Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Br J Cancer ; 94(1): 85-92, 2006 Jan 16.
Article in English | MEDLINE | ID: mdl-16306877

ABSTRACT

Skin toxicity, a common drug-related adverse event observed in cancer patients treated with epidermal growth factor receptor (EGFR)-directed therapies is rarely seen with therapies targeting HER2. This study reports the significance of the EGFR and HER2 dimerization status in skin with regard to these dermatologic side effects. We demonstrate the differential effect of HER-directed therapies on the ligand driven activation status of EGFR, HER2 and MAPK in normal human epidermal keratinocytes. EGFR-directed therapies, such as gefitinib and cetuximab, inhibited ligand-induced activation of EGFR and MAPK in human keratinocytes. Pertuzumab, an antibody interfering with functional HER2 heterodimerization, failed to block ligand-induced HER signaling in primary keratinocytes. Using a novel proximity-based dimerization assay (eTagtrade mark) we show that EGFR homodimers are the predominant HER dimer pair in normal primary kertinocytes and in normal skin tissue from 16 patients with solid malignancies. The presence of [p]EGFR and [p]MAPK, but the absence of [p]HER2, demonstrates productive signaling via EGFR but not HER2 in human skin. These data illustrate the importance of the EGFR dimerization partner in human skin and suggests that inhibition of EGFR homodimer signaling rather than EGFR/HER2 heterodimer signaling maybe the key molecular event determining dermatologic toxicity discrepancies observed between EGFR-targeted versus HER2-targeted therapies.


Subject(s)
Antibodies, Monoclonal/adverse effects , Antineoplastic Agents/adverse effects , ErbB Receptors/physiology , Exanthema/chemically induced , Exanthema/physiopathology , Quinazolines/adverse effects , Receptor, ErbB-2/physiology , Antibodies, Monoclonal, Humanized , Cell Culture Techniques , Cetuximab , Dimerization , ErbB Receptors/drug effects , Gefitinib , Humans , Keratinocytes/physiology , Ligands , Receptor, ErbB-2/drug effects , Signal Transduction
2.
Proc Natl Acad Sci U S A ; 98(20): 11720-4, 2001 Sep 25.
Article in English | MEDLINE | ID: mdl-11573006

ABSTRACT

Neuronal injury in ischemic stroke is partly mediated by cytotoxic reactive oxygen species. Although the antioxidant ascorbic acid (AA) or vitamin C does not penetrate the blood-brain barrier (BBB), its oxidized form, dehydroascorbic acid (DHA), enters the brain by means of facilitative transport. We hypothesized that i.v. DHA would improve outcome after stroke because of its ability to cross the BBB and augment brain antioxidant levels. Reversible or permanent focal cerebral ischemia was created by intraluminal middle cerebral artery occlusion in mice treated with vehicle, AA, or DHA (40, 250, or 500 mg/kg), either before or after ischemia. Given before ischemia, DHA caused dose-dependent increases in postreperfusion cerebral blood flow, with reductions in neurological deficit and mortality. In reperfused cerebral ischemia, mean infarct volume was reduced from 53% and 59% in vehicle- and AA-treated animals, respectively, to 15% in 250 mg/kg DHA-treated animals (P < 0.05). Similar significant reductions occurred in nonreperfused cerebral ischemia. Delayed postischemic DHA administration after 15 min or 3 h also mediated improved outcomes. DHA (250 mg/kg or 500 mg/kg) administered at 3 h postischemia reduced infarct volume by 6- to 9-fold, to only 5% with the highest DHA dose (P < 0.05). In contrast, AA had no effect on infarct volumes, mortality, or neurological deficits. No differences in the incidence of intracerebral hemorrhage occurred. Unlike exogenous AA, DHA confers in vivo, dose-dependent neuroprotection in reperfused and nonreperfused cerebral ischemia at clinically relevant times. As a naturally occurring interconvertible form of AA with BBB permeability, DHA represents a promising pharmacological therapy for stroke based on its effects in this model of cerebral ischemia.


Subject(s)
Antioxidants/metabolism , Ascorbic Acid/pharmacology , Dehydroascorbic Acid/pharmacology , Dehydroascorbic Acid/pharmacokinetics , Neuroprotective Agents/pharmacology , Neuroprotective Agents/pharmacokinetics , Stroke/prevention & control , Animals , Biological Transport , Brain/drug effects , Brain/metabolism , Cerebrovascular Circulation/drug effects , Cerebrovascular Circulation/physiology , Dose-Response Relationship, Drug , Mice , Middle Cerebral Artery/physiology , Reperfusion , Time Factors , Treatment Outcome
3.
Clin Cancer Res ; 7(4): 962-70, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11309347

ABSTRACT

PURPOSE: We have synthesized a series of hybrid polar compounds that induce differentiation and/or apoptosis of various transformed cells. These agents are also potent inhibitors of histone deacetylases (HDACs). Pyroxamide (suberoyl-3-aminopyridineamide hydroxamic acid) is a new member of this class of compounds that is currently under development as an anticancer agent. We investigated the activity of pyroxamide as an inducer of differentiation and/or apoptosis in transformed cells. EXPERIMENTAL DESIGN AND RESULTS: Pyroxamide, at micromolar concentrations, induced terminal differentiation in murine erythroleukemia (MEL) cells and caused growth inhibition by cell cycle arrest and/or apoptosis in MEL, prostate carcinoma, bladder carcinoma, and neuroblastoma cells. Administration of pyroxamide (100 or 200 mg/kg/day) to nude mice at doses that caused little evident toxicity significantly suppressed the growth of s.c. CWR22 prostate cancer xenografts. Despite the potent growth-inhibitory effects of pyroxamide in this tumor model, serum prostate-specific antigen levels in control versus pyroxamide-treated mice were not significantly different. Pyroxamide is a potent inhibitor of affinity-purified HDAC1 (ID(50) = 100 nM) and causes the accumulation of acetylated core histones in MEL cells cultured with the agent. Human CWR22 prostate tumor xenografts from mice treated with pyroxamide (100 or 200 mg/kg/day) showed increased levels of histone acetylation and increased expression of the cell cycle regulator p21/WAF1, compared with tumors from vehicle-treated control animals. CONCLUSIONS: The findings suggest that pyroxamide may be a useful agent for the treatment of malignancy and that induction of p21/WAF1 in transformed cells by pyroxamide may contribute to the antitumor effects of this agent.


Subject(s)
Aminopyridines/pharmacology , Antineoplastic Agents/pharmacology , Cell Differentiation/drug effects , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Acetylation/drug effects , Aminopyridines/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Cell Division/drug effects , Cell Line, Transformed , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Disease Models, Animal , Enzyme Inhibitors/therapeutic use , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Hydroxamic Acids/therapeutic use , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Prostate-Specific Antigen/blood , Prostatic Neoplasms/blood , Prostatic Neoplasms/drug therapy , Treatment Outcome , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Cancer Res ; 60(21): 6134-41, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11085537

ABSTRACT

Treatment of metastatic prostate cancer with androgen-ablation often elicits dramatic tumor regressions, but the response is rarely complete, making clinical recurrence inevitable with time. To gain insight into therapy-related progression, changes in gene expression that occurred following androgen-deprivation of an androgen-dependent prostate tumor xenograft, CWR22, and the emergence of an androgen-independent tumor, CWR22-R, were monitored using microarray analysis. Androgen-deprivation resulted in growth arrest of CWR22 cells, as evidenced by decreased expression of genes encoding cell cycle components and basal cell metabolism, respiration and transcription, and the induced expression of putative negative regulatory genes that may act to sustain cells in a nonproliferative state. Evolution of androgen-independent growth and proliferation, represented by CWR22-R, was associated with a reentry into active cell cycle and the up-regulation of several genes that were expressed at low levels or absent in the androgen-dependent tumor. Androgen repletion to mice bearing androgen-independent CWR22-R tumors induced, augmented, or repressed the expression of a number of genes. Expression of two of these genes, the calcium-binding protein S100P and the FK-506-binding protein FKBP51, was decreased following androgen-deprivation, subsequently reexpressed in CWR22-R at levels comparable with CWR22, and elevated further upon treatment with androgens. The dysregulated behavior of these genes is analogous to other androgen-dependent genes, e.g., prostate-specific antigen and human kallikrein 2, which are commonly reexpressed in androgen-independent disease in the absence of androgens. Other androgen-responsive genes whose expression decreased during androgen-deprivation and whose expression remained decreased in CWR22 were also identified in CWR22-R. These results imply that evolution to androgen-independence is due, in part, to reactivation of the androgen-response pathway in the absence of androgens, but that this reactivation is probably incomplete.


Subject(s)
Androgens/physiology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Neoplasms, Hormone-Dependent/genetics , Prostatic Neoplasms/genetics , Androgens/deficiency , Animals , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Neoplasms, Hormone-Dependent/metabolism , Oligonucleotide Array Sequence Analysis , Prostatic Neoplasms/metabolism , Transplantation, Heterologous , Up-Regulation
5.
Cancer Res ; 60(18): 5165-70, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-11016644

ABSTRACT

Suberoylanilide hydroxamic acid (SAHA) is the prototype of a family of hybrid polar compounds that induce growth arrest in transformed cells and show promise for the treatment of cancer. SAHA induces differentiation and/or apoptosis in certain transformed cells in culture and is a potent inhibitor of histone deacetylases. In this study, we examined the effects of SAHA on the growth of human prostate cancer cells in culture and on the growth of the CWR22 human prostate xenograft in nude mice. SAHA suppressed the growth of the LNCaP, PC-3, and TSU-Pr1 cell lines at micromolar concentrations (2.5-7.5 microM). SAHA induced dose-dependent cell death in the LNCaP cells. In mice with transplanted CWR222 human prostate tumors, SAHA (25, 50, and 100 mg/kg/day) caused significant suppression of tumor growth compared with mice receiving vehicle alone; treatment with 50 mg/kg/day resulted in a 97% reduction in the mean final tumor volume compared with controls. At this dose, there was no detectable toxicity as evaluated by weight gain and necropsy examination. Increased accumulation of acetylated core histones was detected in the CWR22 tumors within 6 h of SAHA administration. SAHA induced prostate-specific antigen mRNA expression in CWR22 prostate cancer cells, resulting in higher levels of serum prostate-specific antigen than predicted from tumor volume alone. The results suggest that hydroxamic acid-based hybrid polar compounds inhibit prostate cancer cell growth and may be useful, relatively nontoxic agents for the treatment of prostate carcinoma.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Prostatic Neoplasms/drug therapy , Animals , Antineoplastic Agents/toxicity , Cell Death/drug effects , Cell Division/drug effects , Enzyme Inhibitors/toxicity , Growth Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Histones/metabolism , Humans , Hydroxamic Acids/toxicity , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Prostate-Specific Antigen/blood , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Transplantation, Heterologous , Tumor Cells, Cultured/drug effects , Vorinostat
6.
Cancer Res ; 60(9): 2317-22, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10811100

ABSTRACT

Androgen receptors (ARs) belong to the family of hormone receptors that are ligand-dependent transcription factors. Endocrine therapy provides effective treatment for prostate cancer until mutations arise that alter the ligand responsiveness of AR. In this study, structural models were developed for the functional domains of human AR by homology modeling from crystal structures of closely related nuclear receptors. These models were used to locate the sites of two frequently occurring mutations in prostate cancer. The substitutions that develop in LNCaP (threonine-->alanine at residue 877) and CWR22 (histidine-->tyrosine at residue 874) tumor cell lines are both located on helix 11 that forms part of the ligand-binding pocket. However, the results suggest that these mutations influence ligand responsiveness by completely different mechanisms. Residue 877 contacts the ligand directly, and substitution at this site alters the stereochemistry of the binding pocket. Thus, the LNCaP mutation apparently broadens the specificity of ligand recognition. In contrast, residue 874 is located down the helical axis, projects away from the ligand pocket, and does not contact ligand. The side chain of residue 874 lies in a cavity between helices 11 and 12. Substitution of tyrosine for histidine 874 in CWR22 tumors may affect a conformational change of helix 12 and, thus, influence binding of coactivator proteins and their regulatory effect on transcriptional activation.


Subject(s)
Ligands , Neoplasms, Hormone-Dependent/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptors, Androgen/chemistry , Receptors, Androgen/genetics , Amino Acid Sequence , Binding Sites , Humans , Male , Models, Molecular , Molecular Sequence Data , Mutation , Neoplasms, Hormone-Dependent/genetics , Prostatic Neoplasms/chemistry , Receptors, Androgen/metabolism , Receptors, Progesterone/chemistry , Receptors, Progesterone/metabolism , Sequence Homology, Amino Acid , Tumor Cells, Cultured
7.
Semin Oncol ; 27(6 Suppl 11): 53-63; discussion 92-100, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11236029

ABSTRACT

HER-2/neu is overexpressed in most epithelial malignancies. Lung cancer, prostate cancer, and ovarian cancer are common epithelial tumors in which clinical trials are currently in progress to explore the potential therapeutic role for monoclonal antibodies to HER-2/neu (trastuzumab [Herceptin; Genentech, Inc, South San Francisco, CA]). In preclinical studies with tumor cell lines, trastuzumab was found to have additive and synergistic effects with some chemotherapeutic agents. Clinical trials investigating combination chemotherapy with trastuzumab and a variety of chemotherapeutic agents are already in progress in lung cancer.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Genes, erbB-2 , Lung Neoplasms/drug therapy , Ovarian Neoplasms/drug therapy , Prostatic Neoplasms/drug therapy , Animals , Antibodies, Monoclonal, Humanized , Carcinoma, Non-Small-Cell Lung/genetics , Clinical Trials as Topic , Drug Screening Assays, Antitumor , Female , Humans , Lung Neoplasms/genetics , Male , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/genetics , Ovarian Neoplasms/genetics , Prognosis , Prostatic Neoplasms/genetics , Trastuzumab
8.
Semin Oncol ; 27(6 Suppl 11): 76-83; discussion 92-100, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11236032

ABSTRACT

The epidermal growth factor (also known as HER or ErbB) family of receptor tyrosine kinases are important mediators of cell growth, differentiation, and survival. At present there are 10 ligands that bind directly to epidermal growth factor, HER-3, or HER-4. Although none of these ligands bind directly to HER-2, it is recruited to these receptor complexes and also becomes activated. A monoclonal antibody directed against HER-2, 2C4, inhibits the association of HER-2 with other HER family members. Ligand-activated HER-2 may also play a role in cancers, particularly those that do not overexpress HER-2 at high levels. For example, when prostate cancers progress from an androgen-dependent to an androgen-independent phenotype, epidermal growth factor pathways are frequently activated. 2C4 will inhibit the growth of both androgen-dependent and androgen-independent prostate tumors grown as xenografts in athymic mice.


Subject(s)
Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Receptor, ErbB-2 , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Humans , Ligands , Male , Prostate-Specific Antigen/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/physiology , Trastuzumab
9.
J Natl Cancer Inst ; 91(21): 1869-76, 1999 Nov 03.
Article in English | MEDLINE | ID: mdl-10547394

ABSTRACT

BACKGROUND: Androgen withdrawal is a standard therapy for prostate cancer that results in a decrease in tumor volume and a decline in serum prostate-specific antigen in the majority of patients. To understand the factors associated with regression of prostate cancers after androgen withdrawal, we studied cell cycle regulator changes in the CWR22 human prostate cancer xenograft model. METHODS: Established tumors in nude athymic BALB/c mice were sampled at various times after androgen withdrawal and after the development of androgen independence. Changes in the expression of cell cycle regulators were categorized into early and mid-to-late events. RESULTS AND CONCLUSIONS: Early events included a decrease in androgen receptor expression, followed by a short-term increase in expression of the p53 and p21/WAF1 proteins and a marked decrease in the Ki67 proliferative index. Mid-to-late events included progressive and sustained increases in p27 and p16 protein expression, a decrease in retinoblastoma protein expression, and an increase in the transcription factor E2F1. Changes in apoptosis (programmed cell death) were not observed at any time after androgen withdrawal. These data suggest that androgen withdrawal results in a cell stress response, in which increased p53 protein produces a cell cycle arrest, without activation of p53-mediated apoptosis. The proliferative index is further decreased through the action of the cyclin-dependent kinase inhibitors p27 and p16. Androgen-independent sublines emerged 80-400 days after androgen withdrawal, and these sublines had variable growth phenotypes but were associated with mdm2 protein overexpression and increased expression of cyclin D1. These results indicate that tumor regression in this human prostate cancer model is due to cell cycle arrest rather than to apoptosis and that the emergence of androgen independence is associated with a release from cell cycle arrest.


Subject(s)
Muscle Proteins , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Androgens/metabolism , Animals , Antibodies, Monoclonal , Cell Cycle , Cyclin-Dependent Kinase Inhibitor p16 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Ki-67 Antigen/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Microfilament Proteins , Receptors, Androgen/metabolism , Time Factors , Transplantation, Heterologous , Tumor Suppressor Protein p53/metabolism
10.
J Clin Oncol ; 17(12): 3776-85, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10577849

ABSTRACT

PURPOSE: To evaluate a chemotherapy regimen that consisted of ifosfamide administered as an infusion with bolus carboplatin, and etoposide (ICE) supported by granulocyte colony-stimulating factor (G-CSF) for cytoreduction and stem-cell mobilization in transplant-eligible patients with primary refractory or relapsed non-Hodgkin's lymphoma (NHL). PATIENTS AND METHODS: One hundred sixty-three transplant-eligible patients with relapsed or primary refractory NHL were treated from October 1993 to December 1997 with ICE chemotherapy at Memorial Sloan-Kettering Cancer Center. Administration of three cycles of ICE chemotherapy was planned at 2-week intervals. Peripheral-blood progenitor cells were collected after cycle 3, and all patients who achieved a partial response (PR) or complete response (CR) to ICE chemotherapy were eligible to proceed to transplantation. Event-free and overall survival, ICE-related toxicity, and the number of CD34(+) cells collected after treatment with ICE and G-CSF were evaluated. RESULTS: All 163 patients were assessable for response, and there was no treatment-related mortality. A major response (CR/PR) was evident in 108 patients (66.3%); 89% of the responding patients underwent successful transplantation. Patient who underwent transplantation and achieved a CR to ICE had a superior overall survival to that of patients who achieved a PR (65% v 30%; P =.003). The median number of CD34(+) cells/kg collected was 8.4 x 10(6). The dose-limiting toxicity of ICE was hematologic, with 29.4% of patients developing grade 3/4 thrombocytopenia. There were minimal nonhematologic side effects. CONCLUSION: ICE chemotherapy, with ifosfamide administered as a 24-hour infusion to decrease CNS side effects, and the substitution of carboplatin for cisplatin to minimize nephrotoxicity, is a very effective cytoreduction and mobilization regimen in patients with NHL. Furthermore, the quality of the clinical response to ICE predicts for posttransplant outcome.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Hematopoietic Stem Cell Mobilization , Lymphoma, Non-Hodgkin/drug therapy , Adolescent , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carboplatin/administration & dosage , Cell Movement , Disease-Free Survival , Etoposide/administration & dosage , Etoposide/adverse effects , Female , Granulocyte Colony-Stimulating Factor/administration & dosage , Humans , Ifosfamide/administration & dosage , Leukocytes, Mononuclear/immunology , Lymphoma, Non-Hodgkin/therapy , Male , Middle Aged , Prognosis , Stem Cells/physiology
11.
Cancer Res ; 59(19): 4761-4, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10519379

ABSTRACT

Antibody to the Her-2/neu gene product has been shown to inhibit the growth of breast cancer cells overexpressing Her-2/neu and to have clinical utility in treating breast cancer. We studied a recombinant, humanized anti-Her-2/neu antibody (Herceptin) in preclinical models of human prostate cancer. The androgen-dependent CWR22 and LNCaP human prostate cancer xenograft models and androgen-independent sublines of CWR22 were used. Her-2/neu staining of the parental, androgen-dependent, and androgen-independent CWR22 tumors and LNCaP tumors demonstrated variable Her-2/neu expression. Herceptin was administered i.p. at a dose of 20 mg/kg twice weekly after the xenograft had been established. No effect of Herceptin on tumor growth was observed in any of the androgen-independent tumors; however, significant growth inhibition was observed in both of the androgen-dependent xenograft models, CWR22 (68% growth inhibition at the completion of the experiment; P = 0.03 for trajectories of the average tumor volume of the groups) and LNCaP (89% growth inhibition; P = 0.002). There was a significant increase in prostate-specific antigen (PSA) index (ng PSA/ml serum/mm3 tumor) in Herceptin-treated androgen-dependent groups compared with control (CWR22, 18-fold relative to pretreatment value versus 1.0-fold, P = 0.0001; LNCaP, 2.35-fold relative to pretreatment value versus 0.6-fold, P = 0.001). When paclitaxel (6.25 mg/kg s.c., five times/week) was given to animals with androgen-dependent and -independent tumors, there was growth inhibition in each group. Paclitaxel and Herceptin cotreatment led to greater growth inhibition than was seen for the agents individually. Thus, in these prostate cancer model systems, Herceptin alone has clinical activity only in the androgen-dependent tumor and has at least an additive effect on growth, in combination with paclitaxel, in both androgen-dependent and androgen-independent tumors. Response to Herceptin did not correlate with the PSA levels, because the PSA index markedly increased in the Herceptin-treated group, whereas it remained constant in the control group. These results suggest the utility of Herceptin in the treatment of human prostate cancer.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Prostatic Neoplasms/therapy , Receptor, ErbB-2/immunology , Animals , Antibodies, Monoclonal, Humanized , Female , Humans , Immunohistochemistry , Male , Mice , Mice, Nude , Paclitaxel/therapeutic use , Prostatic Neoplasms/pathology , Transplantation, Heterologous , Trastuzumab
12.
Cancer Res ; 59(18): 4555-8, 1999 Sep 15.
Article in English | MEDLINE | ID: mdl-10493506

ABSTRACT

Human tumors may contain high concentrations of ascorbic acid, but little is known about how they acquire the vitamin. Certain specialized cells can transport ascorbic acid directly through a sodium ascorbate cotransporter, but in most cells, vitamin C enters through the facilitative glucose transporters (GLUTs) in the form of dehydroascorbic acid, which is then reduced intracellularly and retained as ascorbic acid. Mice with established hematopoietic and epithelial cell xenografts were studied for the accumulation of injected ascorbic acid and dehydroascorbic acid. Most hematopoietic and epithelial tumor cell lines can only transport vitamin C in the oxidized form (dehydroascorbic acid) in vitro; however, when grown as xenografts in mice, they rapidly accumulated vitamin C after administration of radiolabeled ascorbic acid. The involvement of the GLUTs in vitamin C uptake by the xenografted tumors was demonstrated by competitive inhibition with D-glucose but not L-glucose. Because the malignant cells were not capable of directly transporting ascorbic acid, we reasoned that the ascorbic acid was oxidized to dehydroascorbic acid in the tumor microenvironment. Tumor accumulation of vitamin C in animals injected with ascorbic acid was inhibited by coadministration of superoxide dismutase, implying a role for superoxide anion in the oxidation of ascorbic acid. Whereas the epithelial cancer cell lines could not generate superoxide anion in culture, the minced xenograft tumors did. Our studies show the transport of dehydroascorbic acid by GLUTs is a means by which tumors acquire vitamin C and indicate the oxidation of ascorbic acid by superoxide anion produced by cells in the tumor stroma as a mechanism for generating the transportable form of the vitamin.


Subject(s)
Ascorbic Acid/metabolism , Breast Neoplasms/metabolism , Monosaccharide Transport Proteins/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Prostatic Neoplasms/metabolism , Stromal Cells/metabolism , Animals , Biological Transport/drug effects , Female , Glucose/pharmacology , Glucose Transporter Type 1 , HL-60 Cells , Humans , Male , Mice , Mice, Nude , Models, Biological , Transplantation, Heterologous
13.
Cancer Res ; 58(14): 3009-14, 1998 Jul 15.
Article in English | MEDLINE | ID: mdl-9679964

ABSTRACT

The current means of assessing response in prostate cancer are imprecise because changes in tumor size are often slow and difficult to document, and alterations in serum prostate-specific antigen (PSA) levels do not always correlate with clinical outcomes. Using the CWR22 human prostate cancer xenograft model, serial changes in prostate tumor metabolism were assessed after androgen withdrawal by [3H]deoxyglucose (DOG) accumulation in the tumors and noninvasively using positron emission tomography (PET) with 2-[18F]-fluoro-2-deoxy-D-glucose as a radiotracer. A significant decrease in [3H]DOG accumulation was observed at 48 h after androgen withdrawal [62% of preandrogen withdrawal value (95% confidence interval: 0.59, 0.65)], reaching a maximum decline at day 10 [38% of preandrogen withdrawal value (95% confidence interval: 0.37, 0.40)]. Using PET, parallel changes in tumor metabolism were demonstrated and preceded changes in tumor volume and marked declines in serum PSA. DOG accumulation returned to near baseline upon reintroduction of androgen. The decrease in DOG accumulation was associated with a decline in the proportion of tumor cells in active cell cycle from >60% to <5% at 7-10 days after androgen withdrawal. No increase in the proportion of tumor cells undergoing apoptosis was observed during this time period, implying an arrest in a G0/early G1 state. DOG accumulation in tumor cells, measured directly and by PET, correlated with androgen changes in the host. The results suggest that serial monitoring of DOG accumulation using PET scanning may be useful as an early indicator of treatment efficacy and other outcome measures in prostate cancer.


Subject(s)
Deoxyglucose/metabolism , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/metabolism , Androgen Antagonists/pharmacology , Animals , Antineoplastic Agents, Hormonal/pharmacology , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Prostatic Neoplasms/pathology , Tomography, Emission-Computed , Transplantation, Heterologous
15.
J Clin Invest ; 100(11): 2842-8, 1997 Dec 01.
Article in English | MEDLINE | ID: mdl-9389750

ABSTRACT

Vitamin C concentrations in the brain exceed those in blood by 10-fold. In both tissues, the vitamin is present primarily in the reduced form, ascorbic acid. We identified the chemical form of vitamin C that readily crosses the blood-brain barrier, and the mechanism of this process. Ascorbic acid was not able to cross the blood-brain barrier in our studies. In contrast, the oxidized form of vitamin C, dehydroascorbic acid (oxidized ascorbic acid), readily entered the brain and was retained in the brain tissue in the form of ascorbic acid. Transport of dehydroascorbic acid into the brain was inhibited by d-glucose, but not by l-glucose. The facilitative glucose transporter, GLUT1, is expressed on endothelial cells at the blood-brain barrier, and is responsible for glucose entry into the brain. This study provides evidence showing that GLUT1 also transports dehydroascorbic acid into the brain. The findings define the transport of dehydroascorbic acid by GLUT1 as a mechanism by which the brain acquires vitamin C, and point to the oxidation of ascorbic acid as a potentially important regulatory step in accumulation of the vitamin by the brain. These results have implications for increasing antioxidant potential in the central nervous system.


Subject(s)
Ascorbic Acid/pharmacokinetics , Blood-Brain Barrier , Brain/metabolism , Dehydroascorbic Acid/pharmacokinetics , Monosaccharide Transport Proteins/metabolism , Animals , Autoradiography , Brain/diagnostic imaging , Capillary Permeability , Deoxyglucose/pharmacology , Glucose Transporter Type 1 , Image Processing, Computer-Assisted , Kinetics , Mice , Mice, Inbred BALB C , Oxidation-Reduction , Radiography , Rats , Rats, Inbred F344 , Time Factors
16.
J Exp Med ; 173(5): 1039-46, 1991 May 01.
Article in English | MEDLINE | ID: mdl-2022918

ABSTRACT

To seek information on the capacity of mature T cells to migrate to the thymus, mice were injected with Thy-1-marked populations enriched for resting T cells or T blast cells; localization of the donor cells in the host thymus was assessed by staining cryostat sections of thymus and by FACS analysis of cell suspensions. With injection of purified resting T cells, thymic homing was extremely limited, even with injection of large doses of cells. By contrast, in vivo generated T blast cells migrated to the thymus in substantial numbers. Thymic homing by T blasts was greater than 50-fold more efficient than with resting T cells. Blast cells localized largely in the medulla and remained in the thymus for at least 1 mo post-transfer. Interestingly, localization of T blasts in the thymus was 10-fold higher in irradiated hosts than normal hosts. Thymic homing was especially prominent in mice injected with T blasts incubated in vitro with the DNA precursor, 125I-5-iodo-2'deoxyuridine (125IDUR); with transfer of 125IDUR-labeled blasts to irradiated hosts, up to 5% of the injected counts localized in the host thymus. These data suggest that thymic homing by T blasts might be largely restricted to cells in S phase. The physiological significance of blast cell entry to the thymus is unclear. The possibility that these cells participate in intrathymic tolerance induction is discussed.


Subject(s)
Lymphocyte Activation/physiology , T-Lymphocytes/physiology , Thymus Gland/physiology , Animals , Cell Movement/physiology , DNA/metabolism , Idoxuridine/metabolism , Iodine Radioisotopes , Lymph Nodes/cytology , Lymph Nodes/physiology , Mice , Receptors, Lymphocyte Homing/metabolism , Receptors, Lymphocyte Homing/physiology , S Phase/physiology , T-Lymphocytes/metabolism , T-Lymphocytes/ultrastructure , Thymus Gland/cytology , Thymus Gland/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...