Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
2.
Ann Oncol ; 30(11): 1821-1830, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31504139

ABSTRACT

BACKGROUND: The National Cancer Institute-Molecular Analysis for Therapy Choice (NCI-MATCH) is a national precision medicine study incorporating centralized genomic testing to direct refractory cancer patients to molecularly targeted treatment subprotocols. This treatment subprotocol was designed to screen for potential signals of efficacy of ado-trastuzumab emtansine (T-DM1) in HER2-amplified histologies other than breast and gastroesophageal tumors. METHODS: Eligible patients had HER2 amplification at a copy number (CN) >7 based on targeted next-generation sequencing (NGS) with a custom Oncomine AmpliSeq™ (ThermoFisher Scientific) panel. Patients with prior trastuzumab, pertuzumab or T-DM1 treatment were excluded. Patients received T-DM1 at 3.6 mg/kg i.v. every 3 weeks until toxicity or disease progression. Tumor assessments occurred every three cycles. The primary end point was centrally assessed objective response rate (ORR). Exploratory end points included correlating response with HER2 CN by NGS. The impact of co-occurring genomic alterations and PTEN loss by immunohistochemistry were also assessed. RESULTS: Thirty-eight patients were enrolled and 36 included in efficacy analysis. Median prior therapies in the metastatic setting was 3 (range 0-9; unknown in one patient). Median HER2 CN was 17 (range 7-139). Partial responses were observed in two (5.6%) patients: one mucoepidermoid carcinoma of parotid gland and one parotid gland squamous cell cancer. Seventeen patients (47%) had stable disease including 8/10 (80%) with ovarian and uterine carcinomas, with median duration of 4.6 months. The 6-month progression-free survival rate was 23.6% [90% confidence interval 14.2% to 39.2%]. Common toxicities included fatigue, anemia, fever and thrombocytopenia with no new safety signals. There was a trend for tumor shrinkage with higher levels of gene CN as determined by the NGS assay. CONCLUSION: T-DM1 was well tolerated. While this subprotocol did not meet the primary end point for ORR in this heavily pre-treated diverse patient population, clinical activity was seen in salivary gland tumors warranting further study in this tumor type in dedicated trials.


Subject(s)
Ado-Trastuzumab Emtansine/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Biomarkers, Tumor/genetics , Neoplasms/drug therapy , Receptor, ErbB-2/genetics , Ado-Trastuzumab Emtansine/pharmacology , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Immunological/pharmacology , Drug Resistance, Neoplasm/genetics , Female , Gene Amplification , Humans , Middle Aged , National Cancer Institute (U.S.) , Neoplasms/genetics , Neoplasms/mortality , Neoplasms/pathology , Precision Medicine/methods , Progression-Free Survival , Receptor, ErbB-2/antagonists & inhibitors , United States/epidemiology
3.
Clin Pharmacol Ther ; 101(5): 616-618, 2017 May.
Article in English | MEDLINE | ID: mdl-27981567

ABSTRACT

As part of the White House Cancer Moonshot Initiative, the National Cancer Institute (NCI) has developed a drug formulary to provide investigational anticancer agents to the extramural research community. This article describes how the NCI Formulary functions, how researchers may apply for access to drugs in the formulary, and the NCI's initial goals for formulary participation. Approved investigators may apply for access to formulary agents at: https://nciformulary.cancer.gov.


Subject(s)
Antineoplastic Agents , Drugs, Investigational , Formularies as Topic , National Cancer Institute (U.S.) , Public-Private Sector Partnerships , Humans , Neoplasms/drug therapy , United States
4.
Br J Cancer ; 106(1): 77-84, 2012 Jan 03.
Article in English | MEDLINE | ID: mdl-22134508

ABSTRACT

BACKGROUND: Preclinical studies suggest that histone deacetylase (HDAC) inhibitors may restore tumour sensitivity to retinoids. The objective of this study was to determine the safety, tolerability, and the pharmacokinetic (PK)/pharmacodynamic (PD) profiles of the HDAC inhibitor entinostat in combination with 13-cis retinoic acid (CRA) in patients with solid tumours. METHODS: Patients with advanced solid tumours were treated with entinostat orally once weekly and with CRA orally twice daily × 3 weeks every 4 weeks. The starting dose for entinostat was 4 mg m(-2) with a fixed dose of CRA at 1 mg kg(-1) per day. Entinostat dose was escalated by 1 mg m(-2) increments. Pharmacokinetic concentrations of entinostat and CRA were determined by LC/MS/MS. Western blot analysis of peripheral blood mononuclear cells and tumour samples were performed to evaluate target inhibition. RESULTS: A total of 19 patients were enroled. The maximum tolerated dose (MTD) was exceeded at the entinostat 5 mg m(-2) dose level (G3 hyponatremia, neutropenia, and anaemia). Fatigue (G1 or G2) was a common side effect. Entinostat exhibited substantial variability in clearance (147%) and exposure. CRA trough concentrations were consistent with prior reports. No objective responses were observed, however, prolonged stable disease occurred in patients with prostate, pancreatic, and kidney cancer. Data further showed increased tumour histone acetylation and decreased phosphorylated ERK protein expression. CONCLUSION: The combination of entinostat with CRA was reasonably well tolerated. The recommended phase II doses are entinostat 4 mg m(-2) once weekly and CRA 1 mg kg(-1) per day. Although no tumour responses were seen, further evaluation of this combination is warranted.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Histone Deacetylase Inhibitors/therapeutic use , Neoplasms/drug therapy , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Benzamides/administration & dosage , Blotting, Western , Chromatography, Liquid , Female , Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylase Inhibitors/adverse effects , Humans , Isotretinoin/administration & dosage , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/metabolism , Pyridines/administration & dosage , Tandem Mass Spectrometry , Treatment Outcome
6.
Cancer Gene Ther ; 8(9): 636-48, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11593332

ABSTRACT

Cancer metastasis accounts for a significant proportion of morbidity and mortality in patients. Effective means of treating disseminated disease remains elusive. The purpose of this study was to determine whether genetically modified endothelial cells (GMEC) can selectively target and deliver recombinant therapeutic molecules to sites of tumor metastases. Following the establishment of lung metastases of 4T1 mammary tumor in mice, intravenously (i.v.) administered, lacZ transgene-expressing endothelial cells (lacZ-GMEC) accumulated at the tumor sites. An average of 32% and 90% of the pulmonary metastases were X-gal stained following one and three tail vein injections of 10(5) lacZ-GMEC, respectively. The linear pattern of X-gal staining seen within the tumor sites and the histological appearance of the tumor vasculature were consistent with the incorporation of lacZ-GMEC into blood vessels. In C57Bl/6 mice harboring lung metastases of melanoma, the administration of three sequential i.v. injections of 10(5) endothelial cells expressing a human interleukin 2 transgene abrogated the tumor metastases and prolonged survival of the animals. These results demonstrate that i.v.-administered GMEC can selectively accumulate, survive, and stably express exogenous genes at multiple tumor sites. These findings support a role for i.v.-administered GMEC as a potential therapeutic strategy for the systemic treatment of cancer metastases.


Subject(s)
Endothelium, Vascular/physiology , Genetic Therapy/methods , Interleukin-2/genetics , Lung Neoplasms/therapy , Melanoma, Experimental/therapy , Animals , Female , Gene Targeting/methods , Genetic Vectors/administration & dosage , Green Fluorescent Proteins , Humans , Immunoenzyme Techniques , Lac Operon , Luminescent Proteins , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Melanoma, Experimental/secondary , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Retroviridae/genetics , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
7.
Semin Oncol ; 28(4): 336-43, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11498828

ABSTRACT

By far, cancer accounts for the majority of gene therapy trials that are being carried out worldwide. Seventy percent of the gene therapy protocols that have been reviewed by the National Institutes of Health Recombinant DNA Advisory Committee (NIH RAC) are for the treatment of cancer. Of these, two thirds involve immunotherapy, with transfer of genes for cytokines, immune accessory molecules, or tumor antigens into a variety of cellular targets. Other clinical protocols include chemoprotection, prodrug activation, or tumor-suppressor gene replacement. Either local or distal bystander effects may mediate antitumor effects. These bystander mechanisms may help to overcome poor transduction efficiencies by currently available vectors. Replicating oncolytic viruses entering the clinic include herpes virus, Newcastle disease virus, reovirus, and others. These viruses have been shown to replicate selectively in cancer cells, albeit by different mechanisms. Reovirus, for example, requires the presence of an activated Ras signaling pathway in order to replicate and destroy cells. Tumor selectivity can be achieved by placing an essential viral gene under the control of a tumor-specific promoter. The tumoricidal effects of replicating viruses may be enhanced by genetic modification-for example, by the insertion of a cytokine gene to elicit antitumor immunity. Clearly, much work needs to be done both in the laboratory and in the clinic in order to exploit the full potential of these novel gene and viral therapies.


Subject(s)
Breast Neoplasms/therapy , Genetic Therapy , Neoplasms/therapy , Animals , Clinical Trials as Topic , Genetic Therapy/methods , Genetic Vectors , Humans
8.
Nat Med ; 7(7): 781-7, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11433341

ABSTRACT

In the search for novel cancer therapies that can be used in conjunction with existing treatments, one promising area of research is the use of viral vectors and whole viruses. This review describes the underlying biological principles and current status of the field, outlines approaches for improving clinical effectiveness and discusses the unique safety and regulatory issues surrounding viral therapies.


Subject(s)
Biological Therapy , Neoplasms/therapy , Virus Replication , Viruses , Humans , Promoter Regions, Genetic , Risk Management , Virus Replication/genetics
10.
Endocrinology ; 142(4): 1497-505, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11250930

ABSTRACT

Many human breast tumors are driven by high intratumor concentrations of 17beta-estradiol that appear to be locally synthesized. The role of aromatase is well established, but the possible contribution of the steroid sulfatase (STS), which liberates estrogens from their biologically inactive sulfates, has been inadequately assessed and remains unclear. To evaluate the role of STS further, we transduced estrogen-dependent MCF-7 human breast cancer cells with a retroviral vector directing the constitutive expression of the human STS gene. Gene integration was confirmed by Southern hybridization, production of the appropriately sized messenger RNA by Northern hybridization, and expression of functional protein by metabolism of [(3)H]estrone sulfate to [(3)H]estrone. Maximum velocity estimates of estrone formation are 64.2 pmol estrone/mg protein.h in STS-transduced cells (STS Clone 20), levels comparable to those seen in some human breast tumors. Lower levels of endogenous activity are seen in MCF-7 cells (13.0 pmol estrone/mg protein.h) and in cells transduced with vector lacking the STS gene (Vector 3 cells; 12.0 pmol estrone/mg protein.h). 17beta-Estradiol sulfate induces expression of the progesterone receptor messenger RNA only in STS Clone 20 cells, whereas estrone sulfate produces the greatest stimulation of anchorage-independent growth in these cells. STS Clone 20 cells retain responsiveness to antiestrogens, which block the ability of estrogen sulfate to increase the proportion of cells in both the S and G(2)/M phases of the cell cycle. Consistent with these in vitro observations, only STS Clone 20 cells exhibit a significant increase in the proportion of proliferating tumors in nude ovariectomized mice supplemented with 17beta-estradiol sulfate. The primary activity in vivo appears to be from intratumor STS, rather than hepatic STS. Surprisingly, 17beta-estradiol sulfate appears more effective than 17beta-estradiol when both are administered at comparable concentrations. This effect, which is seen only in STS Clone 20 cells, may reflect differences in the cellular pharmacology of exogenous estrogens compared with those released by the activity of intracellular STS. These studies directly demonstrate that intratumor STS activity can support estrogen-dependent tumorigenicity in an experimental model and may contribute to the promotion of human breast tumors.


Subject(s)
Arylsulfatases/biosynthesis , Arylsulfatases/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Animals , Blotting, Northern , Blotting, Southern , Breast Neoplasms/pathology , Cell Cycle/physiology , Estradiol/metabolism , Female , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Nuclease Protection Assays , Signal Transduction/drug effects , Steryl-Sulfatase , Subcellular Fractions/metabolism , Tumor Cells, Cultured
12.
Semin Oncol ; 27(5): 560-77, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11049023

ABSTRACT

Few chemotherapy agents have demonstrated activity in patients with myelodysplastic syndromes (MDS) and supportive management remains the standard of care. An increasing number of new drugs in development are being directed at specific molecular or biological targets of these diseases. Topotecan, a topoisomerase I inhibitor, has shown single-agent activity and is now being combined with other agents, including cytarabine. The aminothiol amifostine induces responses in about 30% of patients; however, its role is still being clarified. Agents that inhibit histone deacetylase and target DNA hypermethylation, thus permitting derepression of normal genes, include 5-azacytidine, decitabine, phenylbutyrate, and depsipeptide. Arsenic trioxide has demonstrated impressive activity in acute promyelocytic leukemia and preclinical data suggest the potential for activity in MDS. UCN-01 is a novel agent that inhibits protein kinase C and other protein kinases important for progression through the G1 and G2 phases of the cell cycle. Dolastatin-10 has extremely potent in vitro activity against a variety of tumor cell lines. Since its dose-limiting toxicities include myelosuppression, it is being studied in acute myelogenous leukemia (AML) and MDS. Ras may play a role in MDS, and activation of this gene and its signaling pathways may require farnesylation. Several farnesyl transferase inhibitors are now available for study in patients with MDS. An increasing body of data suggests a possible role for angiogenesis in MDS, and several antiangiogenesis agents are in clinical trials, including thalidomide, SU5416, and anti-vascular endothelial growth factor (VEGF) antibodies. Development of new drugs and regimens will be facilitated by recently developed standardized response criteria. Future clinical trials should focus on rational combinations of these agents and others with the goal of curing patients with MDS.


Subject(s)
Antineoplastic Agents/therapeutic use , Myelodysplastic Syndromes/drug therapy , Antineoplastic Agents/pharmacology , Humans
13.
Leukemia ; 14(3): 488-90, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10720147

ABSTRACT

New agents for the treatment of acute myelogenous leukemia are discussed that reflect different treatment mechanisms. These include histone acetylation, angiogenesis inhibition, protein kinase inhibitors, and a novel retinoid. Efficacy and safety in phase I and phase II trials reviewed, as well as the problems involved in crossing over from treatment of solid tumors to blood disorders.


Subject(s)
Antineoplastic Agents/therapeutic use , Leukemia, Myeloid/drug therapy , Saccharomyces cerevisiae Proteins , Acetylation/drug effects , Acetyltransferases/metabolism , Acute Disease , Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/pharmacology , Clinical Trials as Topic , Enzyme Inhibitors/therapeutic use , Forecasting , Gene Expression Regulation, Leukemic/drug effects , Histone Acetyltransferases , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Processing, Post-Translational/drug effects , Retinoids/pharmacology , Retinoids/therapeutic use
14.
Semin Oncol ; 25(1): 42-59, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9482526

ABSTRACT

While both the Rai and Binet staging systems continue to provide the most useful tools for assessing prognosis in chronic lymphocytic leukemia (CLL), both fail to identify subsets of patients that may, or may not, benefit from therapy. While numerous factors have been identified that correlate with stage, few have proven to be independent predictors of disease progression, survival, or resistance to chemotherapy. Several factors, including beta2-microglobulin, soluble CD23, and lymphocyte doubling time show particular promise as independent prognostic factors. Their usefulness, however, will remain uncertain until they have been adequately evaluated in prospective, randomized studies. Future studies should focus on the characterization of relevant biologic and immunologic markers in order to direct appropriate treatment for different subsets of patients and to lead to improved outcome in CLL.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Bone Marrow/pathology , Chromosome Aberrations , Drug Resistance, Multiple/genetics , Drug Resistance, Neoplasm , Humans , Immunophenotyping , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Neoplasm Staging , Prognosis
16.
Clin Exp Metastasis ; 15(2): 173-83, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9062394

ABSTRACT

Bone is a common metastatic site in human breast cancer (HBC). Since bone metastasis occurs very rarely from current spontaneous or experimental metastasis models of HBC cells in nude mice, an arterial seeding model involving the direct injection of the cells into the left ventricle has been developed to better understand the mechanisms involved in this process. We present here a sensitive polymerase chain reaction (PCR) method to detect and quantitate bone and soft organ metastasis in nude mice which have been intracardially inoculated with Lac Z transduced HBC cells. Amplification of genomically incorporated Lac Z sequences in MDA-MB-231-BAG HBC cells enables us to specifically detect these cells in mouse organs and bones. We have also created a competitive template to use as an internal standard in the PCR reactions, allowing us to better quantitate levels of HBC metastasis. The results of this PCR detection method correlate well with cell culture detection from alternate long bones from the same mice, and are more sensitive than gross Lac Z staining with X-gal or routine histology. Comparable qualitative results were obtained with PCR and culture in a titration experiment in which mice were inoculated with increasing numbers of cells, but PCR is more quantifiable, less time consuming, and less expensive. This assay can be employed to study the molecular and cellular aspects of bone metastasis, and could easily be used in conjunction with RT-PCR-based analyses of gene products which may be involved with HBC metastasis.


Subject(s)
Bone Neoplasms/secondary , Breast Neoplasms/pathology , Polymerase Chain Reaction/methods , Soft Tissue Neoplasms/secondary , Animals , Breast Neoplasms/genetics , Cell Count , Female , Genes, Reporter , Humans , Lac Operon , Mice , Mice, Nude , Sensitivity and Specificity , Tumor Cells, Cultured
17.
Zhonghua Bing Li Xue Za Zhi ; 25(4): 228-31, 1996 Aug.
Article in Chinese | MEDLINE | ID: mdl-9275658

ABSTRACT

Since interleukin 2 (IL-2) can stimulat host immune response, the cDNA encoding human IL-2 (hIL-2), mediated by retroviral vector, was transferred into human breast carcinoma cell line "MDA-MB-435". The characterization and tumorigenesis of hIL-2 gene transduced cells were studied. The results showed that provirus genome had been inserted to the genome of transduced cells demonstrated by Southern hybridization. One million transduced cells secreted up to 15ng hIL-2 in 24 hours with ELISA method specific to hIL-2. The mRNA expression of hIL-2 was identified by RT-PCR. The hIL-2 gene modified cells lost their tumorigenesis and the tumorigenesis by mixed injection with their parent tumor cells was inhibited in nude mouse. These data suggest that hIL-2 gene transduced tumor cells may represent a potential vaccine in the management of cancer patients.


Subject(s)
Breast Neoplasms/genetics , Interleukin-2/genetics , Transduction, Genetic , Animals , Breast Neoplasms/pathology , Female , Humans , Immunotherapy, Active , Interleukin-2/biosynthesis , Mice , Mice, Nude , Neoplasm Transplantation , RNA, Messenger/biosynthesis , Retroviridae/genetics , Tumor Cells, Cultured/metabolism
18.
Cytokines Mol Ther ; 2(2): 89-101, 1996 Jun.
Article in English | MEDLINE | ID: mdl-9384693

ABSTRACT

Recent studies have demonstrated the feasibility of cytokine gene transfer to enhance the antitumor activities of host immune cells. Endothelial cells forming the vascular supply of tumors may be useful vehicles for the delivery of cytokine molecules in order to effect tumor immunotherapy. In order to determine whether primary endothelial cells can express cytokine transgenes efficiently, we constructed two retroviral vectors containing a cDNA encoding either recombinant human interleukin-1 alpha (rhIL-1 alpha) or recombinant human interleukin-2 (rhIL-2), called LNCIL-1 alpha and LNCIL-2 respectively, and studied the expression of the two cytokines in vitro in non-immortalized endothelial cells. Human umbilical vein endothelial cells (HUVEC) transduced with LNCIL-1 alpha or LNCIL-2 secreted 1.8-33 ng/10(6) cells/24 h and 40-246.7 ng/10(6) cells/24 h of biological active rhIL-1 alpha and rhIL-2 respectively. Mouse microvascular endothelial cells (MMEC) transduced with LNCIL-1 alpha and LNCIL-2 secreted 1.5 ng/10(6) cells/24 h and 5.8-24.7 ng/10(6) of biologically active rhIL-1 alpha and rhIL-2 proteins respectively. Cocultivation of HUVEC/IL-2 and MMEC/IL-2 with normal human bone marrow cells generated potent cytotoxic activity against K562, Daudi and other cell targets in a 51Cr-release assay. While IL-2 transgene-expressing HUVEC and MMEC retained their normal morphology, rhIL-1 alpha transgene expression inhibited the growth and altered the morphology of both HUVEC and MMEC in culture. The cytokine-gene-transduced endothelial cells retained other endothelial cell features, including uptake of acetylated low-density lipoprotein (Ac-LDL) and expression of von Willebrand factor, and were euploid as shown by flow cytometry. These results demonstrate that endothelial cells, by sustaining the production of biologically active rhIL-2 at levels that are sufficient for the activation of potent cytotoxic lymphocyte activity, may be useful agents for cancer gene therapy.


Subject(s)
Cytokines/biosynthesis , Endothelium, Vascular/cytology , Immunotherapy/methods , Neoplasms/therapy , Transcription, Genetic , Transfection/methods , Animals , Breast Neoplasms , Burkitt Lymphoma , Cell Division/drug effects , Cells, Cultured , Cytotoxicity, Immunologic , Endothelium, Vascular/drug effects , Endothelium, Vascular/immunology , Female , Genetic Vectors , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Humans , Interleukin-1/biosynthesis , Interleukin-2/biosynthesis , Interleukin-6/biosynthesis , Lipopolysaccharides/pharmacology , Lymphocytes/cytology , Lymphocytes/immunology , Mice , Mice, Inbred BALB C , Microcirculation , Polymerase Chain Reaction , RNA, Messenger/biosynthesis , Recombinant Proteins/biosynthesis , Retroviridae , Skin/blood supply , Tumor Cells, Cultured , Umbilical Veins
19.
Cancer Res ; 55(11): 2240-4, 1995 Jun 01.
Article in English | MEDLINE | ID: mdl-7538897

ABSTRACT

By virtue of their location within blood vessels and their ability to express foreign genes, endothelial cells are attractive vehicles for the delivery of therapeutic molecules in vivo. We wished to determine whether i.v.-injected, genetically modified endothelial cells can become incorporated into sites of active angiogenesis in vivo. To do so, we studied the fate of i.v.-injected, lacZ-expressing human umbilical vein endothelial cells in athymic nude mice bearing lethally irradiated NIH 3T3 murine fibroblast cells transfected with a sp-hst/KS3:fibroblast growth factor-1 chimera that forces the secretion of the angiogenic protein, fibroblast growth factor-1. Following i.v. injection, lacZ-labeled human umbilical vein endothelial cells accumulated at sites of fibroblast growth factor-1-induced angiogenesis, persisting for at least 4 weeks. These results suggest that i.v.-administered, genetically modified endothelial cells can migrate into and survive within an angiogenic site. This strategy may be useful for delivery of therapeutic molecules to sites of pathological angiogenesis during tumor metastasis.


Subject(s)
Cell Transplantation/physiology , Endothelium, Vascular/cytology , Neovascularization, Pathologic/pathology , 3T3 Cells/metabolism , 3T3 Cells/physiology , Animals , Chimera , Female , Fibroblast Growth Factor 1/metabolism , Fibroblast Growth Factor 4 , Fibroblast Growth Factors/genetics , Humans , Injections, Subcutaneous , Lac Operon , Mice , Mice, Nude , Proto-Oncogene Proteins/genetics , Transduction, Genetic , Umbilical Veins/physiology
20.
J Immunol ; 152(7): 3294-305, 1994 Apr 01.
Article in English | MEDLINE | ID: mdl-8144916

ABSTRACT

Recent studies of transgenic mice have confirmed that clonal deletion is involved in the development of B cells. However, little is known about intercellular and intracellular molecular events regulating B cell clonal deletion. We investigated the role of bcl-2 and cytokines in the regulation of B cell clonal deletion using anti-IgM-induced growth arrest and apoptosis in immature B cell lines as a model. We show here that overexpression of Bcl-2 protein in stably transfected immature B cells partially inhibits anti-Ig M-induced apoptosis but does not affect growth arrest. Similarly, IL-5 has a strong inhibitory effect on anti-IgM-mediated apoptosis but has a weak inhibitory effect on growth arrest. Finally, although both bcl-2 overexpression and exogenous IL-5 cooperate with bacterial LPS to block apoptosis, bcl-2 overexpression and exogenous IL-5 have no additive inhibitory effect on anti-Ig induced apoptosis. These findings indicate that anti-IgM-induced apoptosis is independently regulated from growth arrest and is controlled by at least two independent pathways: One is regulated by either Bcl-2 protein or IL-5 and the other is regulated by LPS. Activation of both the bcl-2/IL-5 and LPS pathways is necessary for complete inhibition of apoptosis, and presumably, clonal selection of the immature B cells.


Subject(s)
Apoptosis , B-Lymphocytes/cytology , Immunoglobulin M/physiology , Interleukin-5/physiology , Proto-Oncogene Proteins/physiology , src-Family Kinases , Animals , Cell Cycle , Cell Line , Clonal Deletion , Drug Synergism , Gene Expression , Lipopolysaccharides/administration & dosage , Mice , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-bcl-2 , RNA, Messenger/genetics , Receptors, Antigen, B-Cell/physiology
SELECTION OF CITATIONS
SEARCH DETAIL