Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
PLoS One ; 18(10): e0292231, 2023.
Article in English | MEDLINE | ID: mdl-37788264

ABSTRACT

Biosimilars are increasingly available for the treatment of many serious disorders, however some concerns persist about switching a patient to a biosimilar whose condition is stable while on the reference biologic. Randomized controlled studies and extension studies with a switch treatment period (STP) to or from a biosimilar and its reference biologic were identified from publicly available information maintained by the U.S. Food and Drug Administration (FDA). These findings were augmented with data from peer reviewed publications containing information not captured in FDA reviews. Forty-four STPs were identified from 31 unique studies for 21 different biosimilars. Data were extracted and synthesized following PRISMA guidelines. Meta-analysis was conducted to estimate the overall risk difference across studies. A total of 5,252 patients who were switched to or from a biosimilar and its reference biologic were identified. Safety data including deaths, serious adverse events, and treatment discontinuation showed an overall risk difference (95% CI) of -0.00 (-0.00, 0.00), 0.00 (-0.01, 0.01), -0.00 (-0.01, 0.00) across STPs, respectively. Immunogenicity data showed similar incidence of anti-drug antibodies and neutralizing antibodies in patients within a STP who were switched to or from a biosimilar to its reference biologic and patients who were not switched. Immune related adverse events such as anaphylaxis, hypersensitivity reactions, and injections site reactions were similar in switched and non-switched patients. This first systematic review using statistical methods to address the risk of switching patients between reference biologics and biosimilars finds no difference in the safety profiles or immunogenicity rates in patients who were switched and those who remained on a reference biologic or a biosimilar.


Subject(s)
Anaphylaxis , Biosimilar Pharmaceuticals , Humans , Biosimilar Pharmaceuticals/adverse effects , Biological Factors , Research Design , Anaphylaxis/chemically induced , Antibodies
2.
Clin Cancer Res ; 23(8): 1882-1885, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28034906

ABSTRACT

The Biologics Price Competition and Innovation (BPCI) Act, enacted as part of the Affordable Care Act, created a new licensure pathway for biological products demonstrated to be biosimilar with or interchangeable with an FDA-licensed biological product (the "reference product"). The FDA's approach to the regulation of biosimilars is based on the requirements set forth in the BPCI Act. A biosimilar product is highly similar to the reference product, notwithstanding minor differences in clinically inactive components, and there are no clinically meaningful differences between products in terms of safety, purity, and potency. The foundation of a biosimilar development program is an analytic similarity assessment that directly compares the structural/physiochemical and functional properties of the proposed biosimilar with the reference product. Data from clinical studies, which include an assessment of immunogenicity and pharmacokinetics/pharmacodynamics, are used to assess for clinically meaningful differences and not to independently establish the safety and effectiveness of the biosimilar. Like all products that the FDA regulates, the FDA requires that biosimilar products meet the agency's rigorous standards of safety and efficacy for approval. That means patients and health care professionals are able to rely upon the safety and effectiveness of biosimilar products in the same manner as for the reference product. Clin Cancer Res; 23(8); 1882-5. ©2016 AACR.


Subject(s)
Biosimilar Pharmaceuticals , Drug Approval/legislation & jurisprudence , Humans , United States , United States Food and Drug Administration
3.
Oncologist ; 20(6): 653-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25964306

ABSTRACT

BACKGROUND: First-in-human (FIH) trials of low-molecular-weight anticancer agents conventionally derive a safe start dose (SD) from one-tenth the severely toxic dose in 10% of rodents or one-sixth the highest nonseverely toxic dose (HNSTD) in nonrodent species. No consensus has been reached on whether this paradigm can be safely applied to biotechnology-derived products (BDPs). MATERIALS AND METHODS: A comprehensive search was conducted to identify all BDPs (excluding immune checkpoint inhibitors and antibody drug conjugates) with sufficient nonclinical and clinical data to assess the safety of hypothetical use of one-sixth HNSTD in an advanced cancer FIH trial. RESULTS: The search identified 23 BDPs, of which 21 were monoclonal antibodies. The median ratio of the maximum tolerated or maximum administered dose (MTD or MAD) to the actual FIH SD was 36 (range, 8-500). Only 2 BDPs reached the MTD. Hypothetical use of one-sixth HNSTD (allometrically scaled to humans) would not have exceeded the MTD or MAD for all 23 BDPs and would have reduced the median ratio of the MTD or MAD to a SD to 6.1 (range, 3.5-55.3). Pharmacodynamic (PD) markers were included in some animal toxicology studies and were useful to confirm the hypothetical SD of one-sixth HNSTD. CONCLUSION: One-sixth HNSTD would not have resulted in unacceptable toxicities in the data available. Supporting its use could reduce the number of dose escalations needed to reach the recommended dose. A low incidence of toxicities in animals and humans underscores the need to identify the pharmacokinetic and PD parameters to guide SD selection of BDPs for FIH cancer trials. IMPLICATIONS FOR PRACTICE: Start dose (SD) for biotechnology-derived products (BDPs) can be safely derived from one-sixth the highest nonseverely toxic dose in nonrodent species and may reduce the number of dose escalations needed to reach the recommended dose in first-in-human studies while limiting unnecessary exposure to high drug levels in humans. The use of this type of SD could improve the design of phase I studies of BDPs by making them more efficient. The role of preclinical pharmacodynamic markers was useful in confirming the hypothetical SD, and attempts should be explored in future animal studies to identify such parameters.


Subject(s)
Antineoplastic Agents/administration & dosage , Clinical Trials, Phase I as Topic , Dose-Response Relationship, Drug , Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacokinetics , Humans , Maximum Tolerated Dose , Neoplasms/pathology
4.
Clin Cancer Res ; 21(12): 2666-70, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25802282

ABSTRACT

On July 3, 2014, the FDA granted accelerated approval for belinostat (Beleodaq; Spectrum Pharmaceuticals, Inc.), a histone deacetylase inhibitor, for the treatment of patients with relapsed or refractory peripheral T-cell lymphoma (PTCL). A single-arm, open-label, multicenter, international trial in the indicated patient population was submitted in support of the application. Belinostat was administered intravenously at a dose of 1000 mg/m(2) over 30 minutes once daily on days 1 to 5 of a 21-day cycle. The primary efficacy endpoint was overall response rate (ORR) based on central radiology readings by an independent review committee. The ORR was 25.8% [95% confidence interval (CI), 18.3-34.6] in 120 patients that had confirmed diagnoses of PTCL by the Central Pathology Review Group. The complete and partial response rates were 10.8% (95% CI, 5.9-17.8) and 15.0% (95% CI, 9.1-22.7), respectively. The median duration of response, the key secondary efficacy endpoint, was 8.4 months (95% CI, 4.5-29.4). The most common adverse reactions (>25%) were nausea, fatigue, pyrexia, anemia, and vomiting. Grade 3/4 toxicities (≥5.0%) included anemia, thrombocytopenia, dyspnea, neutropenia, fatigue, and pneumonia. Belinostat is the third drug to receive accelerated approval for the treatment of relapsed or refractory PTCL.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Approval , Histone Deacetylase Inhibitors/therapeutic use , Hydroxamic Acids/therapeutic use , Lymphoma, T-Cell, Peripheral/drug therapy , Lymphoma, T-Cell, Peripheral/pathology , Sulfonamides/therapeutic use , United States Food and Drug Administration , Humans , United States
5.
PLoS One ; 6(7): e21877, 2011.
Article in English | MEDLINE | ID: mdl-21789189

ABSTRACT

p63 is critical for squamous epithelial development, and elevated levels of the ΔNp63α isoform are seen in squamous cell cancers of various organ sites. However, significant controversy exists regarding the role of p63 isoforms as oncoproteins or tumor suppressors. Here, lentiviruses were developed to drive long-term overexpression of ΔNp63α in primary keratinocytes. Elevated levels of ΔNp63α in vitro promote long-term survival and block both replicative and oncogene-induced senescence in primary keratinocytes, as evidenced by the expression of SA-ß-gal and the presence of nuclear foci of heterochromatin protein 1γ. The contribution of ΔNp63α to cancer development was assessed using an in vivo grafting model of experimental skin tumorigenesis that allows distinction between benign and malignant tumors. Grafted lenti-ΔNp63α keratinocytes do not form tumors, whereas lenti-GFP/v-ras(Ha) keratinocytes develop well-differentiated papillomas. Lenti-ΔNp63α/v-ras(Ha) keratinocytes form undifferentiated carcinomas. The average volume of lenti-ΔNp63α/v-ras(Ha) tumors was significantly higher than those in the lenti-GFP/v-ras(Ha) group, consistent with increased BrdU incorporation detected by immunohistochemistry. The block in oncogene-induced senescence corresponds to sustained levels of E2F1 and phosphorylated AKT, and is associated with loss of induction of p16(ink4a)/p19(arf). The relevance of p16(ink4a)/p19(arf) loss was demonstrated in grafting studies of p19(arf)-null keratinocytes, which develop malignant carcinomas in the presence of v-ras(Ha) similar to those arising in wildtype keratinocytes that express lenti-ΔNp63α and v-ras(Ha). Our findings establish that ΔNp63α has oncogenic activity and its overexpression in human squamous cell carcinomas contributes to the malignant phenotype, and implicate its ability to regulate p16(ink4a)/p19(arf) in the process.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p16/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p19/antagonists & inhibitors , Gene Expression Regulation , Keratinocytes/pathology , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Survival , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p19/metabolism , Gene Expression , HEK293 Cells , Humans , Keratinocytes/metabolism , Lentivirus/genetics , Mice , Phenotype , Time Factors , Up-Regulation/genetics , ras Proteins/metabolism
6.
Toxicol Appl Pharmacol ; 243(2): 125-33, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20045015

ABSTRACT

The drug development of new anti-cancer agents is streamlined in response to the urgency of bringing effective drugs to market for patients with limited life expectancy. FDA's regulation of oncology drugs has evolved from the practices set forth in Arnold Lehman's seminal work published in the 1950s through the current drafting of a new International Conference on Harmonization of Technical Requirements for Registration of Pharmaceuticals for Human Use (ICH) safety guidance for anti-cancer drug nonclinical evaluations. The ICH combines the efforts of the regulatory authorities of Europe, Japan, and the United States and the pharmaceutical industry from these three regions to streamline the scientific and technical aspects of drug development. The recent development of new oncology drug classes with novel mechanisms of action has improved survival rates for some cancers but also brings new challenges for safety evaluation. Here we present the legacy of Lehman and colleagues in the context of past and present oncology drug development practices and focus on some of the current issues at the center of an evolving harmonization process that will generate a new safety guidance for oncology drugs, ICH S9. The purpose of this new guidance will be to facilitate oncology drug development on a global scale by standardizing regional safety requirements.


Subject(s)
Antineoplastic Agents/toxicity , Legislation, Drug/trends , Animals , Antineoplastic Agents/adverse effects , Drug Discovery , Drugs, Investigational/adverse effects , Guidelines as Topic , Humans , Neoplasms/drug therapy , Reproduction/drug effects , Safety , United States , United States Food and Drug Administration
7.
Cancer Res ; 68(7): 2062-4, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18381408

ABSTRACT

The proapoptotic cytokine tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is being evaluated presently as a selective anticancer agent, but its limited effects against cancer cell lines has raised some concerns about its ultimate clinical utility. Here, we review recent findings that cancer cell sensitivity to TRAIL is greatly increased when the Bcl-2 family protein Mcl-1 is down-regulated by the Raf/vascular endothelial growth factor kinase inhibitor sorafenib, a Food and Drug Administration-approved cancer drug. Using the TRAIL-sorafenib combination as a tactic to more effectively kill cancer cells may provide an effective tool to attack a variety of human cancers that are largely presently untreatable.


Subject(s)
Neoplasm Proteins/metabolism , Neoplasms/drug therapy , Proto-Oncogene Proteins c-bcl-2/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzenesulfonates/administration & dosage , Benzenesulfonates/pharmacology , Down-Regulation , Drug Resistance, Neoplasm , Drug Synergism , Humans , Leukemia/drug therapy , Leukemia/genetics , Leukemia/metabolism , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/metabolism , Niacinamide/analogs & derivatives , Phenylurea Compounds , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Pyridines/administration & dosage , Pyridines/pharmacology , Sorafenib , TNF-Related Apoptosis-Inducing Ligand/administration & dosage
8.
Cancer Biol Ther ; 6(9): 1490-5, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17881904

ABSTRACT

Cancer stem cells are resistant to chemotherapy and provide an important target for drug development. We found that, surprisingly, the dye-effluxing side population (SP) within SW480 human colon tumor cells, a population defined to possess stem cell characteristics, expresses a 10-fold higher level of pro-apoptotic TRAIL receptor DR4 as compared to non-SP cells. The TRAIL receptors are activated by the anti-tumor host immune system through the TRAIL ligand. SW480 SP-cells express similar levels of another TRAIL receptor (DR5), as non-SP cells. SP-cells from multiple tumorigenic human cell lines, which are most often resistant to chemotherapeutic agents such as etoposide, cisplatin and 5-FU, are more sensitive to TRAIL than non-SP cells. SP-cells express higher levels of c-Myc than non-SP cells which may explain their sensitivity to TRAIL. We have found c-Myc activates DR4 transcription through E-box DNA-response elements located in the DR4 promoter, thereby increasing the expression of cell-surface pro-apoptotic death receptors in TRAIL-resistant cell lines. TRAIL sensitivity of SP-cells may represent a safeguard against malignancy, and therefore, offers a therapeutic window and opportunity.


Subject(s)
Colonic Neoplasms/drug therapy , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Proto-Oncogene Proteins c-myc/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Apoptosis , Cell Line, Tumor , Cell Separation , Flow Cytometry , Humans , Microscopy, Fluorescence , Models, Biological , Stem Cells/metabolism , Tumor Cells, Cultured
9.
Cancer Cell ; 12(1): 66-80, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17613437

ABSTRACT

Cells expressing oncogenic c-Myc are sensitized to TNF superfamily proteins. c-Myc also is an important factor in determining whether a cell is sensitive to TRAIL-induced apoptosis, and it is well established that the mitochondrial pathway is essential for apoptosis induced by c-Myc. We investigated whether c-Myc action on the mitochondria is required for TRAIL sensitivity and found that Myc sensitized cells with defective intrinsic signaling to TRAIL. TRAIL induced expression of antiapoptotic Mcl-1 and cIAP2 through activation of NF-kappaB. Both Myc and the multikinase inhibitor sorafenib block NF-kappaB. Combining sorafenib with TRAIL in vivo showed dramatic efficacy in TRAIL-resistant tumor xenografts. We propose the combination of TRAIL with sorafenib holds promise for further development.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/physiology , Benzenesulfonates/pharmacology , Inhibitor of Apoptosis Proteins/physiology , Neoplasm Proteins/physiology , Proto-Oncogene Proteins c-bcl-2/physiology , Proto-Oncogene Proteins c-myc/physiology , Pyridines/pharmacology , TNF-Related Apoptosis-Inducing Ligand/physiology , Animals , Apoptosis/drug effects , Baculoviral IAP Repeat-Containing 3 Protein , Cell Line, Tumor , Female , Humans , Inhibitor of Apoptosis Proteins/genetics , Mice , Mice, Nude , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Proteins/genetics , Niacinamide/analogs & derivatives , Phenylurea Compounds , Proto-Oncogene Proteins c-bcl-2/genetics , Sorafenib , Transcription, Genetic/physiology , Ubiquitin-Protein Ligases
10.
Oncologist ; 11(4): 342-57, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16614230

ABSTRACT

For several decades, apoptosis has taken center stage as the principal mechanism of programmed cell death in mammalian tissues. It also has been increasingly noted that conventional chemotherapeutic agents not only elicit apoptosis but other forms of nonapoptotic death such as necrosis, autophagy, mitotic catastrophe, and senescence. This review presents background on the signaling pathways involved in the different cell death outcomes. A re-examination of what we know about chemotherapy-induced death is vitally important in light of new understanding of nonapoptotic cell death signaling pathways. If we can precisely activate or inhibit molecules that mediate the diversity of cell death outcomes, perhaps we can succeed in more effective and less toxic chemotherapeutic regimens.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Death/drug effects , Neoplasms/metabolism , Signal Transduction/drug effects , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Autophagy/drug effects , Cellular Senescence/drug effects , Clinical Trials as Topic , Genes, Neoplasm/drug effects , Humans , Intracellular Signaling Peptides and Proteins/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Mitosis/drug effects , Necrosis/chemically induced , Neoplasms/drug therapy , Neoplasms/pathology , Receptors, Tumor Necrosis Factor/drug effects , Receptors, Tumor Necrosis Factor/metabolism
11.
Cancer Biol Ther ; 4(11): 1285-94, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16319523

ABSTRACT

HIF-1, a hypoxia inducible transcription factor, plays a pivotal role in the cellular response to hypoxia by activating genes involved in glucose metabolism, vascular remodeling, and erythropoiesis. We identified Mxi1, a c-Myc antagonist, as a novel target gene induced in hypoxia. Mxi1 was not induced in cells deficient in ARNT (HIF-1beta), suggesting that Mxi1 is a transcriptional target of the HIF-1 complex. Notably, c-Myc protein levels decreased during hypoxia but were stabilized by a proteasome inhibitor. Analysis of downstream transcriptional targets of c-Myc during hypoxia revealed that genes regulated by c-Myc, such as ornithine decarboxylase (ODC), were downregulated during hypoxia. In contrast, genes that are regulated by c-Myc and HIF-1, such as LDH-A, were upregulated. Mxi1 protects against c-Myc-dependent sensitization to hypoxia-induced apoptosis. The results suggest a coordinated mechanism for opposing c-Myc signaling during hypoxia that is mediated by a reduction in c-Myc levels, the induction of Mxi1, and a dominant effect of HIF-1 transcriptional activity.


Subject(s)
Apoptosis/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Hypoxia/physiology , Cytoprotection/physiology , Hypoxia-Inducible Factor 1/physiology , Proto-Oncogene Proteins c-myc/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Line , Cell Line, Tumor , HeLa Cells , Humans , Mice , Models, Biological
12.
Cancer Biol Ther ; 4(10): 1068-74, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16294025

ABSTRACT

Hypoxia induces Hif-1alpha and selects for loss of wild-type p53 function, both of which can promote tumor cell survival. We evaluated the ability of TRAIL to induce apoptosis of human tumor cell lines exposed to hypoxia. H460 lung cancer cells express low levels of Hif-1alpha, stabilize wild-type p53 during hypoxia, and undergo TRAIL-induced apoptosis. In U2OS osteosarcoma or PA1 ovarian teratocarcinoma cells, high levels of Hif-1alpha and low levels of stable p53 are detected during hypoxia, and cells undergo low levels of TRAIL-induced apoptosis as compared to H460 cells. H460 cells are sensitized to TRAIL-induced apoptosis, whereas U2OS are protected, and little apoptosis is observed in relatively TRAIL-resistant PA1 during hypoxia. Forced expression of Hif-1alpha is also surprisingly a potent inducer of apoptosis in wild-type p53 expressing H460 cells and further promotes TRAIL-induced apoptosis. TRAIL-sensitive wild-type p53-expressing HCT116 colon carcinoma cells modestly elevate Hif-1alpha levels and are equally or slightly more sensitive to TRAIL during hypoxia. In contrast, p53-null HCT116 have higher levels of Hif-1alpha during normoxia and are extremely sensitive to TRAIL, but are protected from TRAIL-induced apoptosis during hypoxia. We hypothesize that a hypoxic tumor microenvironment may alter sensitivity to TRAIL, which may be impacted by Hif-1alpha levels and p53 status. These findings suggest that particular attention to hypoxic regions of tumors and sensitizers to hypoxia-induced cell death may be required to optimize therapeutic combinations using TRAIL.


Subject(s)
Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/pharmacology , Apoptosis/drug effects , Cell Hypoxia/physiology , Colonic Neoplasms/drug therapy , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/pharmacology , Cell Line, Tumor , Dose-Response Relationship, Drug , HCT116 Cells , Humans , Hypoxia-Inducible Factor 1/genetics , Hypoxia-Inducible Factor 1/metabolism , TNF-Related Apoptosis-Inducing Ligand , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
13.
Mol Cell Biol ; 24(19): 8541-55, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15367674

ABSTRACT

Tumor necrosis factor alpha (TNF-alpha)-related apoptosis-inducing ligand (TRAIL) is a member of the TNF-alpha family of death receptor ligands and holds great therapeutic potential as a tumor cell-specific cytotoxic agent. Using a panel of established tumor cell lines and normal cells, we found a significant difference between the number of TRAIL-sensitive cells expressing high levels of c-myc and TRAIL-resistant cells expressing low levels of c-myc (P < 0.05, n = 19). We also found a direct linear correlation between c-myc levels and TRAIL sensitivity in TRAIL-sensitive cell lines (r = 0.94, n = 6). Overexpression of c-myc or activation of a myc-estrogen receptor (ER) fusion sensitized TRAIL-resistant cells to TRAIL. Conversely, small interfering RNA (siRNA)-mediated knockdown of c-myc significantly reduced both c-myc expression and TRAIL-induced apoptosis. The gene encoding the inhibitor of caspase activation, FLICE inhibitory protein (FLIP), appears to be a direct target of c-myc-mediated transcriptional repression. Overexpression of c-myc or activation of myc-estrogen receptor (ER) decreased FLIP levels both in cell culture and in mouse models of c-myc-induced tumorigenesis, while knocking down c-myc using siRNA increased FLIP expression. Chromatin immunoprecipitation and luciferase reporter analyses showed that c-myc binds and represses the human FLIP promoter. c-myc expression enhanced TRAIL-induced caspase 8 cleavage and FLIP cleavage at the death-inducing signaling complex. Combined siRNA-mediated knockdown of FLIP and c-myc resensitized cells to TRAIL. Therefore, c-myc down-regulation of FLIP expression provides a universal mechanism to explain the ability of c-myc to sensitize cells to death receptor stimuli. In addition, identification of c-myc as a major determinant of TRAIL sensitivity provides a potentially important screening tool for identification of TRAIL-sensitive tumors.


Subject(s)
Carrier Proteins/genetics , Gene Expression Regulation/physiology , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Apoptosis/physiology , Apoptosis Regulatory Proteins , CASP8 and FADD-Like Apoptosis Regulating Protein , Carrier Proteins/biosynthesis , Humans , Mice , Promoter Regions, Genetic , RNA Interference , RNA, Messenger/metabolism , TNF-Related Apoptosis-Inducing Ligand , Transcription, Genetic/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...