. 27. The method according to claim 2 wherein the tumor suppressor protein or tumor suppressor nucleic acid as well as paclitaxel or the paclitaxel derivative are dispersed in a single composition. The method according to claim 1, wherein the contacting comprises injecting the tumor suppressor protein or the tumor suppressor nucleic acid into a tumor. 29. The method according to claim 1, wherein the contacting comprises the intra-arterial injection of the tumor suppressor protein or the tumor suppressor nucleic acid. 30. The method according to claim 29, wherein the contacting is selected from the group consisting of injection into the intra-hepatic arterial of the protein or nucleic acid, tumor suppressors, for the treatment of liver cancer and intraperitoneal administration of the protein or nucleic acid, tumor suppressors, for the treatment of ovarian cancer. The method according to claim 3, wherein the contacting comprises injecting the paclitaxel or paclitaxel derivative into a tumor. 32. The method according to claim 3, wherein the contacting comprises the intravenous injection of paclitaxel or paclitaxel derivative. 33. The method according to claim 1, wherein contacting comprises systemic, regional, local, topical, intraperitoneal administration in the intra-pleural cavity, oral, buccal, sublingual, intratracheal, transmucosal, in the bladder. , in the vesicle, vaginal, uterine, rectal or nasal. 34. The method according to claim 2 comprising bringing the cells into contact with A / C / N / 53 and paclitaxel. 35. The method according to claim 1 in which the contacting of the cells with a protein or a nucleic acid, tumor suppressors, comprises putting the cells in contact with the protein or the nucleic acid, tumor suppressors, in a multiplicity of treatments, each separated by at least 6 hours. 36. The method according to claim 1, which comprises at least 3 separate treatments for about 24 hours. 37. The method according to claim 1, wherein: the tumor suppressor protein or nucleic acid is administered in a total dose ranging from 1 x 109 to about 7.5 x 10 15 adenovirus particles in a treatment regimen selected from the group consisting of: the total dose in a single dose, the total dose distributed between 5 days and administered daily, the total dose distributed during 15 days and administered daily and the total dose distributed between 30 days and administered daily; and paclitaxel or the paclitaxel derivative is administered in a total dose ranging from about 75 to 350 mg / m2 for 24 hours in a treatment regimen selected from the compound group: administration in a single dose, a dose administered daily on days 1 and 2, a dose administered daily on days 1, 2 and 3, a daily dosage for 15 days, a daily dosage for 30 days, a daily continuous infusion for 15 days and a daily continuous infusion for 30 days. 38. The method according to claim 37 which is repeated for 2 or more cycles. 39. The method according to claim 38 wherein the 2 or more cycles are spaced by 3 or 4 weeks. 40. The method according to claim 38 which is repeated for 3 cycles. 41. A kit for the treatment of cancer in mammals or hyperproliferative cells, which comprises: a first container comprising a protein or a nucleic acid, of the tumor suppressor type, selected from the group consisting of the wild-type p53 protein or a nucleic acid, either a retinoblastoma protein (RB) or the corresponding nucleic acid, and a second container comprising at least one adjunctive anticancer agent. 42. The kit according to claim 41, wherein the tumor suppressor nucleic acid encodes a wild-type p53 protein. 43. The kit according to claim 41 wherein the adjunctive anti-cancer agent is paclitaxel or a paclitaxel derivative. 44. The kit according to claim 41 further comprising instructions describing the administration of both the tumor suppressor-type protein or nucleic acid, and for the adjunctive anti-cancer agent, in order to inhibit the growth or the proliferation of cells. 45. The kit according to claim 41, wherein the protein or nucleic acid, of the tumor suppressor type, is selected from the group consisting of p53, pllORB and p56RB. 46. The kit according to claim 41, wherein the first container contains a nucleic acid that is contained in a recombinant adenoviral vector. 47. The kit according to claim 46, wherein the nucleic acid is contained in a recombinant adenoviral vector comprising a partial or total deletion of a DNA IX protein and comprising a nucleic acid encoding a p53 protein. 48. The kit according to claim 47 wherein said oppression of the protein IX gene sequence extends from about 3500 bp for the 5 'viral terminals to about 4000 bp from the 5' viral terminals. 49. The kit according to claim 48 further comprising a deletion of the DNA sequence, designated Ela and Elb. 50. The kit according to claim 46 wherein the recombinant adenoviral vector comprises the adenovirus type 2 major late promoter or the human CMV promoter, the tripartite type 2 cDNA leader of the adenovirus as well as a human p53 cDNA. 51. The kit according to claim 46, wherein the vector is A / C / N / 53. 52. A pharmacological composition comprising a protein or a nucleic acid, both of the tumor suppressor type and at least one adjunct anticancer agent. 53. The composition according to claim 52 wherein the adjunctive anti-cancer agent is paclitaxel or a derivative of paclitaxel. 54. The composition according to claim 52 in which the protein or nucleic acid, both of the tumor suppressor type, is selected from the group consisting of a nucleic acid encoding a wild-type p53 protein, a nucleic acid encoding for a retinoblastoma protein (RB), a wild type p53 protein and a retinoblastoma protein (RB). 55. The composition according to claim 52 wherein the nucleic acid encodes a wild-type p53 protein. 56. The composition according to claim 52 wherein the nucleic acid codes for retinoblastoma pllORB or p56RB. 57. The composition according to claim 52 wherein the nucleic acid is contained in a vector recombinant adenoviral. 58. The composition according to claim 57 wherein the nucleic acid is contained in a recombinant adenoviral vector comprising a partial or total deletion of a DNA IX protein and comprising a nucleic acid encoding a p53 protein. 59. The composition according to claim 58 in which the aforementioned deletion of the protein IX gene sequence extends from about 3500 bp for the 5 'viral terminals to about 4000 bp of the 5' viral terminals. 60. The composition according to claim 59 further comprising a deletion of the DNA sequence designated Ela and Elb. 61. The composition according to claim 57 wherein the recombinant adenoviral vector comprises the adenovirus type 2 major late promoter or the human CMV promoter., the tripartite leader cDNA of adenovirus type 2 and a human p53 cDNA. 62. The composition according to claim 57 wherein the vector is A / C / N / 53. 63. The composition according to claim 53 wherein paclitaxel or a paclitaxel derivative is used. 64. A composition comprising a cancer of mammal or a hyperproliferative cell in which the cell contains a nucleic acid or a protein, of the tumor suppressor type, of an exogenous nature, thus paclitaxel or a paclitaxel derivative. 65. The composition according to claim 64 wherein the tumor suppressor nucleic acid is a nucleic acid encoding a tumor suppressor protein selected from the group consisting of wild-type p53 protein and a retinoblastoma protein (RB). 66. The composition according to claim 64 wherein the tumor suppressor nucleic acid codes for a wild-type p53 protein. 67. The composition according to claim 65 wherein the renoblastoma protein is pll0RB or p56RB. 68. The composition according to claim 64 wherein the cells are present in a mammal. 69. The composition according to claim 64 wherein the cells are neoplastic cells. 70. The composition according to claim 69 wherein the neoplastic cells comprise a cancer selected from the group consisting of an ovarian, pancreatic cancer, in the lung in non-small cells, in the lung in small cells, of hepatocarcinoma type, melanoma, retinoblastoma, breast tumor, carcinoma in the colon-rectal tract, leukemia, lymphoma, brain tumor, cervical carcinoma, sarcoma, prostate tumor, vesicle tumor, tumor of the reticuloendothelial tissues, Wilm's tumor, astrocytoma, glioblastoma, neuroblastoma, osteosarcoma, renal cancer and head and neck cancer. 71. A method for treating a metastatic cell comprising bringing the cell into contact with a nucleic acid or a polypeptide, both of which are tumor suppressors. 72. The method according to claim 71, wherein the contacting comprises the topical administration of a tumor suppressor nucleic acid to a surgical wound. 73. The method according to claim 71 wherein the cells are further contacted with at least one adjunctive anti-cancer agent. 74. The method according to claim 73 wherein the adjunctive anticancer cancer is paclitaxel. 75. The method according to claim 73 wherein the adjunctive anticancer agent is an agent that affects microtubules. 76. The method according to claim 71 which further comprises placing the cell in contact with a chemotherapeutic agent. 77. The method according to claim 76 wherein the chemotherapeutic agent is cisplatin, carboplatin or Navelbine
MXPA/A/1999/007571A 1997-02-18 1999-08-17 Combined tumor suppressor gene therapy and chemotherapy in the treatment of neoplasms MXPA99007571A (en)

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US801765 1991-11-29
US60/038065 1997-02-18
US801681 1997-02-18
US038065 1997-02-18
US801755 1997-02-18
US801285 1997-02-18
US047834 1997-05-28
US60/047834 1997-05-28

Publications (1)

Publication Number Publication Date
MXPA99007571A true MXPA99007571A (en) 2000-02-02

Family

ID=

Similar Documents

Publication Publication Date Title
US7157079B2 (en) Combined tumor suppressor gene therapy and chemotherapy in the treatment of neoplasms
AU737621B2 (en) Combined tumor suppressor gene therapy and chemotherapy in the treatment of neoplasms
US20080286239A1 (en) Combined tumor suppressor gene therapy and chemotherapy in the treatment of neoplasms
EP2351575B1 (en) Grs for use in treating a cancer
US7001770B1 (en) Calpain inhibitors and their applications
JP4764872B2 (en) Gene transfer system containing relaxin gene and pharmaceutical composition using relaxin
KR101163248B1 (en) Methods and compositions for treatment of interferon-resistant tumors
KR100739118B1 (en) 1 a novel use of msx1 or gene encoding the same
US20030199472A1 (en) Adenovirus-mediated therapy for uterine fibroids
MXPA99007571A (en) Combined tumor suppressor gene therapy and chemotherapy in the treatment of neoplasms
US20020082204A1 (en) Treatment of inflammation with p20
WO2004081030A2 (en) Peptides selectively lethal to malignant and transformed mammalian cells
US20010044420A1 (en) Combination use of gemcitabine and tumor suppressor gene therapy in the treatment of neoplasms
US7883888B2 (en) Peptides selectively lethal to malignant and transformed mammalian cells
US20090233848A1 (en) Pea15 as a Tumor Suppressor Gene
WO2000021575A2 (en) Calpain inhibitors and their applications
Florea Characterization of an immunocompetent mouse model for gene therapy in ovarian cancer
WO2000056351A2 (en) Combination use of gemcitabine and tumor suppressor gene therapy in the treatment of neoplasms