![]() Antioxidant enhancement of therapy for hyperproliferative conditions
专利摘要:
A method of increasing the cytotoxic activity of an anticancer agent, the method comprising administering an effective amount of an anticancer agent to a host exhibiting abnormal cell proliferation with an amount of antioxidant effective to increase cell death. In addition, the present invention relates to the treatment index of anticancer drugs administered for the treatment of fidelity of cells that reduce or abnormally proliferate the toxicity to anticancer drugs, which consists of administering an antioxidant agent before, simultaneously with and after administration of the anticancer agent. It includes a method of increasing. 公开号:KR20010020611A 申请号:KR1019997012630 申请日:1998-07-01 公开日:2001-03-15 发明作者:쉬너리레베카;뷰캄프알.다니엘;코페이로버트제이.;메드포드러셀엠.;와드신스키브라이언 申请人:아테로제닉스, 인코포레이티드; IPC主号:
专利说明:
ANTIOXIDANT ENHANCEMENT OF THERAPY FOR HYPERPROLIFERATIVE CONDITIONS} Various diseases are associated with hyperproliferative cells, from psoriasis to malignant tumors. In general, these diseases result from loss of control over the course of normal cell growth, differentiation or programmed apoptosis. Many abnormalities associated with these diseases, especially tumors, occur at the genetic stage. Anticancer agents (also known as cell death agents) are often used for the treatment of hyperproliferative diseases. Although chemotherapy is successful in many malignant diseases, it is mostly used to prolong the life or reduce the symptoms of patients with advanced disease. The two groups of drugs used for the treatment of hyperproliferative diseases are anti-metabolic and alkylating agents. Antimetabolites are subdivided back into folic acid, furin, and pyrimidine derivatives. In addition, several natural products or derivatives thereof have been used as mitosis inhibitors. These include vaccinia, alkaloids, and derivatives of podophyllotoxin. Efforts remain to understand the genetic basis of abnormal cell hyperproliferation that can be expressed in a variety of ways and to develop treatments for the successful treatment of these serious diseases. For about 40 years, the antimetabolic 5-fluorouracil (5-FU), and nucleosides containing these bases (eg, 5-fluoro-2'-deoxyuridine or FdUrd) have been It remains one of the 'standard' drugs effective against solid tumors. 5-Fluorouracil is mainly used for the treatment of colorectal, ovarian, kidney, breast, head, and neck tumors. 5-Fluoro-2'-deoxyuridine is used for the treatment of solid tumors, including advanced gastrointestinal adenocarcinomas, renal cell carcinoma, advanced uterine carcinoma, and squamous cell carcinoma of the head and neck. The clinical utility of fluoropyrimidines is limited because of the host-toxicity induced by the administration of fluuropyrimidine. Host-toxic expression of fluoropyrimidine mainly includes gastrointestinal epithelial ulcers, myelosuppression, less cardiac toxicity, hepatotoxicity and neurotoxicity. Tumor patient populations are not resistant to the treatment of 5-fluorouracil and 5-fluoro-2'-deoxyuridine. Intolerance to 5-fluorouracil initially contributes to the low activity or deficiency of the first enzyme in the degradation pathway of dihydrouracil dehydrogenase (DHUDase, EC1.3.1.2), ie 5-fluorouracil. However, not all intolerant patients exhibit reduced dihydrouracil dehydrogenase activity. Moreover, it was shown that tumors treated with fluoropyrimidine became resistant, ie developed resistance to these drugs. Colorectal cancer (CRC) is a multistep process that results from the accumulation of mutations in a colony of colon cells. Mutations in the p53 tumor suppressor gene are relatively slow but are common in hospitals of colorectal cancer and occur in more than 80% of late adenocarcinomas and carcinomas (Fearon, et al., FASEB J. 6, 2789 (1992); Srivastarva, et al., Contemp. Oncol. April 63 (192); Kline, et al., Cancer (Phila ,. 73, 28 (1994)). Conventional treatments for advanced disease, such as cell death chemotherapy and Gamma-radioactivity induces DNA damage in proliferating cells, although this is an undefined mechanism, but induces P53 and disrupts G1 cell cycle in turn, inhibiting cell proliferation and, in some cases, causing cell death. Thus, tumors lacking functional P53 are often resistant to such treatment (SC Fightetti et al., Cancer Res. 56, 689 (1996); JS Kovack et al., Proc. Natl. Acad, Sci. USA 93). , 1093 (1996), which suggests the development of treatment for advanced colorectal cancer that is independent of functional P53. Importance is emphasized. The single most effective chemotherapeutic agent for advanced colorectal cancer so far is still 5-FU. 5-Fluorodeoxyuridine-5-monophosphate (FdUMP), an active metabolite of 5-FU, inhibits enzyme activity by complexing with thymidylate synthase (TS) in the presence of reduced folate And the precursor of DNA synthesis is removed. 5-FU is also incorporated into RNA to alter the processing and function of RNA. It is not known how this relates to cellular lethality. Previous data suggest that although loss of function of p53 significantly reduces the efficacy of 5-FU (B. Cohen et al., Cancer (Phila.) 67, 1859 (1991); Advanced Cancer Meta-Analysis Project, J.). .clin.Oncol. 10, 896 (1992)), 5-FU uses p53-dependent and independent paths (Pritchard, et al., Pharmacol. Ther. 72, 149 (1996)). Given that there are no successful treatments for many diseases, we identify important biological pathways that mediate impairment of normal cell function, including programmed cell death (eg, apoptosis), and identify methods and compositions for the treatment of such diseases. It would be advantageous to sympathize. US Pat. Nos. 5,035,878 and 5,294,430 describe that dithiocarbamate can reverse the myelosuppression of bone marrow's blood producing function caused by anticancer agents. Accordingly, it is an object of the present invention to provide methods and compositions for the treatment of abnormal cell proliferative diseases, including benign and malignant tumors. Another object of the present invention is to provide a method and composition for the treatment of colon cancer. Another object of the present invention is to provide a method and composition for the treatment of solid tumors. Another object of the present invention is to provide a method and composition for the treatment of diffuse tumors. Summary of the Invention Antioxidants, including those specifically described herein, have been found to be useful in enhancing the efficacy of anticancer agents for the treatment of abnormal cell proliferation by inducing cell cycle disruption (G1, G2, S and M types). Thus, in one example, the present invention provides a method of enhancing the cytotoxic activity of an anticancer agent, comprising administering an effective amount of an anticancer agent to a patient exhibiting abnormal cell proliferation in combination with an amount of antioxidant effective to increase cell death. . In addition, it was found that without antioxidants, antioxidants inhibited enzymes that could terminate cell cycle disruption, leading to cell cycle disruption as well as allowing cells to remain in this interrupted state, possibly leading to cell death. Importantly, it has also been found that antioxidants not only improve cell death of anticancer agents against abnormal proliferative cells but also reduce cell death against normal cells. Thus, antioxidants increase the efficacy of anticancer agents and reduce their toxicity. Mitigating effects on normal cells are found in epithelial cells. In particular, as previously reported by Borch in US Pat. Nos. 5,035,878 and 5,294,430, it has been found that antioxidants have such effects on cells other than leukocytes. Accordingly, the present invention is directed to a method of increasing the therapeutic index of an anticancer agent administered for the treatment of congestive growth of abnormally proliferating cells consisting of administering an antioxidant agent before, with or after administration of the anticancer agent. And a method of reducing the toxicity of the administered anticancer agent for the treatment of hyperemic growth of abnormally proliferating cells consisting of administering an antioxidant after, prior to, along with the administration of the anticancer agent. In at least some cell lines, antioxidants increase cell death of anticancer agents by affecting post-translational changes in C / EBβ. C / EBPβ is also known as NF_IL6, AGP / EBPβ, LAP, IL-6DBP, rNF_IL6 and CRP2 (5-11), which are a diverse group of nuclear transcription factors, which are enhancers and / or promoters of these factors and target genes. A basic DNA binding domain that facilitates interaction between regulatory domains of and leucine zipper motifs necessary for dimer formation. C / EBPβ activates several acute-phase protein genes via NF_IL6 reactive elements, which are NF_IL6 reactive elements as nuclear targets, and C / EBPβ is also albumin, c-fos, and some adipocyte-specific proteins Indicates that it regulates the gene that encodes. Moreover, C / EBPβ is involved in the activation of various genes related to inflammatory immune responses, including granulocyte macrophage / colony-stimulating factors, and immunoglobulin genes, including interleukin-1 (IL-1) and interleukin-8 (IL-8). It is. Thus, C / EBPβ is a pleiotropic transactivator involved in a series of signaling and cell differentiation reactions. In one pathway, which may not be unique, antioxidants have been found to enhance the cell death of anticancer drugs through a series of reactions: (1) antioxidants increase the level of cAMP and cAMP phosphorylates C / EBPβ Activating protein kinase A, and phosphorylation of C / EBPβ changes the position of C / EBPβ from the cytoplasm to the nucleus, where it mediates the induction of p21, which stops cell growth; (2) Antioxidants inhibit dephosphorylation of C / EBPβ in the nucleus through inhibition of protein phosphatase 2A (PP2A). Reduction of methyltransferase activity results in inhibition of PP2A activity, and methyltransferase is involved in carboxymethylating the active subunit of PP2A to maintain PP2A in active form. Reduction of methylcarboxylation reduces the PP2A enzymatic dephosphorylation of the substrate C / EBPβ. Antioxidants simultaneously induce phosphorylation of C / EBPβ and inhibit dephosphorylation of C / EBPβ, thereby maintaining the active form of C / EBPβ in the nucleus of cells, thereby maintaining the continuous expression of p21 WAF1 / CIP1 , a cyclin-dependent kinase inhibitor. Induces disruption of the cell cycle. Thus, more generally, the present invention encompasses a method for vaporizing nuclear localization of C / EBβ protein in a cell, comprising administering an antioxidant into the cell. This method maintains C / EBPβ in an activated form, i.e., in phosphorylated form, inducing cell growth disruption and cell death. As a non-limiting example, the present invention is directed to compounds in which antioxidant activity (eg, pyrrolidinedithiocarbamate ("PDTC") and vitamin E homologue, Trolox®) prevent G1 cell cycle interruption and / or cell death. Induction reduces DNA replication in human colorectal cancer cells. However, antioxidant compounds have no effect on normal human colon cells, epidermal cells or mammalian epithelial cells (see Table 1). Cell cycle interruption is more pronounced in colorectal cancer cells expressing mutant P53 compared to wild type P53. Induction of cell cycle interruption and apoptosis is associated with the continuous induction of P21 WAF1CIP1 , a cyclin-dependent kinase inhibitor. Treatment with antioxidants in combination with 5-FU significantly reduces the growth of NCC colorectal cancer cells. Moreover, antioxidants alone significantly reduce the growth of established colorectal cancer in athymic mice, stop tumor growth when combined with 5-FU (Trolox®), or cause tumor degeneration. (Pyrrolidinedithiocarbite). Subsequently, DKO-1 cells (human colorectal cancer cell line) expressing epitope-attached C / EBβ protein were used to investigate whether post-translational modification (phosphorylation) of C / EBβ was the cause of increased C / EBβ activity. In vivo labeling with [32P] orthophosphate after immunoprecipitation showed PDTC forskolin (3R-3α, 4aβ, 5β, 6β, 6aα, 10, 10aβ, 10bα) -5- (acetyloxy) -3- ( Ethenyldodecahydro-6,10,10b-trihydroxy-3,4a, 7,7,10a-pentamethyl-1H-naphtho [2,1-b] pyran-1-one) There was no change in amount, but the phosphorylation of epitope-labeled C / EBβ increased 4 to 6 times. Deletion analysis of C / EBPβ was performed by mapping in vitro phosphorylation sites in C / EBPβ. Cleaved C / EBPβ comprising only 160 or 200 COOH-terminal amino acids was not good as a substrate for PDTC-induced phosphorylation, while cleaved C / EBPβ containing 305 COOH-terminal amino acids was as efficient as full length C / EBPβ. Phosphorylated by PDTC. A detailed examination of the amino acid sequence between 236 and 305 amino acids revealed that this region contained a common PKA phosphorylation site (Arg-X-Ser299-X). Activation of the cAMP-dependent protein kinase-mediated pathway, followed by phosphorylation of Ser299 of C / EBβ is critical for the subsequent nuclear cross-activation of genes for the intracellular nucleation and altered redox potential of C / EBβ. Dephosphorylation of the transcription factor by protein phosphatase 2A (PP2A) inactivates C / EBPβ in the nucleus. This enzyme is activated by carboxymethylation of the catalytic subunit of PP2A by methyltransferase. In a further experiment, carboxymethylation of the catalytic subunit of protein phosphatase 2A is inhibited by PDTC, a typical antioxidant, and further loss of carboxymethylation is achieved by inhibiting the activity of methyltransferase. These results support the fact that antioxidants inhibit the dephosphorylation (and thus deactivation) of C / EBβ by protein phosphatase 2A in the nucleus through inhibition of methyltransferase involved in maintaining the activated form of protein phosphatase 2A. Reduction of methylcarboxylation reduces the enzymatic dephosphorylation of the substrate C / EBPβ PP2A. In addition, it was found that the measured antioxidant had little or no effect on protein phosphatase 1 (PP1). This invention was made in part through federal funds under the National Institutes of Health grant numbers CA4613 and GM53319 CA69457, and the United States Government has certain rights in the invention. FIELD OF THE INVENTION The present invention relates to compositions and methods for antioxidants for improving the treatment of medical chemistry, more specifically for hyperproliferative diseases. The drawings presented in this specification merely illustrate preferred embodiments of the invention and are not intended to limit the scope thereof. 1A is a graph of soft agar forming units x 10 6 HCT15 and HCT16 cells (as a set of controls) versus the concentration of PDTC. This graph shows that pyrrolidinedithiocarbamate (PDTC) and vitamin E inhibit adhesion-independent growth in vitro. Soft-agar colony formation was measured by plating HCT115 or HCT116 cells in soft agar supplemented with medium (control) or with increasing concentrations of pyrrolidinedithiocarbamate (2.5-200 μM) or vitamin E (0.1-10 mM). . Colonies were counted after 10 days of incubation at 37 ° C. The measured values are representative of four to three experiments. FIG. 1B shows a series of flow cytometer assays showing that antioxidants induce G1 cell cycle interruption and apoptosis in CRC cells. Asynchronous HCT116 or HCT115 cells grow either with or without pyrrolidinedithiocarbamate (70 μM) or vitamin E (3 mM). 24 hours after exposure to antioxidants, cells were harvested and analyzed by flow cytometry. 1C is a bar graph showing the time after treatment of test compounds for intracellular H 2 O 2 levels. 1C shows the negative relationship between intracellular oxidative potential and cell cycle disruption. Changes in intracellular redox potential were determined by measuring internal H2O2 levels. Background fluorescence was subtracted from each measured value. Values are shown as calibrated DHR mean / 10 4 cells ± sem. Percentage of G1 (cycle) or dead (TUNEL-positive: squared) cells were analyzed by flow cytometry. 1D shows the effect of N-acetylcysteine, vitamin C and catalase on internal H 2 O 2 levels and cell cycle progression. HCT 15 cells were incubated for 24 hours with pyrrolidinedithiocarbamate (70 μM), vitamin E (3 mM), N-acetylcysteine (50 μM) or vitamin C (200 μM). As shown in FIG. 1C, internal H 2 O 2 levels and cell cycle changes were measured. In addition, cells were transiently transfected with empty plasmids or expression plasmids for human catalase and analyzed as described above 24 hours later. Values obtained from catalase-containing cells were subtracted from those obtained from cells transfected with empty plasmids and expressed in mean ± s.e.m. In a double dish. Figure 1E shows that pyrrolidinedithiocarbamate and vitamin E increased growth induction by 5-FU or doxorubicin in vitro. As described above, 5-FU (5 × 10 −8 to 5 × 10 −5 M) or doxorubicin (1 × 10) in the presence or absence of pyrrolidinedithiocarbamate (70 μM) or vitamin E (3 mM) HCT 116 and HCT15 cells were seeded in soap-agar with increased concentration of any of -9 to 1 × 10 −6 M). Colonies were counted after 10 days and IC 50 values were calculated with the concentration of 5-FU or doxorubicin required to reduce 50% (± sem) of basic colony formation. The values are representative of the tests performed four to three times. FIG. 2A is a graph of the mice with tumors and a photograph of it, showing that pyrrolidinedithiocarbamate and vitamin E improved the efficacy of 5-FU in wild type p53 of human CRC tumor xenografts. HCT 116 CRC was injected transdermally between nu / nu mouse scapula. Once the tumor reaches about 150 mm 3 , animals are given weekly pyrrolidindithiocarbamate (70 μM) or vitamin E (3 mM), 5-FU (40 mg / kg) or saline, or both antioxidants and 5-FU. Injected. The picture shows the effect of treatment on tumor size 4 weeks after the treatment. FIG. 2B is a graph of the number of weeks after treatment with various test agents for tumor volume (mm 3 ), which is the first treatment and salvage diet for variant p53 tumors and 5-FU and pyrrolidinedithiocarba The improved efficacy of the mate. As described above, HCT 15 induced-tumor was generated. Then treated with piloridinedithiocarbamate for 3 weeks in the presence or absence of 5-FU. At this point, treatment was discontinued for two months in animals receiving combination treatment of pyrrolidinedithiocarbamate and 5-FU. All other treatment groups received both 5-FU and pilidinedithiocarbamate for the remaining three weeks. 3A-3C show that pyrrolidinedithiocarbamate induces p21 WAF1 / CIP1 via p53. 3A shows that protein levels of p21 WAF1 / CIP1 are increased in CRC cells of humans expressing functional (HCT116) and variant (HCT15) after pyrrolidinedithiocarbamate treatment. CRC cells were treated with pyrrolidinedithiocarbamate (70 μM) as shown and subjected to western blot analysis. 3B shows p53-independent induction of p21 WAF1 / CIP1 mRNA by pyrrolidinedithiocarbamate in human CRC cells. Algebraically growing asynchronous human CRC cells were cultured in serum-containing medium containing 70 μM pyrrolidinedithiocarbamate. In addition, HCT116 cells containing HPV16E6 were analyzed for target degradation of p53. Cells were collected at the indicated time points and prepared for poly (A) mRNA isolation. Samples (3 μg) were electrophoresed on 1% (W / v) formaldehyde / agarose gel and transferred to nitrocellulose membrane. At 42 ℃ with 32 P-labeled p21 WAF1 / CIP1 probe was carried out northern hybridization platform ratting. IB15 was used as a control for the same loading and transfer. 3C shows that p21 WAF1 / CIP1 expression is required for antioxidant-induced apoptosis. HCT16 cells comprising either functional (p21 + / +) p21 WAF1 / CIP1 or deleted p21 WAF1 / CIP1 were treated with a given pyrrolidinedithiocarbamate or vitamin E for 24 hours and killed by TUNEL assay. Was analyzed. Values are expressed as percent GUNEL positive cells and represent mean ± sem of 3 measurements. 4A shows that pyrrolidinedithiocarbamate induces p21 WAF1 / CIP1 transcriptional activity through the NF_IL6 common sequence. The 2.4 kilobase pair p21 WAF1 / CIP1 promoter sequence and variants were fused to the luciferase reporter gene. TATA represents the TATA box of p21 WAF1 / CIP1 located 45 bp from the transcription start position (defined as +1). -2280, -2198, -2078, -1838, -1428 and 1138 define the 5 'end point for the short deletion structure. The -2280ΔNF_IL6 construct contains a complete promoter with two base pair mutations at the NF_IL6 site. All reporter constructs were transfected with HOCT 116 or HCT15 cells, and 24 hours later antioxidant-induced luciferase activity was measured in relative light units (RLU). Luciferase activity was changed to CAT activity and the results were shown as active folds against baseline. 4B shows that pyrrolidinedithiocarbamate treatment induces C / EBPβ DNA binding activity. Left panel: HCT116 and HCT15 cells were treated with 70 μM pyrrolidinedithiocarbamate for a period of time, and the nuclear extract was incubated with [γ- 32 P] labeled p21-NF_IL6 oligonucleotide. Right panel: Lanes 1-3, controls were treated with pyrrolidinedithiocarbamate for 12 hours (lane 1), unlabeled wild-type (lane 2) excess oligonucleotide and variant (lane 3) oligonucleotides Performed on nuclear extracts derived from treated HCT116 cells. Surface transfer assays were performed with lanes 4-6, C / EBPa (pannel 4), β (panel 5), δ (panel 6) polyclonal antibodies. 4C and 4D show that C / EBPβ can stimulate C / EBPβ promoter activity. HCT116 (FIG. C) and HCT15 (FIG. D) cells were transfected with cytomegalovirus (CMV) expression plasmids containing C / EBPa, β, or δ cDNA and 3 μg of p21 WAF1 / CIP1 -luciferase. Control plasmids are included in FIG. 4A. 4E shows that C / EBβ regulates cell sensitivity to antioxidant-induced apoptosis. Control HCT15 cells and sense or antisense C / EBPβ cell lines were cultured for 24 hours in the presence or absence of 10 μM of murinesterone A and / or of pyrrolidinedithiocarbamate (70 μM) or vitamin E (3 mM). The apoptosis index was measured as a percentage of TUNEL positive cells under light microscopy at 200-fold magnification, and the values were expressed as mean ± sem for 3 samples. Inset shows representative western blots for p21 WAF1 / CIP1 protein levels in transfected cell lines grown in the presence or absence of 10 μM of Murosterone A. 4F shows that increasing C / EBβ protein levels improves cell death of chemotherapeutic agents in vitro. Control HCT15 cells and SenseC / EBPβ cell lines were induced with 10 μM of Murosterone A and exposed to 5-FU (1.5 μM) or doxorubicin (0.1 μM) for 24 hours. Apoptosis index was calculated by the method described in Figure 4C. 5A and 5B show the effect of the test compound on proliferation of HCT 116 and HCT 15 cells, derived from athymic mice treated with a mixture of saline, vitamin E, PCTC, 5-FU, and vitamin E and 5-FU. It is a histogram showing the growth of BrDU-labeled cells (percent of total cell nuclei; BrDU refers to bromodeoxyuridine) taken as colorectal cell xenografts. 6A and 6B are colorectal cell xenografts derived from athymic mice treated with a mixture of saline, vitamin E, PCTC, 5-FU, and vitamin E and 5-FU as measurements of the effect of the test compound on apoptosis. Shows histogram of TUNEL-positive cells (percent of total cell nuclei; TUNEL refers to TdT-mediated dUTP nick end markers) as a transplant. The tumor tissue is fixed overnight in 4% (v / v) paraformaldehyde and embedded in paraffin according to standard histological methods. Fragments were previously treated with 10 mM citrate buffer (pH 6.0) and incubated with PC10 monoclonal antibodies against BrDU (Boehringer Mannbeim). TdT labeling of fragmented DNA was performed according to the manufacturer's instructions. Proliferation index (percent of total BrDU cell nuclei) and apoptosis index (TUNEL) were measured as percent of cells under a 200x light microscope. 7A-7D illustrate that PDTC treatment induces C / EBPβ DNA binding activity through post-translational modification. (A) DKO-1 cells were treated with 70 μM PDTC for a period of time, and nuclear extracts were prepared from [γ-32P] -labeled p21-NF_IL6 oligonucleotides (lanes 1-9). Specificity Assay: DKO-1 cells treated with lanes 10-12, control for 3 hours with PDTC (lane 5), unlabeled wild-type (lane 11) excess oligonucleotide and variant (lane 12) oligonucleotides It was performed on nuclear extracts derived from. Surface transfer assays were performed with lanes 13-15, C / EBPa (pannel 13), β (panel 14), δ (panel 15) polyclonal antibodies. (B) The same DKO-1 cell cultures were isolated and changes in C / EBβ mRNA levels related to treatment were measured by Northern blot analysis. IB15 is a control for the same loading and metastasis. (C) The same DKO-1 cell cultures were treated with PDTC (70 μM) in the presence of [ 32 P] orthophosphate. Prior to initiation (0 hours) or at certain times, C / EBPβ of the cytoplasm and nucleus portion was purified from the cells by immunoprecipitation. SDS-PAGE and magnetic radioactivity or Western blot analysis (100 μg / lane of whole cell protein) were used to analyze the localization of treatment-related C / EBPβ. (D) DKO-1 cells were incubated for 1 hour in the presence of PDTC (70 μM), followed by immunocytochemistry to detect differences in the method of fractionation of C / EBPβ. In all experiments, the cultures with prereversal serum or primary anti-C / EBPβ antiserum preincubated with in vitro transcribed C / EBPβ protein showed no fluorescence signal after treatment with the second Cy3-conjugated antibody. Representative photomicrographs show anti-C / EBPβ stained cells before and after PCTC treatment. 8A-8B illustrate the effect of PDTC on cAMP levels and PKA activity. DKO-1 cells were treated with PDTC (70 μM) for some time. Cytolysates were prepared and analyzed for (A) intracellular cAMP levels or (B) PKA activity. The figures are expressed as pmol average ± s.e.m. Per μg of protein and represent representative values of three of four experiments. 9A-9C show that PDTC phosphorylates C / EBPβ at Ser 299 . (A) Anti internal C / EBPβ of [ 32 P] orthophosphate labeled DKO-1 cells (2mCi / ml.3h) treated with 0 μM (lane 1), 70 μM PDTC (lane 2), or 50 μM forskolin Immunoprecipitated with -C / EBPβ antibody. Labeled proteins were labeled with SDS-PAGE and autoradiography. (B) Tryptic phosphopeptide map of in vivo labeled epitope-tack C / EBPβ. Immunoprecipitated wild-type (WT) and variant (Ala 299 ) C / EBPβ were digested with trypsin from PDTC treated or untreated DKO-1 cells with antibodies to FLAG-epitope, and the phosphopeptide was isolated by electrophoresis and TLC. And indicated by an automatic radioactivity method. X 1,2 was phosphorylated continuously. Levels of phosphopeptide X 3 increased after treatment with PDTC in cells transfected with non-variant wild type proteins. The circle represents its origin. (C) Comparison of phosphorylation of in vivo phosphorylation of wild-type C / EBPβ and Ala substitution of C / EBPβ from cells treated with and without PDTC. Autoradioactivity (top) and C / EBβ immunoblot are shown. 10A-10B are essential for the PKA phosphorylation of C / EBβ to be located in the nucleus. (A) The same DKO-1 cells were treated with PDTC (0 μM or μ70M) for 3 hours. Poly (A) + mRNA and protein were isolated from each group, and changes related to the treatment on C / EBβ mRNA and protein levels were measured by Norte or Western blot methods. IB15 was shown as a control for the same loading and metastasis. (B) DKO-1 cells were treated for 3 hours with PDTC (0 μM or μ70M) or PDTC and mPKI (myristylized protein kinase A, 1 μM). Cells were fixed in paraformaldehyde and C / EBβ protein was visualized by immunofluorescence staining. Cells treated with mPKI alone could not induce nuclear localization of C / EBβ (data not shown). FIG. 11 shows that PDTC inhibits carboxymethylation of catalytic subunitite of protein phosphatase 2A. DKO-1 cells were cultured in serum-containing medium containing [methyl- 3 H] S-adenosyl methionine and / or 70 μM PDTC for 3 hours. The cytoplasm or nuclear sections were prepared and immunoprecipitated C / EBβ using standard methods. Antigen / antibody complexes were separated by SDS-PAGE and the presence of PP2Ac was detected by fluorescence. PDTC inhibits methyltransferase in the nucleus and to a lesser extent the cytoplasm. Figure 12 shows that PDTC inhibits the methyltransferase activity of PP2Ac. PP2A (a and c dimers) were incubated in the presence of [methyl- 3 H] S-adenosyl methionine with increasing concentrations of methyltransferase in PDTC and partially purified rats at 37 ° C. for 30 minutes. The reaction was terminated with SDS-sample buffer. Samples were separated by SDS-PAGE and visualized the presence of methylated PP2A catalytic subunits by fluorescence. As shown, PDTC selectively inhibits the activity of methyltransferase to carboxymethylate the catalytic subunit of PP2A in a dose dependent manner. 13 is a graph of percent radioactivity remaining in the time zone protein substrate. The numbers indicate that PDTC inhibits PP2A but not PP1. The activity of PDTC was compared to I2 (selective PP1 inhibitor), and compared to okadaic acid (inhibitors of PP2A and PP1) and I2, and to okadaic acid and PDTC. DKO-1 cells were cultured in the presence and absence of PDTC (test) (control). After lysing the cells, radiophosphorylated C / EBPβ was added. Test compounds were added and reacted with the lysate. The protein was collected and the amount of radioactive phosphate remaining in the protein was measured. Figure 14 shows the formation of the complex with the transcription factor C / EBPβ with PP2Ac protein phosphatase. Water-soluble brain extracts of rats were fractionated with phenylsepharose and methyltransferase activity was analyzed using an external PP2A heterodimer (a-c complex). In addition, the active peak of methyltransferase was fractionated by source Q, strong anion exchange and gel filtration chromatography. The partially purified methyltransferase shown in FIG. 14 shows the highest methyltransferase activity from the gel filtration column. This peak portion of methyltransferase activity was further subjected to DEAE, weak anion exchange and monoQ, other strong anion exchange resins, and columns. Both C / EBβ and PP2A were detectable after this additional step. Rat brain extracts were expressed as positive controls (C / EBβ and PP2Ac migrated at about 45 and 36 kDa on SDS-PAGE). Through the mechanism of inducing p53 independent of p21 expression by binding to a specific position on the p21 promoter, it was found that antioxidants interrupt the cell cycle of abnormally proliferating cells and induce apoptosis. It was also found that site specific phosphorylation at the Ser299 position of C / EBβ by protein kinase A after treatment with antioxidants is essential for nuclear localization of this protein. In addition, through inhibition of PP2A, it was found that antioxidant prevented dephosphorylation of C / EBPβ in the nucleus, thereby inactivating and delocalizing C / EBPβ. Inhibition of PP2A is caused by a decrease in methyltransferase, which is involved in the maintenance of active PP2A as an enzyme that carboxymethylates the catalytic subunit of PP2A. Reduction of methylcarboxylation reduces the enzymatic dephosphorylation of the substrate C / EBPβ PP2A. By inducing phosphorylation of C / EBPβ and inhibiting dephosphorylation of C / EBPβ, antioxidants maintain C / EBPβ activity in the cell nucleus to continuously induce the expression of p21 WAF1 / CIP1 to disrupt the cell cycle. Methyltransferases, which are responsible for the carboxymethylation of PP2A subunits in vivo and in vitro, are a unique type of carboxymethyltransferase. Type II and Type III carboxymethyltransferases in mammals appear to have substantially different properties from enzymes that carboxymethylate PP2Ac subunits. Protein Carboxy Methyltransferase Type II modifies D-aspartyl and L-aspartyl residues and accumulates in proteins as they age, thus methylating other proteins containing these amino acids. Carboxymethyltransferase type III modifies the protein in cysteine in front of the C-terminus of the protein (G-protein), requiring isoprenylation of cysteine and cleavage of the last three carboxy terminal residues. Antioxidant treatment of the DKO-1 colorectal cell line in vitro does not alter the activity of this carboxymethyltransferase. Thus, in vitro data indicate that antioxidants selectively inhibit the methyltransferase activity of PP2Ac but do not inhibit G-protein methylation. In addition, a novel high dimensional protein complex consisting of C / EBβ, PP2A and methyltransferase was identified. Thus, another embodiment of the present invention is that the novel complex in isolated form is present, for example at 70%, preferably at 80% or 90% purity. A method for isolating this enzyme is provided in Example 27. I. Embodiments of the Invention Based on the basic findings described herein, there is provided a method of increasing the nuclear localization of C / EBPβ, which comprises administering an antioxidant intracellularly. This method stops cell growth and induces apoptosis by keeping C / EBPβ protein in phosphorylated active state. In one example, the invention is a method of increasing the cytotoxic activity of an anticancer agent comprising administering to the host an effective amount of an anticancer agent in need of treatment with an amount of antioxidant effective to increase cell death. Antioxidants, including those specifically described herein, have been found to be useful for inducing cell cycle interruption (G1, G2, S and M types) to increase the efficacy of anticancer agents for the treatment of diseases associated with abnormal cell proliferation. This method revealed that C / EBPβ protein was maintained in phosphorylated active state to stop cell growth and induce apoptosis. In another example, a cell of an anticancer or chemotherapeutic agent for abnormal cell proliferative disease, comprising administering to the patient or animal an effective amount of an antioxidant to or in combination with a pharmacologically effective amount of a chemotherapeutic agent or, optionally, to increase cell death. Increasing lethality provides a method for increasing phosphorylated activity of C / EBβ protein in a host, such as a subject or animal in need of such treatment. In another example, the present invention treats a patient with a tumor disease comprising administering to the host a therapeutically effective amount of cytotoxic chemotherapy selected from among chemotherapy and radiation theraphy of a tumor with an antioxidant. It is about how to. Representative tumor chemotherapeutic agents and antioxidants are listed below. Any radiotherapy that can ameliorate abnormal cell proliferative conditions is suitable for the present invention and includes ionizing radition, either particulate or electromagnetic. Suitable and effective dosages of radiation therapy for various tumor conditions are well known. In one non-limiting example, radiation therapy is gamma irradiation at a suitable time, for example, from about 3,000 centigrey to about 5,000 centigrey for up to six weeks. The invention also increases the expression of p21 as a means of inducing cell disruption and apoptosis in patients in need of such treatment, including administering a pharmacologically effective amount of an antioxidant or a combination of an antioxidant and an anticancer agent. It provides a method to make it. The present invention further relates to a method for controlling cell cycle interruption and apoptosis in patients in need of such treatment comprising administering a pharmacologically effective amount of an antioxidant or an antioxidant and an anticancer agent. In another example, to achieve the effect described in detail herein, a therapeutic effect may be achieved by administering an effective amount of C / EBB, or a protein substantially homologous to C / EBB. The protein or protein homolog can be administered alone or in adjuvant treatment with an anticancer agent. Proteins that are substantially homologous to C / EBPβ are form-X1-Arg-X2-Ser-X3 (where X2 is a C / EBPβ amino acid at position 298 and X1 and X3 are substantially identical to C / EBPβ sequences). It is defined as comprising a peptide sequence of the peptide sequence having the homologous) or consisting of a peptide sequence of the form -X1-Arg-X2-Ser-X3. Substantial homology refers to a protein or peptide sequence that performs substantially the same function as the parent sequence and has at least 60%, or more preferably at least 75%, more preferably 90% or 95% or more sequence identity. it means. Effective methods of protein delivery are known and will be employed with this embodiment to increase the effectiveness of this therapy. In another example, one may administer a synthetic Ser299 phosphorylated C / EBβ homologue with stabilized phosphate bonds that can withstand dephosphorylation. Such stabilized phosphates include, but are not limited to, phosphoroamidates and phosphonate homologues. The present invention also provides a method of inhibiting protein phosphatase 2A (PP2A) in a cell, comprising administering an inhibitory amount of an antioxidant of protein phosphatase intracellularly. In another embodiment of the present invention, a method of reducing the carboxymethylation state of a catalytic subunit of PP2A is provided, comprising contacting a cell with a methyltransferase or methylesterase-lowering antioxidant. In one pathway, but not exclusively, antioxidants have been found to increase cell death of anticancer agents through a series of reactions, including: In one pathway, which may not be unique, antioxidants have been found to enhance the cell death of anticancer drugs through a series of reactions: (1) antioxidants increase the level of cAMP and cAMP phosphorylates C / EBPβ By activating protein kinase A, the phosphorylation of C / EBβ changes the position of C / EBβ from the cytoplasm to the nucleus, where it mediates the induction of p21, which stops cell growth; (2) Antioxidants inhibit dephosphorylation of C / EBPβ in the nucleus through inhibition of protein phosphatase 2A (PP2A). Inhibition of PP2A activity is caused by a decrease in methyltransferase activity, and methyltransferase is involved in carboxymethylating the active subunit of PP2A to maintain PP2A in an activated form. Reduction of methylcarboxylation reduces the PP2A enzymatic dephosphorylation of the substrate C / EBPβ. Antioxidants simultaneously induce phosphorylation of C / EBPβ and inhibit dephosphorylation of C / EBPβ, thereby maintaining the active form of C / EBPβ in the nucleus of cells, thereby maintaining the continuous expression of p21 WAF1 / CIP1 , a cyclin-dependent kinase inhibitor. Induces disruption of the cell cycle. Based on this finding, a method of identifying a therapeutically effective compound for the treatment of abnormal cell proliferation is provided, including determining whether the compound can increase the localization of C / EBβ protein in the nucleus of the cell. In another example, there is provided a method of identifying a therapeutically effective compound for the treatment of abnormal cell proliferation, comprising measuring the ability of the compound to increase phosphorylation at Ser29 of the C / EBp protein. The method comprises performing C / EBβ immunoprecipitation of the nucleus after incubating the selected cell line with the test compound at 37 ° C. for a predetermined time (eg 3 hours). Trypsin digestion and TLC were performed to confirm phosphorylation of C / EBPβ. Based on the findings detailed in this specification, those skilled in the art will appreciate that the present invention further includes, but is not limited to, the following examples. (1) A method for identifying a therapeutically effective compound by measuring the ability of the compound to change the phosphorylation status of C / EBβ in Ser299. In this method, the test compound is included in a solution comprising at least a dimer form of protein phosphatase 2A, methyltransferase and [methyl-3H] S-adenosyl methionine, including at least phosphorylated C / EBPβ, a and c subunits. do. (2) A method for identifying a therapeutically effective compound by measuring the ability of the compound to inhibit protein phosphatase 2a using (1) or another method apparent or well known to those skilled in the art. (3) A method for identifying a therapeutically effective compound by measuring the ability of the compound to change the carboxymethylation state of protein phosphatase 2a. (4) A method for identifying a therapeutically effective compound by measuring the ability of the compound to change methyltransferase activity. (5) a peptide sequence of the form X1-Arg-X2-Ser-X3, wherein X2 is a C / EBPβ amino acid at position 298 and X1 and X3 are peptides surrounding the peptide sequence having substantially homology with the C / EBPβ sequence Says.) (6) cAMP-dependent protein kinase, protein kinase C, in a patient or animal in need of treatment comprising administering to the patient or animal a pharmacologically effective amount of an antioxidant that increases the nuclear retention time and function of C / EBPβ. A method for enhancing the phosphorylation status and function of C / EBPβ induced by modulators including but not limited to ras-dependent MAP kinase and calcium-chalmodulin dependent kinase. (7) cAMP-dependent protein kinase, protein kinase C, ras-dependent MAP kinase and calcium-chalmodulin dependent kinase in patients or animals in need of treatment comprising administering an antioxidant to a patient or animal in a pharmacologically effective amount. Method for improving the phosphorylation status and function of C / EBPβ induced by a regulator, including but not limited to. (8) A method of treating a host, such as a patient or animal, exhibiting or at risk for developing a cancer disease, comprising administering an antioxidant to a patient or animal in a pharmacologically effective amount. (9) A method of treating a patient or animal at risk of developing cancer disease, comprising increasing nuclear localization of C / EBβ expression and function. (10) administering to the patient or animal a therapeutic agent which increases the intranuclear retention time of C / EBPβ to a patient or animal, wherein the therapeutic agent consists of an antioxidant alone or a combination of an antioxidant and an anticancer agent. A method of treating a patient having an abnormal cell proliferative disease, including but not limited to. (11) tumors and by measuring C / EBPβ activation, C / EBPβ phosphorylation and nuclear retention time, PP2A inhibition of carboxymethylation of catalytic subunits of PP2A, and inhibition of methyltransferase or methylesterase alone or together A method of diagnosing and measuring the response to treatment of a patient with a cell proliferative disease. II. Antioxidant As used herein, the term antioxidant refers to a substance that prevents oxidation of an oxidizable compound under physiological conditions. In one example, if a compound can produce oxygen radicals in vitro, the compound is considered an antioxidant for this disclosure. Under oxidized conditions, antioxidants can be added to the cell extract to assess the effect of the oxidizable compounds. As a non-limiting example, the antioxidant scavenger oxygen, superoxide anion, hydrogen peroxide, superoxide radical, lipooxide radical, hydroxy radical, or a reactive metal to bind to the lipid protein, nucleic acid and the like to prevent oxidative damage. Antioxidants include, but are not limited to, the following classes of compounds. A) dithiocarbamate Dithiocarbamates are widely described in patents and scientific literature. Dithiocarbamates and related compounds have been extensively reviewed, for example, GD Thorn et al. "The Dithiocarbamates and Related Compounds" Elsevier, New York, 1962. US Pat. Nos. 5,035,878 and 5,294,430 cause dithiocarbamates to be treated with anticancer drugs. It is described that the bone marrow can reverse the damage to the blood producing function (blood production damage). Both of these patents describe all pharmacologically acceptable dithiocarbamates that can increase the nuclear localization of C / EBβ that is suitable for use in the present invention. Active compound Dithiocarbamate is a precursor metal chelate used clinically for heavy metal poisoning. Baselt, R. C. F.W. J. Sunderman, et al (1977), "Comparisons of antidotal efficacy of sodium diethyldithiocarbamate, D-penicillamine and triethylenetetraamine upon acute toxicity of nickel carbonyl in rats" Res Commun Chem Pathol Pharmacol 18 (4): 677-88; Menne, T. and K. Kaaber (1978), "Treatment of pomphoyx due to nickel allergy with chelating agens" Contact Dermatitis 4 (5): 289-90; Sunderman, F.W. (1978), "Clinical response to therapeutic agents in poisoning from mercury vapor" Ann Clin Lab Sci 8 (4): 259-69; Sunderman, F. W. (1979), "Efficacy of sodium diethyldithiocarbamate (dithocarb) in acute nicke carbonyl poisoning" Ann Clin Lab Sci 9 (1): 1-10; Gale, G.R., A.B. Smith, et al., (1981), "Diethyldithiocarbamate in treatment of acute cadmium poisoning" Ann Clin Lab Sci 11 (6) :: 476-83; Jones, M.M. and M G. Cherian (1990), "The search for chelate antagonists for chronic cadmium intoxication" Toxicology 62 (1): 1-25; Jones, S. G., M. A. Basinger, et al (1982), "A comparison of diethyldithiocarbamate and EDTA as antidotes for acute cadmium intoxication" RE Commun Chem Pathol Pharmacol 38 (2): 271-8; Pages, A., JSCases, et al. (1985), "Dithiocarbamates in heavy metal poisoning: complexes of N, N-di (I-hydroxyethyl) ditocarbamate with Zn (II), Cd (II), Hg (II) , CH3Hg (II), and C6H5Hg (II): J. Inonrg Biochem 25 (I): 35-42; Tandon, SK, NS Hashmi, et al. (1990), "The lead-chelating effects of substituted dithiocarbamates" Biomed Environ Sci 3 (3): 299-305. Dithiocarbamates are also used incidentally in cis-platinum chemotherapy to prevent kidney toxicity. Hacker, M.P., W.B. Ershler, et al. (1982). "Effect of disufiram (tetraethlthiuram disulfide) and diethydithocarbamates on the bladder toxicity and antitumor antivity of cyclophosphamide in mice" cancer Res 42 (11): 4490-4, Bodenner., 1986 # 733; Saran, M. and Bors, W. (1990). "Radical ractions in vivo--an overvieww" Radiat. Environ. Biophys. 29 (4): 249-62. Dithiocarbamates currently used for alcohol abuse are disulphiram, dimers of diethyldithiocarbamate. Disulfuram inhibits liver aldehyde dehydrogenase. Inoue, K., and Fukunaga, et al., (1982). "Effect of disulfiram and its reduced matabolite, diethylthiocarbamate on aldehyde dehydrogenase of human erythrocyte" Life Sci 30 (5) "419-24. It has been reported that dithiocarbamate inhibits HIV virus replication and enhances maturation of specific T cell subgroups. This led to clinical trials of diethyldithiwabamate in AID patients. Reisinger, E., et al., (1990). "Inhibition of HIV progression by dithocarb." Lancet 335: 679. Dithiocarbosylate is a compound of the A-SC (S) -B structure and is a member of the general group known as thiol antioxidants, which are alternatively referred to as carbodithiol or carbodithiolate. The SC (S) moiety is essential for therapeutic activity, and A and B can be any group that does not adversely affect the efficacy or toxicity of the compound. In another embodiment, one or two sulfur atoms are replaced with selenium atoms in dithiocarbamate. Replacing the sulfur atom with selenium can reduce the toxicity of the molecule in some cases so that the patient can be better tolerated. One of ordinary skill in the art can select A and B to give the desired properties to the compound, including size, charge, toxicity and stability (including stability in basic environments such as the intestinal tract or acidic environments such as the gastrointestinal tract). In addition, the choice of A and B has a significant effect on the tissue-distribution and pharmacokinetics of the compound. The compound is preferably discharged by the kidneys and removed. The advantage of pharmacologically administering dithiocarboxylates is that they are not enzymatically cleaved in vivo by thioesterases and thus have an extended life in vivo. In a preferred embodiment, A is hydrogen or pharmacologically acceptable cation, including but not limited to sodium, potassium, calcium, magnesium, aluminum, zinc, bismuth, barium, copper, cobalt, nickel or cadmium; Generally carboxylic acids, including but not limited to acetic acid, oxalic acid, tartaric acid, succinic acid, maleic acid, ascorbic acid, benzoic acid, carbonic acid, pamoic acid, alginic acid, polyglutamic acid, naphthalenesulfonic acid, naphthalenedisulfonic acid, or polygalactonic acid Organic acid forming salts; Nitrogen-containing heterocycle, or structural formula NR4R5R6R7, Where R4, R5, R6, R7Are each independently hydrogen, C1-6Straight, branched, or (C4-6Cyclic alkyl, hydroxy (C)1-6Alkyl, wherein one or more hydroxy groups are located on any one of the carbon atoms, or aryl, N, N-dibenzylethylene-diamine, D-glucosamine, choline, tetraethylammonium, or ethylenediamine Cations formed from unrestricted ammonia or other hydrogen generating bases. In another example, it may be a physiologically cleavable leaving group cleavable in vivo from an A-attached molecule, including acyl (including acetyl, propionyl, and butyryl), alkyl, phosphate, sulfate or sulfonate One is not limited thereto. In another example, B is alkyl, akenyl, alkynyl, alkaryl, aralkyl, haloalkyl, haloalkenyl, haloalkynyl, aryl, alkaryl, hydrogen, C 1-6 alkoxy-C 1- 10 alkyl, C 1-6 alkylthio-C 1-10 alkyl, NR 2 R 3 ,-(CHOH) nCH 2 OH, where n is 0, 1,2,3,4,5, or 6 (CH 2 ) nCO 2 R 1, hydroxy (C 1-6 ) alkyl, wherein at least one hydroxy group is present at any one of the carbon atoms, including acetyl, alkylpropionyl, and alkylbutyryl. In another example, B is NR 2 R 3 , wherein R 2 and R 3 are each independently alkyl ;-( CHOH) n (CH 2 ) nOH, where n is 0,1,2,3,4,5 , 6; -(CH 2 ) nCO 2 R 1 ,-(CH 2 ) nCO 2 R 4 ; Hydroxy (C 1-6 ) alkyl-; Alkenyl (including vinyl, allyl and CH 3 CH═CH—CH 2 —CH 2 ); Alkyl (CO 2 H), alkenyl (CO 2 H), alkynyl (CO 2 H) or aryl, wherein the aryl group is substitutable as described above, for example NO 2 , CH 3 , t-butyl, A CO 2 H, halo, p-OH group; Or R 2 and R 3 may constitute a bridge, such as — (CH 2 ) m —, where m is 3,4,5,6,7,8,9, or 10 and R 4 is acetyl, propionyl And alkyl, aryl, alkaryl, or aralkyl, including butyryl. In another example, B is a heterocyclic or alkylheterocyclic group. The heterocycle may optionally be partially or fully hydrogenated. Non-limiting examples are those listed above including phenazine, phenothiazine, pyridine and dihydropyridine. In another embodiment, B is a residue of a pharmacologically active compound or drug. The term drug, as used herein, refers to any substance used internally or externally as a drug for the treatment, treatment or prevention of a disease or condition. The -C (S) SA-group can be attached directly to the drug or bound via any suitable binding moiety. In another example, dithiocarbamate is an amino acid derivative of the structure AO 2 CR 9 -NR 10 -C (S) SA, wherein R 9 is a divalent B portion, a linking portion, or an internal residue of naturally occurring amino acids ( For example, CH 3 CH of amines, CH 2 of glycine, CH (CH 2 ) 4 NH 2 of lysine, and R 10 are hydrogen or lower alkyl. In addition, B may be a polymer in which one or more dithiocarbamate groups are attached directly or through a suitable linking group. Preferably the dithiocarbamate is released from the polymer under in vivo conditions to provide a therapeutic effect for a suitable time. In a preferred embodiment, the polymer itself can also be degraded in vivo. The term biodegradable or bioerodible as used herein refers to a physiological solution of PH6-8 at 25-37 ° C. within an acceptable time for the desired application (usually in vivo treatment), usually within 5 years and preferably within 1 year. Refers to a polymer that is exposed to decompose or dissolve. In a preferred embodiment, the polymer decomposes upon application for 1 hour and several weeks. Many degradable polymers are known. Non-limiting examples are peptides, proteins, nucleoproteins, lipoproteins, glycoproteins, synthetic and natural polypeptides and polyamino acids, arginine, lysine, aspartamic acid, cysteine, cystine, glutamic acid, glutamine, hydroxylysine, serine, threonine Polymers, including but not limited to tyrosine; Polylactic acid such as polylactic acid, polyglycolic acid, poly (lactide-co-glycolide), polyanhydride, albumin or collagen, polysaccharides with sugar units such as lactose, and poly (a-hydroxy) such as polycaprolactam Acid) is a polyorthoester. The polymer may be any polymer or block polymer. B may also be a group that increases the water solubility of dithiocarbamate, examples of which are as follows; - lower alkyl, -OR 8 (wherein, R 8 is -PO 2 (OH) - M + or PO 3 (M +) 2, wherein M + is a pharmaceutically acceptable cation Im); - C (O) ( CH 2 ) 2 CO 2 - M + , or -SO 3 - M + ; Lower alkylcarbonyl-lower alkyl; Carboxy lower alkyl; Lower alkylamino-lower alkyl; N, N-di-substituted aminolower alkyl-, wherein the substituents each independently represent lower alkyl; Pyridyl-lower alkyl-; Imidajoil-low work kill; Imidazoyl-Y-lower alkyl, wherein Y is thioene amino; Morpholinyl-lower alkyl; Pyrrolidinyl-lower alkyl; Thiazolinyl-lower alkyl-; Piperidinyl-lower alkyl; Morpholinyl-lower hydroxyalkyl; N-pilyl; Piperazinyl-lower alkyl; N-substituted piperazinyl-lower alkyl, wherein the substituent is lower alkyl; Triazoyl-lower alkyl; Tetrazoyl-loweralkyl; Tetrazoylamino-lower alkyl; Or thiazoyl-lower alkyl. In another embodiment, dimers such as B-C (S) S-SC (S) -B may be administered. The term "alkyl" as used in this specification, unless stated otherwise, refers to a saturated straight chain, branched or cyclic (if C 5 or higher) C 1-10 hydrocarbon (or lower alkyl, ie C 1-5 ), Specifically methyl, ethyl, propyl, isopropyl, butyl, isobutyl, t-butyl, pentyl, cyclopentyl, isopentyl, neopentyl, hexyl, isohexyl, cyclohexyl, cyclohexylmethyl, 30 methyl pentyl, 2,2 -Dimethylbutyl, and 2,3-dimethylbutyl. The alkyl group may be optionally substituted with any one of carbons having one or more moieties selected from the group consisting of hydroxy, amino, mono- or di-substituted amino, and the substituents are independently alkyl, aryl, alkaryl, or Aralkyl; Aryl, alkoxy, aryloxy, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate or phosphonate, or if unprotected or protected, if necessary, are known to those skilled in the art, such as Greene et al. in Organic Synthesis "John Wiley and Sons, Second Edition, 1991. The term "alkenyl" in this specification, unless stated otherwise, refers to a straight chain, branched, or cyclic C 2-10 hydrocarbon having one or more double bonds. The term "aralkyl" refers to an aryl group having one or more alkyl substituents. The term "alkaryl" refers to an alkyl group having one or more aryl substituents. The term "halo (alkyl, alkenyl, or alkynyl) refers to an alkyl, alkenyl, or alkynyl group in which one or more hydrogens in the group are replaced with halogen atoms. As used herein, the term "aryl" refers to phenyl, biphenyl, or naphthyl, preferably phenyl. The aryl group may be optionally substituted with one or more moieties selected from the following group; Alkyl, hydroxy, amino, ilkyamino, aramino, alkoxy, aryloxy, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, CO2H or pharmacologically acceptable salts thereof, CO2 (alkyl , Aryl, alkaryl or aralkyl), or glutamine, if necessary protected or unprotected, these are known to those skilled in the art and are described, for example, in " Protective Groups in Organic Synthesis " Second Edition, 1991. As used in this specification, the term "alkoxy" refers to a part of the -O-alkyl structure unless otherwise specified. As used herein, the term "acyl" refers to a group having the structure C (O) R 'unless otherwise specified, where R' is an alkyl, aryl, alkaryl, or aralkyl group. As used herein, the term “heterocyclic” refers to an aromatic moiety having at least one sulfur, oxygen, or nitrogen in the aromatic ring. Non-limiting examples include ferrazine, phenothiazine, furyl, pyridyl, pyrimidyl, thienyl, isothiazolyl, imidazolyl, tetrazolyl, pyrazinyl, benzofuranyl, benzothiophenyl, quinolyl, isoxet Nolyl, benzothienyl, isobenzofuryl, piazolyl, indolyl, isoindolyl, benzimidazolyl, furinyl, morpholinyl, carbozolyl, oxazolyl, thiazolyl, isothiazolyl, 1,2,4 -Thiadazolyl, isoxalzolyl, pyrrolyl, pyrazolyl, quinazolinyl, pyridazinyl, pyrazinyl, cynolinyl, phthalazinyl, quinoxalinyl, santinyl, hypoxanthinyl, putridinyl , 5-azacytidinyl, 5-azaurasilyl, thiazolopyridinyl, imidazolopyridinyl, pyrrolopyrimidinyl, pyrazolopyrimidinyl, adenine, N 6 -alkylpurine, N 6 -benzylpurin, N 6 -halopurin, N 6 -vinylpurine, N 6 -acetylenicpurin, N 6 -acylpurin, N 6 -hydroxyalkylpurine, N 6 -thioalkylpurine, thymine, cytosine , 6-aza-pyrimidine, 2-mercapto toffee limiter Dean, uracil, N 5 - alkyl pyrimidine, N 5 - benzyl pyrimidine, N 5 - halo-pyrimidine, N 5 - vinyl-pyrimidine, N 5 - acetyl renik Pyrimidine, N 5 -acylpyrimidine, N 5 -hydroxyalkylpurine, and N 5 -thioalkylpurine and isotazolyl. The heterocyclic group may be optionally substituted with a substituent as described for aryl. The heterocyclic group may be preferably partially or wholly hydrogenated. As a non-limiting example, dihydropyridine can be used in place of pyridine. Functional oxygen and nitrogen groups on the heterocyclic base may be protected during the reaction sequence if necessary or desired. Suitable protecting groups are well known to those skilled in the art and include: Acyl groups, such as trimethylsilyl, dimethylhexylsilyl, t-butyldimethylsilyl, and t-butyldiphenylsilyl, tritylmethyl, alkyl, acetyl and propionyl groups, methylsulfonyl groups, and p-tolulsulfonyl groups. As used herein, the term "hydroxyalkyl" refers to a C 1-6 alkyl group wherein at least one hydrogen group bonded to a carbon atom is substituted with a hydroxy group. The term "pharmacologically acceptable derivative" refers to a derivative of the active compound which can provide the parent compound directly or indirectly upon administration to the receptor or exhibits its activity. The term "pharmacologically acceptable cation" refers to an inorganic or organic moiety that has a positive charge and can be administered with a coaion in a medicament, such as a salt. Pharmacologically acceptable cations include, but are not limited to, sodium, potassium, and tetravalent amines as known to those skilled in the art. The term "physiologically cleavable leaving group" refers to a moiety cleavable in vivo from a molecule to which the group is bound, and includes inorganic or organic anions, pharmacologically acceptable cations, acyl (acetyl propionyl and butyryl) But not limited to (alkyl) C (O)), alkyl, phosphates, sulfates and sulfonates. The term "composition or compound plus stereoisomer" refers to a composition or compound in which a single stereoisomer of the compound contains at least 95%, preferably at least 97, 98, 99 or 100% by weight. The term "amino acid" includes synthetic and natural amino acids, such as alanyl, valenyl, leucineyl, isolecinyl, prolinyl, phenylalanyl, tryptophanyl, methioninyl, glycinyl, seri Nil, threoninyl, cysteinyl, tyrosinyl, asparazinyl, glutaminyl, aspanoyl, glutayl, lysinyl, arginyl, and histidinyl. As used herein, the term "linking moiety" refers to a divalent group connecting two chemical moieties and includes alkyl, alkenyl, alkynyl, aryl, polyalkyleneoxy (eg, [(CH 2 ) nO-] n, -C 1-6 alkoxy-C 1-10 alkyl-, C 1-6 alkylthio-C 1-10 alkyl-, -NR 3 _, and-(COHO) nCH 2 OH, wherein n is each 0,1, 2,3,4,5, or 6). As described in Chapter 2 of Thorn et al., The preparation of dithiocarbamates is very simple. In general, compounds of formula R 1 R 2 NCSSH or R 1 R 2 NSSNa were prepared by reaction of carbon disulfide with secondary amines in alcohol or aqueous solution. It is common to carry out this method in the presence of NaOH so that sodium dithiocarbamate is produced. Thus, for example, sodium dimethyldithiocarbamate is prepared from CS2NaOH and dimethylamine. See page 14 of Thorn et al., Incorporated herein by reference. Other common dithiocarbamate compounds described in Thorn et al. Are: N-methyl, N-ethyldithiocarbamate, hexamethylenedithiocarbamic acid, sodiumdi (beta-hydroxyethyl) dithiocarbamate, sodium N -Methyl, N-cyclobutylmethyldithiocarbamate, sodium N-allyl-N-cyclopropylmethyl-dithiocarbamate, cyclohexylamyldithiocarbamate, dibenzyl-dithiocarbamate, sodium dimethylene-diti Ocarbamate, various pentamethylene dithiocarbamates, sodium piloridine-N-cardithioate, sodium piperidine-N-cardithioate, sodium morpholine-N-carbodithioate, alpha-fur Ruryl dithiocarbamate and imidazoline dithiocarbamate. B) probucol and its derivatives Probucol is chemically formulated with 2, [3] -ter-butyl-4-hydroxyanisole (BHA) and 2,6-di-tert-butyl-4-methylphenol (BHT), which are widely used food additives. Related. Its full chemical name is 4,4 '-(isopropylidenedithio) bis (2,6-di-tert-butylphenol). US Pat. No. 5,262,439 to Parthasarathy discloses water-soluble probucol homologs substituted with one or two hydroxy groups with ester groups that increase the water solubility of probucol. In one example, the water soluble derivative is selected from the group consisting of mono or di-succinic acid esters, glutaric acid esters, adipic acid esters, suveric acid esters, sebacic acid esters, azelaic acid or malic acid esters of probucol. In another embodiment, the probucol derivatives are mono or diesters which are esters containing alkyl or alkenyl containing functional groups selected from carboxylic acid groups. Compounds of the '439 patent are used in the present invention. US Pat. No. 5,155,250 (incorporated by reference herein) discloses 2,6-dialkyl-4-silylphenol as an atherosclerotic agent. The same compound is described as serum cholesterol lowering agent in PCT publication WO95 / 15760 (published June 15, 1995). U.S. Patent 5,608,095 (incorporated herein by reference) discloses atherosclerosis by alkylated-4-silyl-phenols inhibiting the peroxidation of LDL, lower plasma cholesterol and inhibiting the expression of VCAM-1. It is described as being useful for the treatment of. These compounds may also be used in the present invention. C) N-acetyl cysteine and derivatives thereof Cysteine is an amino acid with one asymmetric carbon. It is present as L-stereoisomers, D-stereoisomers, or mixtures of L-stereoisomers and D-stereoisomers. L-stereoisomers are forms that exist in nature. N-acetylcysteine (acetamide-mercaptopropionic acid, NAC) is an N-acetylated derivative of cysteine. It is also a talented addition of L-stereoisomers, D-stereoisomers, and one stereoisomer. The term "composition or compound with addition of stereoisomers" comprises at least 95%, preferably at least 97% by weight of the single stereoisomer of said compound. Any of these forms of NAC are delivered as antioxidants in the present invention. In one example, single isomers of thioethers or thioesters of NAC or its salts, preferably naturally occurring L-stereoisomers, are used in the method of treatment. N-acetylcysteine exhibits antioxidant activity (Smilksten, Knapp, Kuling and Rumack, N. Engl. J. Med. 1988, Vol 319, pp. 1557-62; Knight, KR, MacPhadyen, K., Lepore, DA, Kuwata, N., Eadie, PA, O'Brien, B. clinical Sci., 1991, Vol. 81, pp. 31-36; Ellis, EF, Dodson, LY, Police, RJ, J. Neurosurg., 1991, Vol. 75, pp. 774-779). Sulfahydryl functional groups are well known in nature and are free radical scavengers with high reactivity. N-acetylcysteine is known to promote the production of glutathione (tri-peptide also known as glutamylcysteinylglycine), which is important for keeping cellular components in a reduced state (Berggren, M,., Dawson, J., Moldeus, P. FEBS Lett., 1984, Vol. 176, pp. 189-192). The production of glutathione may enhance the activity of glutathione peroxidase, an enzyme that inactivates hydrogen peroxide, known as a precursor of hydroxy radicals (Lalitha, T., Keren, D., Yanni, S., Pharmacology and Toxicololgy, 1990, Vol. 66, pp. 56-61). N-acetylcysteine exhibits low toxicity in vivo, and has a fairly low deprinilboza toxicity (eg, LD 50 is 1140 and 81 mg / kg for N-acetylcysteine and deprinil, respectively, intravenously in rats). .). N-acetylcysteine and derivatives thereof are described, for example, in WO95 / 26719. The derivatives described in this publication can be used according to the present invention. D) Peroxide Scavenger, including but not limited to catalase and pyruvate E) thiols, including dithiothreitol and 2-mercaptoethanol F) antioxidants that are inhibitors of lipid peroxidation, including but not limited to TroloxTM, BHA, BHT, aminosteroid antioxidants, tocopherols and their analogs and lazaroids G) Dietary antioxidants comprising oxidant vitamins (vitamin C, E, or synthetic or natural pharmacological precursors or their analogs), either alone or in combination with flavonoids, phenolic compounds, caratenoids and alpha lipoic acid, alone or in combination. H) Inhibitors of lipoxygenase and cyclooxygenase, including but not limited to nonsteroidal anti-inflammatory drugs, COX-2 inhibitors, aspirin-based compounds and quercetin I) Antioxidants prepared by the body, including but not limited to ubiquinol and thiol antioxidants, glutathione, Se and lipoic acid J) Synthetic Phenolic Antioxidants: Inducers of Page I and II Drug Metabolizing Enzymes III anticancer drugs The term "anticancer agent" as used herein refers to any substance that reduces abnormal cell proliferation. Anticancer agents are described in various documents and include Martindale, "The Extra Pharmacopoeia, 31th Edition, Royal Pharmaceutical Society (1996). Anticancer agents include: (i) antifolates; (ii) antimetabolites (purine antimetabolites, cytarabine, fudarabine, phloxuridine, 6-mercaptopurine, methotrexate, 5-fluoropyrimidine, including 5-fluorouracil, beta-L-1 Including cytidine and 6-thiogluanine, such as, 3, -dioxoanyl cytidine); (iii) hydroxyurea; (iv) mitotic inhibitors (including CPT-11, etoposide (VP-21), taxol and vincristine); (v) alkylating agents, including but not limited to busulfan, chloroambucil, cyclophosphamide, ifopamide, mechloretamine, melphalan, and thiotepa; (vi) nonclassical alkylating agents, platinum-containing compounds, pleomycins, anti-tumor antibiotics, antitracycline, anthracenedione, topoisomer II inhibitors, hormones (corticosteroids, dexamethasone, pridnis) Hand, and methylpridnisone); And (vii) androgens such as fluoromesterone and methyltestosterone, estrogens such as diethylstilbestol, antiestrogens such as tamoxifen, LJRH homologues such as leuprolide, antiandrogens such as flutamide, aminoglutetide, Megestrol acetate and hydroxyprogensterone, asparaginase, carmustine, romustine, hexamethylmelanin, dacarbazine, mitotan, streptozosin, cisplatin, carboplatin, levamasol, and leuboline. A more comprehensive list of anticancer agents includes: the anticancer agent is aceglacon; Aclarubicin; Altretamine; Aminoglutetimides; 5-aminoglybulic acid; Amsacrine; Anastrozole; Ancitabine hydrochloride; 17-1A antibody; Antilymphocyte immunoglobulins; Anticancer agent A10; Asparinase; Pegaspargase; Azacytidine; Azathioprine; Batimastad; Benzoporphyrin derivatives; Bicalutamide; Bisantrene hydrochloride; Bleomycin; Sulfates; Brequinar Sodium; Broxuridine; Busulfan; Campath-IH; Carracemide; Carbetimers; Carboplatin; Carbocuone; Carmorpher; Carmustine; Chlorambucil; Chlorozotocin; Chromomycin; Cisplatin; Cladribine; Corinbacterium Parum; Cyclophosphamide; Cyclosporin; Cytarabine; Dacarbazine; Dactinomycin; Daunorubicin hydrochloride; Decitabine; Divajikuon; Dichlorodiethyl sulfide; Didemnin B; Docetaxel; Doxyfluidine; Doxorubicin hydrochloride; Droloxifene; Dekinomycin; Dedacrexate; Elftinium; Elmustine; Enroplatin; Enositabine; Epirubicin hydrochloride; Etamustine sodium phosphate; Ethanidazol; Etogluside; Etoposide; Padrosol hydrochloride; Kazarabine; Fenretinide; Phloxuridine; Fludarabine phosphate; Fluorouracil; Flutamide; Forestane; Potemustine; Gallium nitrate; Sencitabine; Gusperimus; Homoharingtonin; Hydroxyurea; Idarubicin hydrochloride; Ifosfamide; Monomorphine; Improsulfan tosylate; Inolimob; Interleukin-2; Irinotecan; JM-216; Letrozole; Lithium gamolenate; Lovaplatin; Romustine; Rodidamine; Mapposamide; Melparan; Menogaryl; Mercaptopurine; Methotrexate; Methotrexate sodium; Miboplatin; Miltefosine; Misimidazole; Mitobrotol; Mitoguazone dihydrochloride; Miso bean; Fur mall; Multialkyl peptides; Muromonab-CD3; Mustin hydrochloride; Mycophenolic acid; Mycophenolate mofetil; Nedalplatin; Nilutamide; Nimustine hydrochloride; Oxaliplatin; Paclitaxel; PCNU; Phenostatin; Peplomycin sulfate; Fifobroman; Pyrarrubicin; Pyritrexime; Isethionate; Pyroxanthrone hydrochloride; Plicamycin; Porpimer sodium; Friednimustine; Procarbazine hydrochlorite; Raltitrexed; Rannimustine; Lakamic acid; Rogletimide; Loquinimax; Sebriplatin; Semustine; Sirolimus; Sizopyran; Small aliphatic acid; Sodium bromebrate; Spartic acid; Sparfosate sodium; Sertozosin; Sulofener; Tacrolimus; Tamoxifen; Tegaper; Teloxtron hydrochloride; Temozolomide; Teniposide; Testosterone; Tetrasodium nesotetraphenylporpin-sulfenate; Thioguanine; Thiinosine; Thiotepa; Topotecan; Toremifene; Treosulfan; Trimetrexate; Trophosphamide; Tumor metastasis factor; Ubenimex; Uramustine; Vinblastine sulfate; Vincristine sulfate; Vindesine sulfate; Vinorelbine tartrate; Borosol; Ginostatin; Zolimob aritox; And zorubicin hydrochloride. IV abnormal cell proliferation Antioxidants may be used to increase the cell death of anticancer agents against abnormal cell proliferative diseases, including but not limited to: Abnormal cell proliferation may include papillomas, adenomas, fibromas, chondromas, osteomas, lipomas, hemangiomas, lymphangiomas, leiomyomas, rhabdomyomas, meningiomas, neuromas, ganglion neuromas, birthmarks, pheochromocytoma, schwannomas, fibroadenomas, teratomas, hydatidiforms Benign tumors, including granules, Brenner tumors, South Asian blastomas, hilar cell tumors, vas deferens, mesothelial cell tumors, and hypoxic tumors; Renal cell carcinoma, prostate cancer, bladder cancer, adenocarcinoma, fibrosarcoma, cartilage cancer, bone cancer, fatty cancer, hematosarcoma, lymphatic carcinoma, smooth muscle cancer, rhabdomyocarcinoma, myeloid leukemia, red leukemia, multiple myeloma , Glioma, epidural hematoma, gonorrhea, bladder sarcoma lobe (CTCL), skin preferential or invasive skin tumor (eg, basal cell carcinoma, squamous cell carcinoma, melanoma, and Bowen's disease), Kaposi cancer, and Malignant tumors including premalignant and malignant diseases of mucosal tissues, central nervous system tumors (neuroblastoma); Hyperproliferative and total tumor injuries, including fungal sarcoma, psoriasis, dermatitis, rheumatoid arthritis, virus, infectious sequencing, realignment and malignant diseases of the female reproductive tract; Of course, specific conditions treatable with this method include colorectal cancer, ovarian cancer, bone cancer, kidney cancer, breast cancer, gastrointestinal cancer, pancreatic cancer, melanoma, hematopoietic tumors such as lymphoma, leukemia, plasmacytoma, multiple myeloma and amylodosis ( amylodosis). Antioxidants can also be used in combination with anticancer agents for treating cardiovascular hyperproliferative diseases such as post- and. V Pharmacological Composition By administering systemically or locally an effective amount of an antioxidant, optionally in combination with an anticancer agent, in the presence of a pharmacologically acceptable carrier or diluent, a host, including a mammal, particularly a human, suffering from any of the above diseases can be treated. Antioxidants may be administered before, simultaneously, or after treatment of the anticancer agent when used to increase the cytotoxic effect of the anticancer agent. Methods and dosages for the administration of anticancer agents are well known to those skilled in the art and are described in a number of documents including The Physician's Desk Reference, Martindale's The Extra Pharmacopeia and Goodman & Gilman's The Pharmacological Basis of Therapeutics, using standard methods. It's easy to decide. Effective doses of antioxidants are administered subcutaneously, intravenously, intramuscularly, intraperitoneally, parenterally, orally, submucosa, inhalation, transdermally using sustained-release patches, or topically, to treat a target disease. May be administered. In general, systemic dosages for all diseases described in the present specification range from 0.01 mg / kg to 500 mg / Kg per kg body weight per day in single or divided daily doses. Typical dosages for topical application range from 0.001 to 100% by weight of the active compound. The compound is administered for a sufficient time to alleviate undesirable symptoms and clinical signs associated with the disease to be treated. In order to deliver a therapeutic amount of a compound to a patient in vivo without serious toxic effects, the active compound is included in a pharmaceutically acceptable carrier or diluent. The concentration of active compound in the pharmaceutical composition depends on the adsorption, inactivation, and secretion rate of the drug as well as other factors known to those skilled in the art. Dosage levels will also depend on the severity of the disease to be alleviated. In addition, for particular subjects, specific dosage regimens will have to be determined over time, depending on the expert judgment of the person administering and supervising the composition and the patient, and the dosage ranges set out in this specification are merely exemplary and claimed. It is not intended to limit the actual or the scope of the composition. The active ingredient may be administered at one time, or divided into smaller dosages and at different intervals. The preferred mode of administration for systemic delivery is oral. Oral compositions will generally include a stable diluent or edible carrier. They will be packed into gelatin capsules or compressed into tablets. For oral therapeutic administration, the active compound is incorporated into excipients and used in the form of tablets, troches, and capsules. Pharmacologically compatible binders and / or auxiliaries are included as part of the composition. Tablets, pills, capsules, troches, and the like may include compounds having the following ingredients or similar properties; Binders such as microcrystalline cellulose, gum tragacanth or gelatin; Excipients such as starch or lactose, alginic acid, primogel, or corn starch and silver disintegrants; Lubricants such as magnesium stearate or steotes; Brightening agents such as colloidal silicon dioxide; Sweetening agents such as sucrose or saccharin; Flavorings such as peppermint, methylsalicylate or orange flavor. When the dosage unit is a capsule, the capsule may comprise a liquid carrier such as fatty oil in addition to the above substances. In addition, the dosage unit form may further include various other substances that may modify the physical form of the dosage unit, such as sugar coating, shellac, or other enteric agents. It can also be mixed with other active substances that do not impair the desired action, or with substances which can supplement the desired substance, such as antibiotics, antibacterial agents, anti-inflammatory agents, antiviral agents, or other immunosuppressive agents. Parenteral, endothelial, subcutaneous, or topically applied solutions or suspensions may comprise the following components: sterile diluents such as injectable water, saline, fixed oils, polyethylene glycols, glycerin, propylene glycol or other synthetic solvents ; Antibacterial agents such as benzyl alcohol or methylparabens; Antioxidants such as ascorbic acid or sodium bisulfite; Chelating agents such as ethylenediamineacetic acid; Buffers such as acetates, citrate or phosphates; Osmotic pressure regulators such as sodium chloride or sodium hydroxide. Parenteral preparations may be enclosed in ampoules made of glass or plastic, or may be included in disposable syringes or multidose vials. In the case of intravenous administration, preferred carriers are physiological saline, sterile water, Cremophor EL (trade name, BASF, Parsipany, NJ) or phosphate buffered saline (PBS). In a preferred embodiment, the active compound is prepared with a carrier to protect the compound from rapid removal from the body, such as a controlled release formulation comprising an implant and a microencapsulated delivery system. Biodegradable and biocompatible polymers such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparing such formulations will be apparent to those skilled in the art. The material can also be purchased commercially from Alza Corporation, Nova Pharmaceutical, Inc. Liposomal suspensions (liposomes that target cells infected with monoclonal antibodies against viral antibodies) are also preferred as pharmacologically acceptable carriers. According to methods known in the art, they may be prepared, for example, by the methods described in US Pat. No. 4,522,811, which is incorporated herein by reference in its entirety. Lipid preparations are prepared by dissolving lipids (e.g., stearol phosphatidyl ethanolamine, stearol phosphatidyl choline, arachidol phosphatidyl choline and cholesterol) suitable for inorganic solvents, followed by evaporation and leaving a thin lipid film dried on the surface of the container. Can be. The vessel is then shaken by hand to form a lipid-free material from the side of the vessel and the lipid aggregate is dispersed to produce a liposome suspension. Suitable carriers or carriers for topical application are often applied to conventional methods such as lotions, suspensions, ointments, creams, gels, sprays, tinctures, powders, pastes, sustained-release transdermal patches, rectal, capsular, nasal or oral mucosa. It may be a dragon suppository. In addition to those listed above for systemic administration, thickeners, emollients, and stabilizers can be used in topical compositions. Examples of thickening agents may include petrolatum, biswax, xanthan gum or wetting agents such as polyethylene, sorbitol, mineral oils, ranoyl and derivatives thereof or emollients such as squalene. Many solutions and ointments are commercially available, especially for eye applications. VI Example Embodiments The following examples are intended to illustrate various embodiments of the present invention and are not intended to limit the protection scope of the present invention. Example 1 HCT 116 and HCT15 human CRC cells were obtained from the Americanl type culture collection. J. Pietenpol (Vanderbilt University, TN) provided p21 WAF1 / CIP1 − / − tumor cells generated from HCT by T. Waldman and HPV E6-transfected HCT 116 was obtained from WS El-Deiry (University of Pennsylvania, PA) [ WSEl-Deiry et al., Cell 75, 817 (1993). All tumor cell lines used in this study were serum (FBS), non-essential amino acids, L-glutamine, and penicillic G sodium (100 U / ml) and streptomycin sulfate in high content glycos and 10% heat activated fetal bovine. (100 mg / ml) were incubated in supplemented Dulbecco's Modified Eagle's Medium (DMEM) (GIBCO BRI.) At 37 ° C. under 50% CO 2 air. Pyrrolidinedithiocarbamate (Sigma Chemical Co., MA), vitamin E homologue (6-hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic acid in attachment-independent culture: vit ( E) (Aldrich), 5-FU (Hoffmann-LaRoche Inc. Nutley, NJ), or 10 × 10 3 in DMEM supplemented with 1% FBS and 0.4% agar with the material to be tested to determine the effect of doxorubicin. Cell / 35 mm plates were plated with HCT 116 and HCT15 Colony numbers were quantified with Omnicon image analyzer after 10 days Colonies larger than 50 microns in diameter were positively counted 25-200 μM and 0.1- in preliminary experiments At 10 mM, pyrrolidinedithiocarbamate and vitamin E had no effect on CRC cell growth efficiency, showing nonspecific cell death at higher concentrations. Example 2 I. G. Nicoletti et al., J. Immunol. Methods. Cell membranes were lysed by the method described in 139, 271 (1991), nuclei stained with propidium iodide (50 mg / ml) and quantified with Becton Dickinson FACSORT fluorescence-activated cell sorter. The nuclear ratio was determined on each cell cycle with MODFIT-DNA analysis software. ApopTaq Plus In Situ Apoptosis Detection Kit (Oncor, Gaithersburg, MD) was used to measure apoptosis cells by fluorescence microscopy or flow cytometry as described by the manufacturer. In summary, digoxigenin-labeled nucleotides were added to the free 3′OH group of DNA generated by DNA fragmentation during killing with terminal deoxynucleotidyl transferase (TdT). Digoxigenin was detected with FTC-conjugated anti-digoxigenin antibodies. Analysis was performed with a fluorescence-activated cell sorter and FITC staining was visualized using a fluorescence microscope (Zeiss). Example 3 Intracellular H 2 O 2 levels were measured by flow cytometry using dihydroortamine 1234 (DHR) as a specific fluorescent dye probe (G.Rothe, A.Emmendorffer, A. Oser, J. Roesler, G. Valet, J. Immun.Methods 138, 133 (1991); JA Royall, H. Ischiropoulos, Arch. Biochem. Biophysics 302, 348 (1993). CRC cells were cultured in DMEM medium containing 1 mM DHR and pyrrolidinedithiocarbamate (70 μM) or vitamin E (3 mM) until 24 hours. After trypsin digestion, trypsin activity was stopped with 2% FBS in phosphate buffer and cells were fixed in 1% paraformaldehyde (Sigma). Cell rhodamine 123 fluorescence intensity of 1 × 10 4 cells was measured for each sample using a Becton-Dickenson FACS Vantage flow cytometer with an excitation source of 488 nm and emission wavelength of 580 nm. Histograms were analyzed using the PC-Lysis (Becton dckenson) software program. The background fluorescence from the empty wells was subtracted from each reading. Example 4 4-6 week old male athymic Balb / c nu / nu mice were obtained from Harlan Sparque-Dawley Company and isolated for more than 2 weeks prior to testing. Animal testing was conducted in accordance with group and federal protection rules. HCT 116 and HCT15 CRC cell lines were cultured in DMEM medium supplemented with 10% FBS as described above. Two consecutive trypsins were treated, centrifuged at 300 g for 5 minutes, washed twice to harvest cells and resuspended in sterile phosphate buffer. In 0.2 ml 1 × 10 6 cells were administered subcutaneously into the scapula of 7-10 week old male nude mice. Example 5 The maximum volume, width, and height were measured weekly to obtain tumor volume. Once tumors average 120-150 mm 3 , weekly pyrrolidindithiocarbamate70), vitamin E, 5-FU, or saline is administered iP, or in combination with pyrrolidinedithiocarbamate, vitamin E or 5-FU was administered to the animals for 6 weeks. As a crossover experiment, the combination of pyrrolidinedithiocarbamate and 5-FU (saline, pyrrolidinedithiocarbamate or 5-FU alone) after the treatment was administered for three weeks (except Vitamin E) Or pyrrolidinedithiocarbamate and 5-FU were discontinuously treated for 3 weeks remaining. In preliminary experiments, a single dose of pyrrolidinedithiocarbamate, vitamin E, 5-FU was administered for 30 days to establish an effective route of administration with LD50 (no data). Tumor volumes were recorded weekly until the end of the study. Example 6 Tumor cells were fixed overnight in 4% (v / v) paraformaldehyde and embedded in paraffin following standard histological procedures. BrDU staining is described by Holmgren et al., Nature Med. 1, 149 (1955)]. TdT labeling of fragmented DNA (TUNEL) was as described. The proliferation index (BrDU) and death index (TUNEL) were measured as the percentage of cells calculated under the microscope at 200 × magnification. Proliferative indices for HCT 116 and HCT15-induced variants (regardless of treatment) were 53.1 ± 5.2 and 63.1 ± 7.2, respectively. Example 7 For western blot analysis, 50 mM Tris-Cl pH 7.4, 300 mM Nacl, 2 mM EDTA, 0.5% Nonidet-40, 0.5 mM phenylmethylsulfonyl fluoride aprotinin (1 μg / ml), pepstatin (1 μg / ml) And lysed the cells in lupeptin (2 μg / ml). 100 mg of extract (as determined by Bradford assay) was applied to a 12% SDS-PAE gel and transferred to 0.2 μM pore nitrocellulose membrane (Schleicher and Schuell). Blots were detected with antibodies to P21WAF1 / CIPI, p53, p27 or C / EBPβ (Santa Cruz) at a final concentration of 0.1 μg / ml. After washing, the blots were incubated with donkey-anti-rabbit or goat-anti-mouse IgG-horseradish peroxadase conjugates and developed using Enhanced Chemiluminescence (amersham, Arlington Height, IL). Example 8 [M. RNA was extracted as described in Schwab, K. Alitalo, HE, Varmus, JM Bishop, Nature (Lond.) 303, 497 (1983)]. Poly (A) + mRNA was electrophoresed on a 1% (w / v) agarose formaldehyde gel, and (not shown by Coffey et al., Cancer Res. 47, 4590 (1987)). Was performed. B.vogelstein (John Hopkins Oncology Center, altimore, MD) provided human p21 WAF1 / CIPI cDNA probes and labeled [ 32 P] dCTP by any primer extension method. Hybridization and post-hybridization washes were performed at 43 ° C. IB15 was used as a control for the same loading and transfer [PE Danielson, et al., DNA 7, 261 (1988)]. Example 9 B. Vogelstein (W.El-Deiry, et al., Cell 75, 817 (1993)) provided human p21 WAF1 / CIPI promoter constructs. According to the manufacturer's instructions (GIBCO BRL), CRC cell lines were incubated with 50% confluence prior to transfection with CELLFECTIN. For all luciferase assays, the transfected whole DNA was kept constant with the addition of pBSKII + pCMV-base. L. Sealy (Vanderbilt University, TN) provided all pCMV-C / EBP expression vectors. pCMV-CAT was transfected as an internal control of gene expression. Twelve hours after transfection, selected cells were treated with 70 μΜ pyrrolidinedithiocarbamate. After 24 hours of treatment, cell lysates were prepared and prepared as described in A. Misra-Press, CSRim, H. Yao, MS Roberson, PJS Stork, J. Biol. Chem. 270, 14587 (1995)] was analyzed luciferase activity. Luciferase activity was standardized to CAT activity and the results were reported as activity multiples of baseline. Example 10 A 2.4Kb genome fragment comprising the human p21 WAF1 / CIPI cDNA initiation site at the 3 'end is subcloned into the Hind III site of the luciferase reporter vector and the designated internal p21 for the published p21 WAF1 / CIPI promoter sequence (GeneBank). PGL -2 basic (Promega) -p21 WAF1 / CIPI deletion variants (D2198-D1138) were prepared by PCR using WAF1 / CIPI primers. In each case, PCR products were subcloned into pGL2-base and analyzed by the double chain DNA sequencing. NF_IL6 recognition sites were mutated using Muta-Gene M13 In vitro Mutagenesis Kit (Bio-Rad, Hercules, CA). The presence of the desired TT to AA base pair changes was confirmed by DNA sequencing. 11 to implementation Complementary oligonucleotides corresponding to bases 1884-1904 were synthesized in wild-type and NF_IL6 variant p21WAF1 / CIPI promoter sequences (wild type, GTACTTAAGAAATATTGAAT and ATTCAATATTTCTTAAGTAC; variant; GTACAAAAGAAATATTGAAT and ATCAATATTTCTTTTGTAC). 200 ng of each oligonucleotide was endlabeled with 200 μCi γ- 32 Plabeled ATP and T4 polynucleotide kinase. The resulting endlabel oligos were conjugated and purified by gel. Conditions for nucleation and electrophoresis rate assay (EMSA) of CRC cells treated with antioxidants are described in Kalioff, et al., Science 253, 786 (1991). When antiserum was added, the nuclear extract and 2 μl C / EBPa, β, and δ polyclonal antibodies (Santa Cruz) were incubated at room temperature for 10 minutes before radiolabeled probes were added. Example 12 SoftAga was treated with two human colorectal cancer cell lines, HCT116 (wild type p53) and HCT15 (variant p53) while increasing the amount of pyrrolidinedithiocarbamate or vitamin E in an oncogenic in vitro model. Both @ and vitamin E resulted in a dose dependent decrease in non-adherent urinary growth of HCT 116 and HCT 15 cells (FIG. 1A). This analysis was extended to various tumor cell lines derived from colon (HCA-7, Dif, RKO, SW620), breast (MCF-7, MDA-MB23), and gastrointestinal (Hs 746T). At these concentrations, both antioxidants inhibited non-adherent growth of tumor cell lines tested independently of the state of P53 (Difi, RKO, data not shown). Treatment of HCT116 HCT15 CRC cells with pyrrolidinedithiocarbamate (70 μM) or vitamin E (3 mM) for 24 hours, followed by staining with propidium iodide and analyzed by flow cytometry, showed that both compounds were significantly reduced in G1 pickle. It has been shown to induce cell accumulation, suggesting that the cause of the growth inhibition effect of @ or vitamin E observed in soft agar is cell interruption and / or apoptosis (FIG. 1B). To investigate the relationship between these cell cycle interruptions and the antioxidant properties of these compounds, the intracellular redox potential (internal H 2 O 2 level) and the percentage of cells showing G1 cell cycle interruption or apoptosis (type 25 cytometry) ) Was quantified for 24 hours in antioxidant-treated cells. As shown in FIG. 1C, both @ and vitamin E significantly reduced internal H 2 O 2 levels in both cell lines, with @ being a more effective reducing agent. In addition, this decrease in H 2 O 2 levels is associated with G1 cell cycle induction and the appearance of TUNEL-positive cell nuclei in these cells. Treatment of HCT15 cells with membrane-passing antioxidant N-acetyl-L-cysteine (NAC) and dietary antioxidant vitamin C resulted in decreased H 2 O 2 and induced cell death (1D). Support the role of reactive coral species. Since antioxidants change intracellular redox levels through reactive oxygen species rather than H 2 O 2 , HCT15 cells were transfected with expression plasmids encoding human catalase. Overexpression catalase reduced levels and induced cell cycle interruption and apoptosis in these cells, involving H 2 O 2 as a major regulator of cell cycle effects observed in these antioxidant treated cells. In addition, to investigate whether antioxidants increase the cytotoxic effects of 5-FU and doxorubicin, the IC 50 values of each drug were determined by HCT 116 cultured in soft agar with or without @ (70 μM) or vitamin E (3 mM). And 15 cells of HCT15 (approximate IC 50 values for these compounds in both cell lines). Compared to cells treated with each of 5-FU and doxorubicin alone, @ or vitamin E reduced IC 50 levels for these two compounds (FIG. 1E). This effect is more pronounced at @, which probably indicates the magnitude of its potential reducing power. The mechanisms of cellular uptake and the mechanisms of 5-FU and doxorubicin are quite different. Thus, it seems that @ or vitamin E regulates cell death of 5-FU and doxorubicin through changes in this pathway. Example 13 The therapeutic efficacy of @ or vitamin E was investigated in vivo by culturing HCT 116 or HCT 15 as tumor xenografts in athymic mice. After establishing palable tumors (mean tumor volume 150 mm 3), saline was injected i.p. into animals weekly as @, vitamin E, and / or 5-FU or as a negative control. The results of HCT116 are shown in Figure 2A. Four weeks later, control mice were sacrificed to measure tumor volume following the procedure. Individually, @, vitamin E and 5-FU significantly reduced tumor volume compared to saline control. The addition of @ or vitamin E significantly increased the effect of 5-FU. In all nine animals completely removed from the field, no signs of regrowth of tumor 25 were observed after the combination treatment was discontinued for two months. Similar results were seen in lactose HCT 15-derived xenografts, except that combination therapy was more effective in these variant p53CRC cells. To further investigate the in vivo efficacy of established HCT 15-derived tumors @ and 5-FU, mice were crossed with combination therapy after significant differences were established in monotherapy (FIG. 2B). Untreated mice initially developed large tumors (2780 ± 257 mm 3) around three weeks. At this point, treatment of these mice with @ and 5-FU reduced the size of these advanced injuries (Week 6: 1184 ± 96 mm 3). By crossing the combination therapy (5-FU and @), the tumor size of mice initially receiving monotherapy alone was also reduced. Initially, tumor size decreased from 1864 ± 190 mm3 to 660 ± 82 mm3 and 1325 ± 210 mm3 to 637 ± 231 mm3 in animals treated with 5-FU and @ only. These results complement the in vitro findings and indicate that antioxidants significantly increased the efficacy of 5-FU in CRC cells. Judging by weight change or gross anatomical and microscopic examination of the major organs, no signs of agonist-induced toxicity were observed in mice. At necropsy, all tumors showed severe central erosion regardless of tumor size or therapy. Mice treated with @ and 5-FU excluded this treatment group from the analysis because the tumors no longer exist. Immunohistologic analysis of the remaining tumors showed a high proliferation index regardless of therapy. However, in both xenograft models, the death index increased approximately 5 times after vitamin E treatment (FIG. 2C). In contrast, the apoptosis index of 5-FU was higher in cells expressing wild type p53 compared to variant p53 (HCT 15), which supports the role of p53-mediated apoptosis in 5-FU apoptosis. Combination treatment with vitamin E and 5-FU further increased the death index in these tumors even in variant p53 genetic background. The clear synergistic effect of vitamin E and 5-FU-induced apoptosis on variant p53 HCT 15 cells suggests that antioxidants can reestablish apoptosis signaling pathways. Example 14 Modulation of G1 cell cycle interruption and subsequent cell death contributes to a number of cellular proteins, including p53 and cyclin-dependent kinase inhibitors such as p21 WAF1 / CIPI . According to Western blot analysis (FIG. 3A), @ had no effect on p53 or p27 protein for 24 hours in HCT 116 and HCT15 cells. In contrast, p21 WAF1 / CIPI protein and mRNA levels increase within one hour after @ treatment and are maintained for 24 hours (FIG. 1B). Induction of p21 WAF1 / CIPI mRNA by @ is independent of p53 because its antioxidant effect is not diminished in HCT 116 cells expressing papillomavirus (HPV) E6 in humans that inactivate p53 via ubiquitin-mediated proteolysis (FIG. 3B) (Scheffner, et al., Proc. Natl. Acad. Sci. USA 88, 5523 (1991); Crook, et al., Oncogene 6, 873 (1991)). A similar increase in p21 WAF1 / CIPI expression was observed in HCT 116 and HCT 15 cells treated with vitamin E. To determine whether expression of p21 WAF1 / CIPI by antioxidants is essential for these cell cycle interruptions, parental HCT116 cells or cells were treated with @ or vitamin E for 24 hours to target p21 WAF1 / CIPI (FIG. 3C). . In both cell types, antioxidant-mediated apoptosis was significantly reduced, indicating that p21 WAF1 / CIPI plays a pivotal role in antioxidant mediated apoptosis. Example 15 To determine whether the induction of p21 WAF1 / CIPI was dependent on the electronic activity of antioxidants, HCT 116 and HCT 15 were transfected with 2.4Kb fragments of the promoter of p21 WAF1 / CIPI linked to the luciferase reporter gene. Treatment of cells transformed with @ induced about 5-fold promoter activity of p21 WAF1 / CIPI in HCT 116 and HCT 15, consistent with p53-independent induction of mRNA and protein of p21 WAF1 / CIPI (FIG. 4A). . A series of deletions of this promoter illustrate that the @reactive element of the p21 WAF1 / CIPI promoter is between nucleotides 2078 and -1874 . Site-specific mutations disrupted this site, eliminating luciferase activity induction of @, indicating that NF_IL6 site is essential for @ -induced p21 WAF1 / CIPI transcription. Members of the CCAAT / enhancer binding protein (C / EBP) family of transcription factors recognize the NF_IL6 consensus sequence (S.Akira and T.Kishimoto, Immunol Rev. 127, 25 (1992); Landschulz et al., Genes Dev. 2 786 (1988); Cao et al., Supra 5,1553 (1991); Chang, et al., Mol. Cell. Biol. 10, 6642 (1990); Williams et al., Supra, 5, 1553 (1991); Akira et al., EMBO, J. 9, 1897 (1990); Poli et al., Cell 63, 25 643 (1990). These factors include basic DNA-binding regions adjacent to leucine zipper (bZIP) dimer-forming domains that facilitate the feminization of other bZIP proteins and homodimers or heterodimers. Interestingly, C / EBPa regulates better transcription of p21 WAF1 / CIPI and inhibits cell proliferation in mouse preadipocytes, although no association with apoptosis has been identified. Result of determination by the 24 hours HCT 116 and HCT 15 nuclear extract and p21 WAF1 / CIPI NF_IL6 cis mutant 32 P oligo-labeled electricity in nucleotides electrophoretic migration assay (EMSA) containing obtained from the p21 WAF1 / CIPI NF_IL6 (Fig. 4B Left panel), DNA binding activity increased after @ treatment. Not a oligonucleotide comprising the variant NF_IL6 consensus sequence (lane 3) but a 50-fold molar excess of an unlabeled oligonucleotide comprising the NF_IL6 common sequence (right panel: lane 2), which competed with the mobile complex It is specific for the NF_IL6 cis element. Supershift analysis of induced complexes is due to the interaction of the migrated complex with C / EBPβ (lane 5) and the NF_IL6 cisele, C / EBPβ (lane 5) and C / EBPα (lane 4) or C Not due to interaction with / EBPδ (lane 6). To determine whether C / EBPβ affects the transcriptional activity of p21 WAF1 / CIPI , eukaryotic expression plasmids encoding C / EBPα, β or δ were converted to HCT 116 with full-length p21 WAF1 / CIPI -luciferase promoter constructs. Or HCT15 cells were transfected (FIGS. 4C and 4D). Transfection of C / EBβ was strongly activated in a p21 WAF1 / CIPI dose-dependent manner, and mutation of NF_IL6 eliminated this stimulus. In contrast, C / EBP α, or δ did not stimulate p21 promoter activity. Finally, HCT15 cell lines safely transformed with human C / EBPβ cDNA in the sense and antisense directions under the control of the ecdysone inducible promoter were generated to investigate the functional role of C / EBPβ in the apoptosis signaling pathway. In order to eliminate the possibility that constitutively expressed C / EBPβ cDNA induces apoptosis, an ecdysone (murosterone A) -induced expression system (Invitrogen, Carlsobad, Calif.) Was used. Human C / EBPβ cDNA was subcloned with pIND at a convenient enzyme cleavage site. Structures comprising the sense and antisense C / EBPβ sequences were identified by double chain DNA sequencing. Prior to transfection, the pIND-C / EBPβ construct was linearized with PmeI and purified. HCT15 cells were transfected with 5 μg pVgRXR (Invitrogen) and 10 μg pIND-C / EBPb using CELLFECTIN according to the manufacturer's instructions. After 24 hours, cells were transferred to medium supplemented with 1 mg / ml Geneticin and 10 mg / ml Furomycin (GIBCO BRL) to select transfected clones. Two weeks later, antibiotic-resistant cells were subcloned with limited dilution. Expression of C / EBPβ protein and subsequent induction of p21 WAF1 / CIPI were determined, induced for 10 hours with 10 mysterone A and Western blot was performed. Three independent positive clones were used to analyze substantially the same results. Representative data of clones derived from each of these cell lines are shown in FIGS. 4E and F. C / EBβ overexpression increased p21 WAF1 / CIPI protein levels relative to the unstimulated baseline (FIG. 4E: insertion). In addition, C / EBβ induction increased the death index of these cells in the presence and absence of antioxidants. In addition, inhibition of C / EBPβ expression by antisense mRNA induction eliminated antioxidant-induced apoptosis in these cells (FIG. 4E). Increasing the killing index for 5-FU or doxorubicin in the presence of overexpressing C / EBPβ determined additional evidence that C / EBPβ induction regulates antioxidant effects on colon cancer cells (FIG. 4F). In the absence of C / EBPβ overexpression, 5-FU increased the killing index by 20%, but doxorubicin did not induce killing. When these cells induce C / EBPβ overexpression in the presence of 5-FU or doxorubicin, apoptosis is increased by 70 and 80%, respectively. Together these data indicate that the induction of killing by antioxidant mediated, at least in part, by p53-independent induction of p21 WAF1 / CIPI through activation of transcription factor C / EBPβ. Although recently reported that induction of NF-kB DNA binding activity in renal epithelium progresses apoptosis after serum degeneration, another transcription factor, NF_IL6, confers resistance to TNFα-mediated apoptosis. @ Regulated low NF-kB activity through inhibition of phosphorylation and degradation of proteasome-mediated inhibitors (IkBs). The amount of @ used in this study did not reduce any NF-kB DNA binding activity in these CRC cells. In addition, the recent induction of p21 WAF1 / CIPI increases NF-kB transcriptional activity, so that the antioxidant effect in these cells does not seem to be mediated by NF-kB activity. This study suggests that the induction of the electron factor, C / EBβ, makes CRC cells susceptible to chemo-mediated apoptosis. Activation of C / EBPβ induced p21 WAF1 / CIPI gene expression directly or indirectly in both colon cancer cell lines, leading to G1 cell cycle interruption and cell death. The antioxidant potency of @ and vitamin E, which induces this transcription factor independently of functional p53, has important biological consequences for the efficacy of DNA damaging agents. Both 5-FU and doxorubicin exhibit their cellular lethal effect through induction of DNA damage. This damage is an unknown mechanism that signals p53 induction, which in turn inhibits cell proliferation and apoptosis. Since mutations in p53 occur over 80% of advanced CRC tumors, these mutations are responsible for the relatively low response rates of advanced colon cancer tumors to DNA-damaging agents such as 5-FU. Although 5-FU is particularly effective in the treatment of local, wild-type p53, colon cancer, this success rate drops to 15-20% of patients with advanced, often mutated, p53-containing colon cancer. Thus, the ability of antioxidants (used throughout this study at doses obtainable in humans) to exceed the need for p53-mediated apoptosis may be combined with antioxidants and chemotherapy for advanced colon cancer and other solid tumors. Explain the usefulness of Example 16 Figures 5a and 5b is a measure of the effect of the test compound on the proliferation of HCT 116 and HCT 15, derived from athymic mice treated with a combination of saline, vitamin E, PDTC, 5-FU and vitamin E and 5-FU It is a histogram of BrDU-labeled cells (percent of total cell nuclei) of isolated colon cell xenografts. Figure 6a and 6b is a measure of the effect of the test compound on apoptosis, colon cells xenografts derived from athymic mice treated with a combination of saline, vitamin E, PDTC, 5-FU and vitamin E and 5-FU It is a histogram of TUNEL-positive cells (percent of total cell nuclei). Tumor tissue was fixed in 4% (v / v) paraformaldehyde overnight and buried in paraffin by standard histological methods. Sections were pretreated with 10 mM citrate buffer (pH 6.0) and reacted with PC 10 monoclonal antibody against BrDU (Boehringer Mannheim). TdT labeling of fragmented DNA was performed according to the manufacturer's instructions. Percentage of cells was counted under a 200x microscope to determine the growth index (BrDU) and death index (TUNEL). Example 17 As shown in Fig. 7, PDTC treatment induces C / EBPβ DNA binding activity by post-translational modification. (A) DKO-1 cells were treated with 70 μM PDTC for a period of time, and nuclear extracts were prepared from [γ- 32 P] -labeled p21-NF_IL6 oligonucleotides (lanes 1-9). Specificity Assay: Nuclear extract derived from lanes 10-12, DKO-1 cells treated with PDTC for 3 hours (lane 5), treated with excess unlabeled wild type oligonucleotide (lane 11) and variant oligonucleotide (12) A comparative control experiment was conducted. Supershift analysis was performed with lanes 13-15, C / EBPa (lane 13), β (lane 14) or δ (lane 15) polyclonal antibodies. (B) The same DKO-1 cells were treated with PDTC (70 μM) for some time. Poly (A) was analyzed and treatment related changes on C / EBβ mRNA were analyzed by Northern blot. IB15 appeared as a control of loading and metastasis. (C) The same DKO-1 culture was treated with PDTC (70 μM) in the presence of [ 32 P] orthophosphate. Prior to initiation (0 hours) or at certain times, C / EBPβ of the cytoplasm and nucleus portion was purified from the cells by immunoprecipitation. SDS-PAGE and magnetic radioactivity or Western blot analysis (100 μg / lane of whole cell protein) were used to analyze the localization of treatment-related C / EBPβ. (D) DKO-1 cells were incubated for 1 hour in the presence of PDTC (70 μM), followed by immunocytochemistry to detect differences in the method of fractionation of C / EBPβ. In all experiments, the cultures with prereversal serum or primary anti-C / EBPβ antiserum preincubated with in vitro transcribed C / EBPβ protein showed no fluorescence signal after treatment with the second Cy3-conjugated antibody. Representative photomicrographs show anti-C / EBPβ stained cells before and after PCTC treatment. Example 18 8 illustrates the effect of PDTC on cAMP levels and PKA activity. DKO-1 cells were treated with PDTC (70 μM) for some time. Cell lysates were prepared and analyzed for (A) intracellular cAMP levels or (B) PKA activity (see experimental method). The figures are expressed as pmol average ± s.e.m. Per μg of protein and represent representative values of three of four experiments. Example 19 9 shows that PDTC phosphorylates C / EBPβ at Ser 299 . (A) Anti internal C / EBPβ of [ 32 P] orthophosphate labeled DKO-1 cells (2mCi / ml.3h) treated with 0 μM (lane 1), 70 μM PDTC (lane 2), or 50 μM forskolin Immunoprecipitated with -C / EBPβ antibody. Labeled proteins were labeled with SDS-PAGE and autoradiography. (B) Tryptic phosphopeptide map of in vivo labeled epitope-tack C / EBPβ. Immunoprecipitated wild-type (WT) and variant (Ala 299 ) C / EBPβ were digested with trypsin from PDTC treated or untreated DKO-1 cells with antibodies to FLAG-epitope, and the phosphopeptide was isolated by electrophoresis and TLC. And indicated by an automatic radioactivity method. X 1,2 was phosphorylated continuously. Levels of phosphopeptide X 3 increased after treatment with PDTC in cells transfected with non-variant wild type proteins. The circle represents its origin. (C) Comparison of phosphorylation of in vivo phosphorylation of wild-type C / EBPβ and Ala substitution of C / EBPβ from cells treated with and without PDTC. Autoradioactivity (top) and C / EBβ immunoblot are shown. (D) Phosphorylation of Ser299 in C / EBPβ is essential for protein translocation to the nucleus. DKO-1 cells were transfected with PCMV-C / EBPβ (WT) or pCMV-C / EBPβ (Ala 299 ) and treated with PDTC for 3 hours. Immunohistochemistry showed C / EBPβ protein as described in the experimental method. Example 20 10 shows that PKA phosphorylation of C / EBβ is essential for positioning into the nucleus. (A) The same DKO-1 cells were treated with PDTC (0 μM or μ70M) for 3 hours. Poly (A) + mRNA and protein were isolated from each group, and changes related to the treatment on C / EBβ mRNA and protein levels were measured by Norte or Western blot methods. IB15 was shown as a control for the same loading and metastasis. (B) DKO-1 cells were treated for 3 hours with PDTC (0 μM or μ70M) or PDTC and mPKI (myristylized protein kinase A, 1 μM). Cells were fixed in paraformaldehyde and C / EBβ protein was visualized by immunofluorescence staining. Cells treated with mPKI alone could not induce nuclear localization of C / EBβ (data not shown). Example 21 FIG. 11 shows that PDTC inhibits carboxymethylation of catalytic subunitite of protein phosphatase 2A. DKO-1 cells were cultured in serum-containing medium containing [methyl- 3 H] S-adenosyl methionine and / or 70 μM PDTC for 3 hours. The cytoplasm or nuclear sections were prepared and immunoprecipitated C / EBβ using standard methods. Antigen / antibody complexes were separated by SDS-PAGE and the presence of PP2Ac was detected by fluorescence. PDTC inhibits methyltransferase in the nucleus and to a lesser extent the cytoplasm. Example 22 Figure 12 shows that PDTC inhibits the methyltransferase activity of PP2Ac. PP2A (a and c dimers) were incubated in the presence of [methyl- 3 H] S-adenosyl methionine with increasing concentrations of methyltransferase in PDTC and partially purified rats at 37 ° C. for 30 minutes. The reaction was terminated with SDS-sample buffer. Samples were separated by SDS-PAGE and visualized the presence of methylated PP2A catalytic subunits by fluorescence. As shown, PDTC selectively inhibits the activity of methyltransferase to carboxymethylate the catalytic subunit of PP2A in a dose dependent manner. Example 23 To determine the direct and specific effect of PDTC on PP2A activity, DKO-1 cells were first treated with 17M PDTC for 3 hours. Cell lysates were made and treated with the following reagents in the presence of phosphorylated C / EBPβ (phosphate group radiolabeled) at 37 ° C .; I2 (selective PP1 inhibitor), Okadaic acid (optional PP2A and PP1 inhibitor), PDTC, I2 and PDTC, and okadaic acid and PDTC. As shown in FIG. 13, PDTC maintains phosphorylated C / EBPβ by inhibiting phosphatase activity in DKO-1 extract. This effect is reversible by removing antioxidants. This result is consistent with the PDTC inhibition of PP2A phosphatase. In contrast, PP2 phosphatase specific inhibitor I2 does not prevent dephosphorylation of C / EBβ under the same conditions. As expected, the nonspecific phosphatase inhibitor, okadaic acid, inhibits all DKO-1 phosphatase activity to prevent dephosphorylation of C / EBPβ. These results indicate that antioxidants such as PDTC are specific inhibitors of a group of phosphatases, such as PP2A, involved in the dephosphorylation of C / EBPβ. Example 24 The effect of PDTC on cell proliferation or apoptosis in a number of normal and tumor cell lines was measured. IC 50 is shown as the concentration of PDTC that inhibits cell proliferation. The results are provided in Table 1. As shown, PDTC does not interfere with normal cell growth but substantially inhibits the growth of breast, gastric, bone and pancreatic cancer cells. Effect of PDTC on Cell Symptoms (IC 50 Required to Inhibit Cell Proliferation or Induce Apoptosis) Normal cellsEpidermal cells600 μM Primary colon cell500 μM Epithelium of Primary Mammals650 μM Intestinal Epithelial Cells of Untransformed Lap450 μM Breast cancer cellMCF-713 μM MCF-10WT5 μM MCF-10HRas5 μM MDA-MB23110 μM MDA-MB-46820 μM Gastrointestinal cancerHs 746 T35 μM N-8740 μM Bone cancerSaos-210 μM Pancreatic cancerAsPol70 μM PANC-175 μM BxPc3100 μM Example 25 To determine whether antioxidants induce apoptosis in normal cells, normal and cancer cells were incubated with 70 μM PDTC for 24 hours and DNA fragments were measured as control percentages. As shown in Tables 2 and 3, normal cell lines (primary colon cells) did not show significant DNA fragments after 24 exposure to PDTC, but tumor cells (wild type p53HCA-7, HCT 116, mutant p53 HCT15, DLD-1, DKO). -3 cells) showed substantial DNA fragments. In vitro, PDC induces death in CRC cells but not in normal cells (I) Cell typeDNA fragments after PDTC treatment (70 μM) (%)3 hours6 hours12 hours24 hours Primary colon cell101 ± 10109 ± 9107 ± 10130 ± 16 Wild type p53 HCA-7111 ± 13126 ± 17154 ± 19302 ± 35 HCT 116108 ± 11131 ± 21198 ± 23367 ± 49 In vitro, PDC induces death in CRC cells but not in normal cells (II) Cell typeDNA fragments after PDTC treatment (70 μM) (%) Primary colon cell101 ± 10109 ± 9107 ± 10130 ± 16 Variant p53 HCT 15145 ± 12259 ± 18673 ± 34979 ± 34 DLD-1213 ± 17296 ± 21712 ± 34876 ± 46 DKO-3223 ± 11478 ± 16896 ± 161116 ± 54 Darkness in the table: AOVA determined significantly different from untreated cells (P <0.01) Example 26 As shown in Tables 2 and 3, PDTC substantially reduced the toxicity of 5-FU in the small intestine and colon of the rat. These results indicate that PDTC not only increases the cellular killing effect of anticancer drugs but also alleviates normal cells exposed to the killing agents. Example 27 A novel multicomponent complex consisting of C / EBβ, PP2A and methyltransferase was isolated and first characterized. This complex plays an important role in the regulation of lower transcription, including but not limited to PP2A and cell division and apoptosis. Coimmunoprecipitation describes for the first time that the transcription factor C / EBPβ forms a complex with PP2Ac protein phosphatase. This novel complex plays a mechanical role in the regulation of phosphorylation of C / EBp by PP2A. In addition, the C / EBβ / PP2Ac complex has also been shown to include a methyltransferase that carboxymethylates the catalytic subunit of C / EBP. Aqueous extracts of rat brain were fractionated with phenyl-sepharose and analyzed for methyltransferase activity using an external PP2A heterodimer (AC complex). In addition, the active peak of methyltransferase was fractionated by source Q (strong anion exchange) and gel filtration chromatography. The partially purified methyltransferase shown in FIG. 14 shows the highest methyltransferase activity from the gel filtration column. This peak portion of methyltransferase activity was further subjected to DEAE (weak anion exchange) and monoQ (other strong anion exchange resin) columns. Both C / EBβ and PP2A were detectable after this additional step. Rat brain extracts were expressed as positive controls (C / EBβ and PP2Ac migrated at about 45 and 36 kDa on SDS-PAGE). Modifications and variations of the present invention will become apparent to those skilled in the art from the detailed description previously described. Such changes will be included within the scope of the following claims.
权利要求:
Claims (30) [1" claim-type="Currently amended] A method of enhancing the cytotoxic activity of an anticancer agent against abnormal cell proliferative diseases, comprising administering an effective amount of an anticancer agent to a host in need thereof, with an effective amount of an antioxidant to increase cell death. [2" claim-type="Currently amended] A method for reducing toxicity to an anticancer agent administered for the treatment of solid growth of abnormally proliferating cells, which comprises administering an antioxidant agent, such as before, or after administration of the anticancer agent. . [3" claim-type="Currently amended] A method of increasing the therapeutic index of an anticancer agent administered for the treatment of faithful growth of abnormally proliferating cells, comprising administering an antioxidant agent, such as prior to or after administration of the anticancer agent. [4" claim-type="Currently amended] A method of increasing nuclear localization of C / EBβ in a cell, comprising administering an antioxidant into the cell. [5" claim-type="Currently amended] A method of inhibiting carboxymethylation of a catalytic subunit of protein phosphatase 2A by methyltransferase acting on protein phosphatase 2A, comprising contacting methyltransferase with an amount of antioxidant sufficient to inhibit. [6" claim-type="Currently amended] A method for identifying a compound that increases the cell death of an anticancer agent, comprising analyzing the ability of the compound to promote phosphorylation at the Ser299 position of C / EBβ. [7" claim-type="Currently amended] A method for identifying a compound that increases the cell death of an anticancer agent, which comprises analyzing the ability of the compound to inhibit carboxymethylation of protein phosphatase 2A. [8" claim-type="Currently amended] X2 is a C / EBPβ amino acid at position 298, and X2 and X3 are peptide sequences having the form of X1-Arg-X2-Ser-X3, which is a side peptide sequence having substantial homology to C / EBPβ. [9" claim-type="Currently amended] The method of claim 1, wherein the abnormal cell proliferation is colorectal cancer. [10" claim-type="Currently amended] The method of claim 1, wherein said abnormal cell proliferation is breast cancer. [11" claim-type="Currently amended] A protein complex, consisting of C / EBβ, PP2A, and methyltransferase that carboxymethylates a subunit of PP2A and having a purity of at least 70%. [12" claim-type="Currently amended] A method of treating abnormal host cell proliferative disease in a host, comprising administering to the host a protein having substantial homology to C / EBPβ or C / EBPβ in phosphorylated or non-phosphorylated form. [13" claim-type="Currently amended] 13. The protein of claim 12, wherein the protein having substantial homology to C / EBPβ is X1-Arg-X2, wherein X2 is a C / EBPβ amino acid at position 298 and X2 and X3 are flanking peptide sequences that have substantial homology to C / EBPβ. Consisting or comprising a peptide sequence in the form of Ser-X3, wherein substantial homology refers to a protein or peptide sequence having at least 60% identity and having substantially the same function as a parent sequence, Way. [14" claim-type="Currently amended] Synthetic C / EBβ homologues having their analogs resistant to stabilized phosphate bonds or dephosphorylation. [15" claim-type="Currently amended] 16. The synthetic C / EBβ homolog of claim 15, wherein said homologues that are resistant to dephosphorylation are phosphoramidates or phosphonate homologs. [16" claim-type="Currently amended] The method of claim 1, 2, 3, 4, or 5, wherein the antioxidant is dithiocarbanate. [17" claim-type="Currently amended] The method of claim 16, wherein the dithiocarbonate is A-SC (S) -B structure, Wherein A is hydrogen or a pharmacologically acceptable cation, B is alkyl, akenyl, akynyl, alkaryl, aralkyl, haloalkyl, haloalkenyl, haloalkynyl, aryl, alkaryl, hydrogen, C 1-6 alkoxy-C 1-10 alkyl, C 1 -6 alkylthio-C 1-10 alkyl, NR 2 R 3 ,-(CHOH) nCH 2 OH, where n is 0, 1,2,3,4,5, or 6, alkylacetyl, aylpropionyl (CH 2 ) nCO 2 R 1, hydroxy (C 1-6 ) alkyl comprising alkylbutyryl, wherein one or more hydroxy groups are present at any one of the carbon atoms, Or NR 2 R 3 , wherein R 2 and R 3 are each independently alkyl ;-( CHOH) n (CH 2 ) nOH, wherein n is 0,1,2,3,4,5,6; -(CH 2 ) nCO 2 R 1 ,-(CH 2 ) nCO 2 R 4 ; Hydroxy (C 1-6 ) alkyl-; Alkenyl (including vinyl, allyl and CH 3 CH═CH—CH 2 —CH 2 ); Alkyl (CO 2 H), alkenyl (CO 2 H), alkynyl (CO 2 H) or aryl, wherein the aryl group is substitutable as described above, for example NO 2 , CH 3 , t-butyl, A CO 2 H, halo, p-OH group; Or R 2 and R 3 may constitute a bridge, such as — (CH 2 ) m —, where m is 3,4,5,6,7,8,9, or 10 and R 4 is acetyl, propionyl Alkyl, aryl, alkaryl, or aralkyl, including butyryl, or A heterocyclic or alkylheterocyclic group that can be partially or fully hydrogenated. [18" claim-type="Currently amended] The method according to claim 1, 2, 3, 4, 5 or 6, wherein the antioxidant is probucol or monoester or diester of probucol. [19" claim-type="Currently amended] 19. The method of claim 18, wherein one or two hydroxy groups of probucol are substituted with succinic acid, glutaric acid, adipic acid, suberic acid, sebacic acid, magellanic acid, or maleic acid. [20" claim-type="Currently amended] The method according to claim 1, 2, 3, 4, 5 or 6, wherein the antioxidant is 2,6-dialkyl-4-silylphenol. [21" claim-type="Currently amended] The method according to claim 1, 2, 3, 4, 5 or 6, wherein the antioxidant is N-acetyl cysteine. [22" claim-type="Currently amended] The method according to claim 1, 2, 3, 4, 5 or 6, wherein the antioxidant is a scavenger of peroxidase, thiol, inhibitor of lipid peroxidation, dietary antioxidant, lipooxygenase and cycloox A method selected from the group consisting of inhibitors of cigenase, biologically made antioxidants, and synthetic phenolic antioxidants. [23" claim-type="Currently amended] 8. The method of claim 1, 2, 3, 6 or 7, wherein the anticancer agent is selected from aceglacon; Aclarubicin; Altretamine; Aminoglutetimides; 5-aminoglybulic acid; Amsacrine; Anastrozole; Ancitabine hydrochloride; 17-1A antibody; Antilymphocyte immunoglobulins; Anticancer agent A10; Asparinase; Pegaspargase; Azacytidine; Azathioprine; Batimastad; Benzoporphyrin derivatives; Bicalutamide; Bisantrene hydrochloride; Bleomycin; Sulfates; Brequinar Sodium; Broxuridine; Busulfan; Campath-IH; Carracemide; Carbetimers; Carboplatin; Carbocuone; Carmorpher; Carmustine; Chlorambucil; Chlorozotocin; Chromomycin; Cisplatin; Cladribine; Corinbacterium Parum; Cyclophosphamide; Cyclosporin; Cytarabine; Dacarbazine; Dactinomycin; Daunorubicin hydrochloride; Decitabine; Divajikuon; Dichlorodiethyl sulfide; Didemnin B; Docetaxel; Doxyfluidine; Doxorubicin hydrochloride; Droloxifene; Dekinomycin; Dedacrexate; Elftinium; Elmustine; Enroplatin; Enositabine; Epirubicin hydrochloride; Etamustine sodium phosphate; Ethanidazol; Etogluside; Etoposide; Padrosol hydrochloride; Kazarabine; Fenretinide; Phloxuridine; Fludarabine phosphate; Fluorouracil; Flutamide; Forestane; Potemustine; Gallium nitrate; Sencitabine; Gusperimus; Homoharingtonin; Hydroxyurea; Idarubicin hydrochloride; Ifosfamide; Monomorphine; Improsulfan tosylate; Inolimob; Interleukin-2; Irinotecan; JM-216; Letrozole; Lithium gamolenate; Lovaplatin; Romustine; Rodidamine; Mapposamide; Melparan; Menogaryl; Mercaptopurine; Methotrexate; Methotrexate sodium; Miboplatin; Miltefosine; Misimidazole; Mitobrotol; Mitoguazone dihydrochloride; Miso bean; Fur mall; Multialkyl peptides; Muromonab-CD3; Mustin hydrochloride; Mycophenolic acid; Mycophenolate mofetil; Nedalplatin; Nilutamide; Nimustine hydrochloride; Oxaliplatin; Paclitaxel; PCNU; Phenostatin; Peplomycin sulfate; Fifobroman; Pyrarrubicin; Pyritrexime; Isethionate; Pyroxanthrone hydrochloride; Plicamycin; Porpimer sodium; Friednimustine; Procarbazine hydrochlorite; Raltitrexed; Rannimustine; Lakamic acid; Rogletimide; Loquinimax; Sebriplatin; Semustine; Sirolimus; Sizopyran; Small aliphatic acid; Sodium bromebrate; Spartic acid; Sparfosate sodium; Sertozosin; Sulofener; Tacrolimus; Tamoxifen; Tegaper; Teloxtron hydrochloride; Temozolomide; Teniposide; Testosterone; Tetrasodium nesotetraphenylporpin-sulfenate; Thioguanine; Thiinosine; Thiotepa; Topotecan; Toremifene; Treosulfan; Trimetrexate; Trophosphamide; Tumor metastasis factor; Ubenimex; Uramustine; Vinblastine sulfate; Vincristine sulfate; Vindesine sulfate; Vinorelbine tartrate; Borosol; Ginostatin; Zolimob aritox; And zorubicin hydrochloride. [24" claim-type="Currently amended] The method of claim 1, 2, 3 or 12, wherein said abnormal cell proliferation is a benign tumor. [25" claim-type="Currently amended] 13. The method of claim 1, 2, 3 or 12, wherein said abnormal cell proliferation is a malignant tumor. [26" claim-type="Currently amended] 13. The method of claim 1, 2, 3 or 12, wherein said abnormal cell proliferation is hyperproliferative or preneoplastic injury. [27" claim-type="Currently amended] The method of claim 1, 2, 3 or 12, wherein the abnormal cell proliferation is induced tumor, adenoma, fibroma, chondroma, osteoma, lipoma, hemangioma, lymphoma, leiomyoma, rhabdomyoma, meningioma, neuroma, ganglion neuroma, birthmark , Pheochromocytoma, schwannoma, fibroadenoma, teratoma, hydatidiform, granulosa, Brenner's tumor, South blastoma, hilar cell tumor, vasculature, mesenchymal cell tumor, subtractive tumor, kidney Renal cell carcinoma, prostate cancer, bladder cancer, adenocarcinoma, fibrosarcoma, cartilage cancer, bone cancer, fatty cancer, hematosarcoma, lymphatic carcinoma, smooth muscle cancer, rhabdomyocarcinoma, myeloid leukemia, red leukemia, multiple myeloma, Glioma, epidural hematoma, gonorrhea, bladder sarcoma lobe (CTCL), skin-priority or skin-infiltrating skin tumor, kaposi's cancer, and premalignant and malignant diseases of mucosal tissue, central nervous system tumor, fungal sarcoma, Psoriasis, dermatitis, rheumatoid arthritis, A method selected from the group consisting of viruses, infectious serial species, realignment and malignant diseases of the female reproductive tract. [28" claim-type="Currently amended] 13. The method of claim 1, 2, 3 or 12, wherein the abnormal cell proliferation is colon cancer, ovarian cancer, bone cancer, kidney cancer, breast cancer, gastric cancer, pancreatic cancer, melanoma and hematopoietic tumor. The method selected from the group consisting of. [29" claim-type="Currently amended] 13. The method of claim 1, 2, 3, or 12, wherein said abnormal cell proliferation is cardiovascular disease. [30" claim-type="Currently amended] 30. The method of claim 29, wherein the cardiovascular disease is restenosis after angioplasty.
类似技术:
公开号 | 公开日 | 专利标题 ES2685947T3|2018-10-15|Anti-aging agents Mittra et al.2000|Luteolin, an abundant dietary component is a potent anti-leishmanial agent that acts by inducing topoisomerase II-mediated kinetoplast DNA cleavage leading to apoptosis US20140336210A1|2014-11-13|Aryl urea compounds in combination with other cytostatic or cytotoxic agents for treating human cancers CN101242817B|2016-08-31|HIF1 alpha modulators purposes in treatment cancer Nutter et al.1991|Menadione: spectrum of anticancer activity and effects on nucleotide metabolism in human neoplastic cell lines US6057361A|2000-05-02|Formulations and methods of reducing toxicity of antineoplastic agents Rey et al.2000|Up-regulation of mitochondrial peripheral benzodiazepine receptor expression by tumor necrosis factor alpha in testicular leydig cells: possible involvement in cell survival JP5390184B2|2014-01-15|Improved treatment of anemia Wolvetang et al.1994|Mitochondrial respiratory chain inhibitors induce apoptosis Sedlacek et al.1996|Flavopiridol |, a new kinase inhibitor for tumor therapy US5457130A|1995-10-10|Eicosapentaenoic acid used to treat cachexia Jonassen et al.2000|Insulin administered at reoxygenation exerts a cardioprotective effect in myocytes by a possible anti-apoptotic mechanism US7867983B2|2011-01-11|Methods to protect skeletal muscle against injury JP4256679B2|2009-04-22|How to treat restenosis EP1255537B1|2006-04-19|Farnesyl protein transferase inhibitors for treating breast cancer US6255347B1|2001-07-03|Methods and compositions comprising R-ibuprofen Dexter et al.1985|Activity of a novel 4-quinolinecarboxylic acid, NSC 368390 [6-fluoro-2-|-3-methyl-4-quinolinecarboxylic acid sodium salt], against experimental tumors US6703382B2|2004-03-09|Small molecule inhibitors targeted at Bcl-2 US5821260A|1998-10-13|Treatment for atherosclerosis and other cardiovascular and inflammatory diseases KR100861428B1|2008-10-02|Fatty acid analogues for the treatment of cancer US7105561B2|2006-09-12|Use of etodolac for the treatment of prostate cancer US8614192B2|2013-12-24|Method for treating ocular cancer EP1558256B1|2011-01-05|Pyrroloquinoline quinone and a beta blocker for the treatment of ischemia or reperfusion injury EP2988767A1|2016-03-02|Agents for downregulation of the activity and/or amount of bcl-xl and/or bcl-w US5545614A|1996-08-13|Controlling nitrogen oxide concentrations to modulate skeletal muscle contraction
同族专利:
公开号 | 公开日 WO1999001118A2|1999-01-14| WO1999001118A3|1999-04-22| US20010049349A1|2001-12-06| WO1999001118A9|1999-05-20| JP2002511878A|2002-04-16| CA2294247A1|1999-01-14| EA200000087A1|2000-08-28| EP1019034A2|2000-07-19| AU8282798A|1999-01-25| CA2294247C|2004-10-26| CN1261803A|2000-08-02| US7071158B2|2006-07-04|
引用文献:
公开号 | 申请日 | 公开日 | 申请人 | 专利标题
法律状态:
1997-07-01|Priority to US88665397A 1997-07-01|Priority to US8/886,653 1997-11-11|Priority to US96749297A 1997-11-11|Priority to US8/967,492 1998-07-01|Application filed by 아테로제닉스, 인코포레이티드 2001-03-15|Publication of KR20010020611A
优先权:
[返回顶部]
申请号 | 申请日 | 专利标题 US88665397A| true| 1997-07-01|1997-07-01| US8/886,653|1997-07-01| US96749297A| true| 1997-11-11|1997-11-11| US8/967,492|1997-11-11| 相关专利
Sulfonates, polymers, resist compositions and patterning process
Washing machine
Washing machine
Device for fixture finishing and tension adjusting of membrane
Structure for Equipping Band in a Plane Cathode Ray Tube
Process for preparation of 7 alpha-carboxyl 9, 11-epoxy steroids and intermediates useful therein an
国家/地区
|