Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Cancer ; 110(5): 1042-9, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17654660

ABSTRACT

BACKGROUND: Bortezomib is active in heavily pretreated multiple myeloma patients; the dose-limiting toxicity is peripheral neuropathy (PN). METHODS: The authors retrospectively reviewed the incidence, severity, and risk factors for PN in 78 patients who received bortezomib. The median age was 57 years (range, 33-80 years), 62% of patients were men, and 37% of patients were African Americans. Seventeen patients (22%) had diabetes mellitus (DM), and 66 patients (85%) had received thalidomide. Before bortezomib treatment, 37% of the patients reported subjective, grade 1 or 2 PN. Patients received bortezomib alone (n = 10 patients) plus dexamethasone (n = 36 patients) and thalidomide (n = 20 patients) or chemotherapy (n = 12 patients). PN affected 52% of patients, including grade 3 and 4 PN in 15% and 7%, respectively. RESULTS: Twelve patients stopped bortezomib because of side effects that included PN (n = 9 patients), diarrhea (n = 2 patients) and cytomegalovirus pneumonia (n = 1 patient); 11 patients had dose reductions because of PN. Grade 4 PN affected 6 patients (sensory, n = 4 patients; motor/sensory, n = 2 patients). The onset of grade 4 PN was sudden rather than cumulative. Factors that were predictive of PN grade were baseline PN (P = .002), prior thalidomide use (P = .03), and the presence of DM (P = .03). Multiple myeloma responses included complete, near complete, and partial responses in 5% of patients, 10% of patients, and 27% of patients, respectively. Responses were independent of PN and of whether bortezomib was combined with chemotherapy or thalidomide. Patients remained on therapy longer for a median of 5 cycles (range, 2-36 cycles) when they received bortezomib plus thalidomide versus 3 cycles (range, 1-19 cycles) for the other combinations. PN therapy was mostly supportive. It was noteworthy that 6 of 9 patients with PN who received lenalidomide as salvage therapy after bortezomib had significant improvement in their symptoms. CONCLUSIONS: The risk of bortezomib-related PN was greater in patients who had PN and DM at baseline. The authors concluded that an unexpected, symptomatic improvement of PN on lenalidomide is worth further investigation.


Subject(s)
Boronic Acids/adverse effects , Boronic Acids/therapeutic use , Multiple Myeloma/drug therapy , Peripheral Nervous System Diseases/chemically induced , Pyrazines/adverse effects , Pyrazines/therapeutic use , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols , Boronic Acids/administration & dosage , Bortezomib , Dexamethasone/administration & dosage , Drug Administration Schedule , Female , Humans , Male , Middle Aged , Multiple Myeloma/pathology , Neoplasm Recurrence, Local , Pyrazines/administration & dosage , Retrospective Studies , Thalidomide/administration & dosage , Treatment Outcome
2.
Haematologica ; 91(3): 390-3, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16503551

ABSTRACT

Flavopiridol downregulates anti-apoptotic regulators including Mcl-1, upregulates p53, globally attenuates transcription through inhibition of P-TEFb, binds to DNA, and inhibits angiogenesis. Eighteen myeloma patients were treated with 1-hour flavopiridol infusions for 3 consecutive days every 21 days. Immunoblotting for Mcl-1, Bcl-2, p53, cyclin D, phosphoRNA polymerase II and phosphoSTAT 3 was conducted on myeloma cells. Ex vivo flavopiridol treatment of cells resulted in cytotoxicity, but only after longer exposure times at higher flavopiridol concentrations than were anticipated to be achieved in vivo. No anti-myeloma activity was observed in vivo. As administered, flavopiridol has disappointing activity as a single agent in advanced myeloma.


Subject(s)
Flavonoids/pharmacology , Flavonoids/therapeutic use , Multiple Myeloma/drug therapy , Multiple Myeloma/prevention & control , Neoplasm Recurrence, Local/prevention & control , Piperidines/pharmacology , Piperidines/therapeutic use , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Treatment Outcome
3.
J Clin Oncol ; 23(18): 4089-99, 2005 Jun 20.
Article in English | MEDLINE | ID: mdl-15867202

ABSTRACT

PURPOSE: Bcl-2 regulates the mitochondrial apoptosis pathway that promotes chemotherapy resistance. Bcl-2 antisense oligonucleotide, G3139, targets Bcl-2 mRNA. PATIENTS AND METHODS: G3139 was administered (3 to 7 mg/kg/d for 7 days) by continuous intravenous infusion. On day 4, patients started thalidomide (100 to 400 mg as tolerated) and dexamethasone (40 mg daily for 4 days) on 21-day cycles for three cycles. Stable and responding patients continued on 35-day cycles for 2 years. RESULTS: Thirty-three patients (median age, 60 years; range, 28 to 76 years) received 220 cycles. Patients received a median of three prior regimens including thalidomide (n = 15) and stem-cell transplantation (n = 31). The regimen was well tolerated; the median number of cycles per patient was eight (range, one to 16+ cycles). Toxicities included reversible increase in creatinine, thrombocytopenia, neutropenia, fatigue, anorexia, constipation, fever, neuropathy, edema, electrolyte disturbances, and hyperglycemia. Fifty-five percent of patients had objective responses, including two complete responses (CRs), four near CRs (positive immunofixation), and 12 partial responses; six patients had minimal responses (MRs). Of patients who received prior thalidomide, seven had objective responses, and three had MRs. The median duration of response was 13 months, and estimated progression-free and overall survival times were 12 and 17.4 months, respectively. Responding patients had significant increase in polyclonal immunoglobulin M (P = .005), indicating innate immune system activation. Western blot analysis of Bcl-2 protein isolated from myeloma cells before and after G3139 demonstrated a decrease of Bcl-2 levels in three of seven patients compared with six of nine patients using reverse transcriptase polymerase chain reaction. CONCLUSION: G3139, dexamethasone, and thalidomide are well tolerated and result in encouraging clinical responses in relapsed multiple myeloma patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Multiple Myeloma/drug therapy , Adult , Aged , Blotting, Western , Dexamethasone/administration & dosage , Female , Humans , Infusions, Intravenous , Male , Middle Aged , Oligonucleotides, Antisense/administration & dosage , Recurrence , Reverse Transcriptase Polymerase Chain Reaction , Stem Cell Transplantation , Survival Analysis , Thalidomide/administration & dosage , Thionucleotides/administration & dosage , Treatment Outcome
4.
Clin Cancer Res ; 10(24): 8301-8, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15623606

ABSTRACT

PURPOSE: Inosine monophosphate dehydrogenase (IMPDH) inhibitors have been used to induce leukemia blast cell differentiation but have not been tested in multiple myeloma for activity. Currently, available IMPDH inhibitor, mycophenolate mofetil (MMF), which is known as an immunosuppressant, was shown to induce apoptosis in myeloma cell lines. On the basis of our preclinical studies, we designed a clinical study to test our hypothesis that MMF has antimyeloma activity. EXPERIMENTAL DESIGN: A Phase I MMF dose escalation study was conducted in relapsed and refractory myeloma patients who had documented disease progression by myeloma markers or bone marrow plasmacytosis to determine the maximum tolerated dose, toxicities, and efficacy of the drug. To assess the activity of IMPDH inhibition in the myeloma cells of patients, we measured intracellular nucleotide triphosphate levels by high-performance liquid chromatography-based analysis and examined the correlation with clinical response. RESULTS: Among the 11 study patients, MMF was generally well tolerated and was administered up to a maximum dose of 5 g/day. The most common toxicity was grade 1 fatigue (n = 4, 36%). One patient had a partial response (3 g/day), four patients had stable disease, and six patients had progression of disease. There was a statistically significant difference in the intracellular dGTP level changes between the stable disease/partial response group versus progression of disease. CONCLUSIONS: MMF at 1 to 5 g/day daily dose is well tolerated by patients with relapsed and refractory multiple myeloma patients. Positive correlation between clinical response and depletion of intracellular dGTP level was shown. Future drug development to target this enzyme maybe useful in treating myelomas.


Subject(s)
Enzyme Inhibitors/therapeutic use , IMP Dehydrogenase/antagonists & inhibitors , Immunosuppressive Agents/therapeutic use , Multiple Myeloma/drug therapy , Mycophenolic Acid/analogs & derivatives , Aged , Bone Marrow/drug effects , Bone Marrow/metabolism , Bone Marrow/pathology , Chromatography, High Pressure Liquid , Disease Progression , Dose-Response Relationship, Drug , Female , Guanosine Triphosphate/metabolism , Humans , IMP Dehydrogenase/metabolism , Male , Maximum Tolerated Dose , Middle Aged , Multiple Myeloma/enzymology , Multiple Myeloma/pathology , Mycophenolic Acid/therapeutic use , Plasmacytoma/pathology , Salvage Therapy
5.
Free Radic Biol Med ; 37(11): 1845-53, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15528043

ABSTRACT

Bcl-2 family proteins protect against a variety of forms of cell death, including acute oxidative stress. Previous studies have shown that overexpression of the antiapoptotic protein Bcl-2 increases cellular redox capacity. Here we report that cell lines transfected with Bcl-2 paradoxically exhibit increased rates of mitochondrial H(2)O(2) generation. Using isolated mitochondria, we determined that increased H(2)O(2) release results from the oxidation of reduced nicotinamide adenine dinucleotide-linked substrates. Antiapoptotic Bcl-2 family proteins Bcl-xL and Mcl-1 also increase mitochondrial H(2)O(2) release when overexpressed. Chronic exposure of cells to low levels of the mitochondrial uncoupler carbonyl cyanide 4-(triflouromethoxy)phenylhydrazone reduced the rate of H(2)O(2) production by Bcl-xL overexpressing cells, resulting in a decreased ability to remove exogenous H(2)O(2) and enhanced cell death under conditions of acute oxidative stress. Our results indicate that chronic and mild elevations in H(2)O(2) release from Bcl-2, Bcl-xL, and Mcl-1 overexpressing mitochondria lead to enhanced cellular antioxidant defense and protection against death caused by acute oxidative stress.


Subject(s)
Antioxidants/physiology , Apoptosis , Carbonyl Cyanide m-Chlorophenyl Hydrazone/analogs & derivatives , Mitochondria/metabolism , Oxidative Stress/physiology , Proto-Oncogene Proteins c-bcl-2/physiology , Animals , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Cells, Cultured , Humans , Hydrogen Peroxide/metabolism , Mitochondria/drug effects , Myeloid Cell Leukemia Sequence 1 Protein , NAD/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Oxidation-Reduction , Oxidative Stress/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Rats , bcl-X Protein
6.
Article in English | MEDLINE | ID: mdl-14633785

ABSTRACT

Even during this past year, further advances have been made in understanding the molecular genetics of the disease, the mechanisms involved in the generation of myeloma-associated bone disease and elucidation of critical signaling pathways as therapeutic targets. New agents (thalidomide, Revimid, Velcade) providing effective salvage therapy for end-stage myeloma, have broadened the therapeutic armamentarium markedly. As evidenced in Section I by Drs. Kuehl and Bergsagel, five recurrent primary translocations resulting from errors in IgH switch recombination during B-cell development in germinal centers involve 11q13 (cyclin D1), 4p16.3 (FGFR3 and MMSET), 6p21 (cyclin D3), 16q23 (c-maf), and 20q11 (mafB), which account for about 40% of all myeloma tumors. Based on gene expression profiling data from two laboratories, the authors propose 5 multiple myeloma (MM) subtypes defined by the expression of translocation oncogenes and cyclins (TC molecular classification of MM) with different prognostic implications. In Section II, Drs. Barillé-Nion and Bataille review new insights into osteoclast activation through the RANK Ligand/OPG and MIP-1 chemokine axes and osteoblast inactivation in the context of recent data on DKK1. The observation that myeloma cells enhance the formation of osteoclasts whose activity or products, in turn, are essential for the survival and growth of myeloma cells forms the basis for a new treatment paradigm aimed at reducing the RANKL/OPG ratio by treatment with RANKL inhibitors and/or MIP inhibitors. In Section III, Dr. Fenton reviews apoptotic pathways as they relate to MM therapy. Defects in the mitochrondrial intrinsic pathway result from imbalances in expression levels of Bcl-2, Bcl-XL and Mcl-1. Mcl-1 is a candidate target gene for rapid induction of apoptosis by flavoperidol. Antisense oglionucleotides (ASO) lead to the rapid induction of caspace activity and apoptosis, which was potentiated by dexamethasone. Similar clinical trials with Bcl-2 ASO molecules alone and in combination with doxorubicin and dexamethasone or thalidomide showed promising results. The extrinsic pathway can be activated upon binding of the ligand TRAIL. OPG, released by osteoblasts and other stromal cells, can act as a decoy receptor for TRAIL, thereby blocking its apoptosis-inducing activity. MM cells inhibit OPG release by stromal cells, thereby promoting osteoclast activation and lytic bone disease (by enhancing RANKL availability) while at the same time exposing themselves to higher levels of ambient TRAIL. Thus, as a recurring theme, the relative levels of pro- versus anti-apoptotic molecules that act in a cell autonomous manner or in the milieu of the bone marrow microenvironment determine the outcome of potentially lethal signals. In Section IV, Dr. Barlogie and colleagues review data on single and tandem autotransplants for newly diagnosed myeloma. CR rates of 60%-70% can be reached with tandem transplants extending median survival to approximately 7 years. Dose adjustments of melphalan in the setting of renal failure and age > 70 may be required to reduce mucositis and other toxicities in such patients, especially in the context of amyloidosis with cardiac involvement. In Total Therapy II the Arkansas group is evaluating the role of added thalidomide in a randomized trial design. While data are still blinded as to the contribution of thalidomide, the overriding adverse importance of cytogenetic abnormalities, previously reported for Total Therapy I, also pertain to this successor trial. In these two-thirds of patients without cytogenetic abnormalities, Total Therapy II effected a doubling of the 4-year EFS estimate from 37% to 75% (P <.0001) and increased the 4-year OS estimate from 63% to 84% (P =.0009). The well-documented graft-vs-MM effect of allotransplants can be more safely examined in the context of non-myeloablative regimens, applied as consolidation after a single autologous transplant with melphalan 200 mg/m(2), have been found to be much better tolerated than standard myeloablative conditioning rege conditioning regimens and yielding promising results even in the high-risk entity of MM with cytogenetic abnormalities. For previously treated patients, the thalidomide congener Revimid and the proteasome inhibitor Velcade both are active in advanced and refractory MM (approximately 30% PR). Gene expression profiling (GEP) has unraveled distinct MM subtypes with different response and survival expectations, can distinguish the presence of or future development of bone disease, and, through serial investigations, can elucidate mechanisms of actions of new agents also in the context of the bone marrow microenvironment. By providing prognostically relevant distinction of MM subgroups, GEP should aid in the development of individualized treatment for MM.


Subject(s)
Multiple Myeloma/therapy , Bone Diseases/diagnosis , Bone Diseases/drug therapy , Bone Diseases/etiology , Drug Delivery Systems , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Humans , Multiple Myeloma/etiology , Multiple Myeloma/genetics , Signal Transduction/drug effects , Treatment Outcome
7.
Oncogene ; 22(12): 1848-59, 2003 Mar 27.
Article in English | MEDLINE | ID: mdl-12660820

ABSTRACT

Mcl-1 is a critical antiapoptotic survival factor for human multiple myeloma (MM). We examined the importance of IL-6 for Mcl-1 expression in five MM cell lines and in primary MM cells from 14 patients. While culture of MM.1S cells in IL-6 did induce Mcl-1 expression, four other MM cell lines exhibited high levels of Mcl-1 expression that were unaffected by IL-6. Similarly, Mcl-1 expression in 10 of 14 primary MM isolates was found to be IL-6-independent. An analysis of the mechanisms responsible for IL-6-independent Mcl-1 expression was undertaken. ERK1/2 activity did not influence Mcl-1 expression, distinct from Mcl-1 regulation that occurs during myeloid differentiation from hematopoietic progenitor cells. Inhibition of the PI3K pathway led to growth inhibition of 8226 and ANBL-6 cells without reduction of Mcl-1 levels, and high level Mcl-1 expression was maintained in the absence of activated STAT3. Analysis of the transcriptional activity of 5'-regulatory sequences from the human Mcl-1 gene in MM cells demonstrated high levels of IL-6-independent indicator gene activation as predicted. These data demonstrate that the mechanisms regulating Mcl-1 levels in MM cells are heterogeneous, and are often independent from IL-6 signaling pathways.


Subject(s)
Gene Expression Regulation, Neoplastic/physiology , Interleukin-6/physiology , Multiple Myeloma/genetics , Neoplasm Proteins/genetics , Proto-Oncogene Proteins c-bcl-2 , Base Sequence , Blotting, Western , Culture Media, Serum-Free , DNA Primers , Humans , Multiple Myeloma/enzymology , Multiple Myeloma/pathology , Myeloid Cell Leukemia Sequence 1 Protein , Phosphatidylinositol 3-Kinases , Regulatory Sequences, Nucleic Acid , Transcriptional Activation , Tumor Cells, Cultured
8.
Clin Cancer Res ; 8(11): 3527-38, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12429644

ABSTRACT

Multiple myeloma (MM) is a B-cell malignancy characterized by the accumulation of malignant plasma cells with slow proliferative rate but enhanced survival. MM cells express multiple Bcl-2 family members, including Bcl-2, Bcl-XL, and Mcl-1, which are thought to play a key role in the survival and drug resistance of myeloma. The cyclin-dependent kinase inhibitor flavopiridol has antitumor activity against hematopoietic malignancies, including CLL, in which induction of apoptosis was associated with reduced expression of antiapoptotic proteins. Therefore, we sought to characterize the effect of flavopiridol on the proliferation and survival of myeloma cells and to define its mechanisms of action. Treatment of MM cell lines (8226, ANBL-6, ARP1, and OPM-2) with clinically achievable concentrations of flavopiridol resulted in rapid induction of apoptotic cell death that correlated temporally with the decline in Mcl-1 protein and mRNA levels. Levels of other antiapoptotic proteins did not change. Overexpression of Mcl-1 protected MM cells from flavopiridol-induced apoptosis. Additional analysis demonstrated that flavopiridol treatment resulted in a dose-dependent inhibition of phosphorylation of the RNA polymerase II COOH-terminal domain, thus blocking transcription elongation. These data indicate that Mcl-1 is an important target for flavopiridol-induced apoptosis of MM that occurs through inhibition of Mcl-1 mRNA transcription coupled with rapid protein degradation via the ubiquitin-proteasome pathway.


Subject(s)
Apoptosis , Cyclin-Dependent Kinases/antagonists & inhibitors , Down-Regulation , Flavonoids/pharmacology , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Neoplasm Proteins/metabolism , Piperidines/pharmacology , Transcription, Genetic , Blotting, Northern , Blotting, Western , Caspases/metabolism , Cell Division , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Humans , In Situ Nick-End Labeling , Membrane Glycoproteins/biosynthesis , Myeloid Cell Leukemia Sequence 1 Protein , Phosphorylation , Protein Structure, Tertiary , Proteoglycans/biosynthesis , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/metabolism , Syndecans , Time Factors , Tumor Cells, Cultured , bcl-X Protein
9.
J Cell Physiol ; 193(1): 42-54, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12209879

ABSTRACT

The evolutionarily conserved Ras/Raf/MEK/ERK pathway is thought to be essential for proliferation of eukaryotic cells. The human multiple myeloma (MM) cell line 8226 encodes an activated K-ras allele and proliferates without requirement for the main MM growth and survival factor IL-6. Surprisingly, the addition of the MEK1/2 inhibitors PD98059 or U0126 to 8226 cultures at doses that block virtually all ERK1/2 activity had minimal effects on the rapid proliferation of this cell line. In contrast, proliferation of the IL-6-dependent MM cell line, ANBL-6 was blocked by PD98059. Levels of activated forms of the other classical MAP kinases (JNK and p38) were very low during MM cell proliferation and, therefore, do not substitute for the mitogenic activities normally regulated by ERK kinases. These data demonstrate that proliferation of 8226 cells does not require ERK1/2 activity, and suggest that IL-6-independent growth of MM may correlate with independence from a requirement for ERK activity. Other signal transduction pathways that appear to regulate cell cycle progression in these cells were examined.


Subject(s)
Interleukin-6/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Multiple Myeloma/drug therapy , Multiple Myeloma/enzymology , Protein Serine-Threonine Kinases , Antibiotics, Antineoplastic/pharmacology , Blotting, Western , Butadienes/pharmacology , Cell Cycle/drug effects , Cell Division/drug effects , Chromones/pharmacology , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Genes, ras , Humans , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Morpholines/pharmacology , Multiple Myeloma/genetics , Nitriles/pharmacology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Ribosomal Protein S6 Kinases/antagonists & inhibitors , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction/drug effects , Sirolimus/pharmacology , Tumor Cells, Cultured , p38 Mitogen-Activated Protein Kinases
10.
Blood ; 99(6): 1885-93, 2002 Mar 15.
Article in English | MEDLINE | ID: mdl-11877256

ABSTRACT

Multiple myeloma (MM) is characterized by the accumulation of malignant plasma cells in the bone marrow caused primarily by failure of normal homeostatic mechanisms to prevent the expansion of postgerminal center plasma cells. We have examined the molecular mechanisms that promote the survival of MM cells and have identified a key role for myeloid cell factor-1 (Mcl-1), an antiapoptotic member of the Bcl-2 family. These experiments were initiated by the observation that MM cells were exquisitely sensitive to culture in the presence of actinomycin D: caspase activation occurred within 3 hours of treatment and cells were not protected by interleukin-6, the main MM cell growth and survival factor. Actinomycin D-induced apoptosis was blocked by proteasome inhibitors, suggesting that a labile protein was required for MM cell survival. Further analysis demonstrated that Mcl-1 was likely to be the labile factor governing MM cell survival. Mcl-1 protein levels decreased rapidly after culture in the presence of actinomycin D in concordance with effector caspase activation, but addition of proteasome inhibitors reversed the loss of Mcl-1 and maintained cell viability. The levels of other antiapoptotic proteins, including Bcl-2 and members of the inhibitors-of-apoptosis family, were unaffected by these interventions. Furthermore, Mcl-1 antisense oligonucleotides caused a rapid down-regulation of Mcl-1 protein levels and the coincident induction of apoptosis, whereas overexpression of Mcl-1 delayed actinomycin D-induced apoptosis with kinetics that correlated with expression levels of Mcl-1. These data indicate that Mcl-1 expression is required for the survival of MM cells and may represent an important target for future therapeutics.


Subject(s)
Multiple Myeloma/metabolism , Neoplasm Proteins/physiology , Apoptosis/drug effects , Bone Marrow/pathology , Caspases/drug effects , Caspases/metabolism , Dactinomycin/pharmacology , Enzyme Activation/drug effects , Humans , Multiple Myeloma/pathology , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Proteins/metabolism , Neoplasm Proteins/pharmacology , Protease Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Cells, Cultured , bcl-X Protein
11.
Cancer Res ; 62(2): 450-8, 2002 Jan 15.
Article in English | MEDLINE | ID: mdl-11809695

ABSTRACT

Factors that govern host-tumor interaction play a critical role in tumor progression. In previous studies we have shown that oncogenic Ras inhibits the expression of Fas (CD95) and renders Ras-transformed cells resistant to Fas-induced death. We now demonstrate that culture of Ras-transformed cells in the presence of the farnesyltransferase inhibitor (FTI) LB42722 leads to up-regulation of Fas expression, both under basal growth conditions and in the presence of the inflammatory cytokines IFN-gamma and tumor necrosis factor alpha. This is manifested by an increase in fas mRNA, Fas cell surface expression, and Fas-induced apoptosis. Whereas FTI up-regulates expression of FAS in Ras-transformed cells, it inhibits the expression of vascular endothelial growth factor. Culture of Ras-transformed cells in the presence of the histone deacetylase inhibitor trichostatin A resulted in morphological reversion and G(1) arrest (as observed with FTI); however, no induction of Fas was observed. Furthermore, the effects of FTI on Fas-induced death were shown to be independent of RhoB. Therefore, inhibition of oncogenic Ras by FTI can result in two events that alter host-tumor interactions: up-regulation of Fas, rendering tumors more sensitive to immune cytotoxic effector cells, and down-reglation of VEGF, which may inhibit tumor angiogenesis.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , ras Proteins/physiology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cell Line, Transformed , Cell Membrane/drug effects , Cell Membrane/metabolism , Farnesyltranstransferase , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/physiology , G1 Phase/drug effects , G1 Phase/physiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Histone Deacetylase Inhibitors , Humans , Mice , Mice, Inbred C3H , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Up-Regulation/drug effects , fas Receptor/biosynthesis , fas Receptor/genetics , ras Proteins/antagonists & inhibitors , rhoB GTP-Binding Protein/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...