PMID-sentid Pub_year Sent_text compound_name comp_offset prot_official_name organism prot_offset 11078493-1 2000 PURPOSE: To examine the feasibility of using fenretinide (4-HPR) for the prevention and treatment of prostate cancer. Fenretinide 45-56 haptoglobin-related protein Homo sapiens 60-63 11122886-1 2000 Retinoids are derivatives of vitamin A that include all- trans-retinoic acid (ATRA), 13-cis-retinoic acid, (13-cis-RA), and fenretinide (4-HPR). Fenretinide 124-135 haptoglobin-related protein Homo sapiens 139-142 11054663-5 2000 In this report, we provide evidence that DPH2L is down-regulated during differentiation or apoptosis in several cancer cell lines after treatment with all-trans RA or N-(4-hydroxyphenyl)retinamide and during cell-cycle arrest. Fenretinide 167-196 diphthamide biosynthesis 1 Homo sapiens 41-46 11027625-1 2000 Fenretinide (4-HPR) is a synthetic retinoid with cancer chemopreventative potential and clinically manageable side effects, compared to the prototype retinoid, all-trans retinoic acid (RA). Fenretinide 0-11 haptoglobin-related protein Homo sapiens 15-18 11010810-3 2000 Fenretinide induced high levels of caspase-dependent apoptosis accompanied by an increase in free radicals and the release of cytochrome c in the absence of mitochondrial permeability transition. Fenretinide 0-11 cytochrome c, somatic Homo sapiens 126-138 11010810-7 2000 These results suggest that the effector pathway of fenretinide-induced apoptosis of neuroblastoma is caspase dependent, involving mitochondrial release of cytochrome c independently of permeability changes, and mediated by specific RARs. Fenretinide 51-62 cytochrome c, somatic Homo sapiens 155-167 11027625-0 2000 Apoptosis and restriction of G(1)/S cell cycle by fenretinide in Burkitt"s lymphoma mutu I cell line accessed with bcl-6 down-regulation. Fenretinide 50-61 BCL6 transcription repressor Homo sapiens 115-120 10999486-2 2000 A satisfactory correlation was found between the HINT scores and the experimental dissociation constants of three of the ligands, fenretinide, N-ethylretinamide and all-trans retinol, despite their hydrophobic nature. Fenretinide 130-141 histidine triad nucleotide binding protein 1 Homo sapiens 49-53 10824459-0 2000 Effects of N-(4-hydroxyphenyl) retinamide on urokinase-type plasminogen activator and plasminogen activator inhibitor-1 in prostate adenocarcinoma cell lines. Fenretinide 11-41 serpin family E member 1 Homo sapiens 86-119 10962560-0 2000 pRb and Cdk regulation by N-(4-hydroxyphenyl)retinamide. Fenretinide 26-55 RB transcriptional corepressor 1 Homo sapiens 0-3 10969771-0 2000 Fenretinide-induced caspase 3 activity involves increased protein stability in a mechanism distinct from reactive oxygen species elevation. Fenretinide 0-11 caspase 3 Homo sapiens 20-29 10969771-1 2000 Fenretinide (4-HPR) is a synthetic retinoid that displays a broad range of biological effects and has also demonstrated clinical efficacy as a chemopreventative agent. Fenretinide 0-11 haptoglobin-related protein Homo sapiens 15-18 10917548-1 2000 The effect of the chemopreventive synthetic retinoid N-(4-hydroxyphenyl)-retinamide (4-HPR) on aromatase activity and expression was examined. Fenretinide 53-83 haptoglobin-related protein Homo sapiens 87-90 10918204-0 2000 Time course of fenretinide-induced modulation of circulating insulin-like growth factor (IGF)-i, IGF-II and IGFBP-3 in a bladder cancer chemoprevention trial. Fenretinide 15-26 insulin like growth factor 1 Homo sapiens 61-95 10918204-0 2000 Time course of fenretinide-induced modulation of circulating insulin-like growth factor (IGF)-i, IGF-II and IGFBP-3 in a bladder cancer chemoprevention trial. Fenretinide 15-26 insulin like growth factor 2 Homo sapiens 97-103 10918204-0 2000 Time course of fenretinide-induced modulation of circulating insulin-like growth factor (IGF)-i, IGF-II and IGFBP-3 in a bladder cancer chemoprevention trial. Fenretinide 15-26 insulin like growth factor binding protein 3 Homo sapiens 108-115 10918204-4 2000 We have previously shown that the synthetic retinoid fenretinide (4-HPR) lowers plasma IGF-I in pre-menopausal breast cancer patients. Fenretinide 53-64 haptoglobin-related protein Homo sapiens 68-71 10918204-4 2000 We have previously shown that the synthetic retinoid fenretinide (4-HPR) lowers plasma IGF-I in pre-menopausal breast cancer patients. Fenretinide 53-64 insulin like growth factor 1 Homo sapiens 87-92 10918204-6 2000 Repeated measures analysis, after adjustment for age, body mass index (BMI) and year of study, showed a significant effect of fenretinide on IGF-I levels, which were further lowered after the second year of treatment and only partially recovered after drug discontinuation. Fenretinide 126-137 insulin like growth factor 1 Homo sapiens 141-146 10955773-3 2000 One such strategy is to use retinoids like N-(4-hydroxyphenyl)retinamide (4-HPR), which has been found to possess chemopreventive activities in preclinical studies. Fenretinide 43-72 haptoglobin-related protein Homo sapiens 76-79 10772821-0 2000 BAG-1 promotes apoptosis induced by N-(4-hydroxyphenyl)retinamide in human cervical carcinoma cells. Fenretinide 36-65 BAG cochaperone 1 Homo sapiens 0-5 10772821-1 2000 N-(4-hydroxyphenyl)retinamide (4-HPR) is a synthetic apoptosis-inducing retinoid with cancer chemopreventive properties and lower toxicity than all-trans retinoic acid. Fenretinide 0-29 haptoglobin-related protein Homo sapiens 33-36 10824459-1 2000 Previous investigations have demonstrated that a synthetic retinoid, N-(4-hydroxyphenyl) retinamide (4-HPR), inhibits the invasion of prostate adenocarcinoma in vitro. Fenretinide 69-99 haptoglobin-related protein Homo sapiens 103-106 10448131-3 1999 All-trans-N-(4-hydroxyphenyl)retinamide (4HPR or fenretinide) is a synthetic retinoid that is reported to have fewer side-effects compared to naturally occurring retinoids such as all-trans retinoic acid (ATRA) and 9-cis retinoic acid. Fenretinide 0-39 haptoglobin-related protein Homo sapiens 42-45 10706389-1 2000 Fenretinide, N-(4-hydroxyphenyl)retinamide (4-HPR), is a cancer chemopreventive and antiproliferative agent whose mechanism of action is unknown. Fenretinide 0-11 haptoglobin-related protein Homo sapiens 46-49 10706389-1 2000 Fenretinide, N-(4-hydroxyphenyl)retinamide (4-HPR), is a cancer chemopreventive and antiproliferative agent whose mechanism of action is unknown. Fenretinide 13-42 haptoglobin-related protein Homo sapiens 46-49 10637240-1 2000 PURPOSE: N-(4-hydroxyphenyl) retinamide (?4-HPR, Fenretinide; R.W. Fenretinide 9-39 haptoglobin-related protein Homo sapiens 44-47 10597238-0 1999 Implication of mitochondria-derived reactive oxygen species, cytochrome C and caspase-3 in N-(4-hydroxyphenyl)retinamide-induced apoptosis in cervical carcinoma cells. Fenretinide 91-120 cytochrome c, somatic Homo sapiens 61-73 10597238-0 1999 Implication of mitochondria-derived reactive oxygen species, cytochrome C and caspase-3 in N-(4-hydroxyphenyl)retinamide-induced apoptosis in cervical carcinoma cells. Fenretinide 91-120 caspase 3 Homo sapiens 78-87 10523855-0 1999 4HPR triggers apoptosis but not differentiation in retinoid sensitive and resistant human embryonal carcinoma cells through an RARgamma independent pathway. Fenretinide 0-4 retinoic acid receptor gamma Homo sapiens 127-135 10523855-8 1999 In contrast, N-(4-hydroxyphenyl)retinamide (4HPR), a reported transcriptional activator of RARgamma was shown to potently induce growth inhibition and apoptosis in both NT2/D1 and NT2/D1-R1 cells. Fenretinide 13-42 haptoglobin-related protein Homo sapiens 45-48 10523855-8 1999 In contrast, N-(4-hydroxyphenyl)retinamide (4HPR), a reported transcriptional activator of RARgamma was shown to potently induce growth inhibition and apoptosis in both NT2/D1 and NT2/D1-R1 cells. Fenretinide 13-42 retinoic acid receptor gamma Homo sapiens 91-99 10762020-5 2000 Both tamoxifen and the synthetic retinoid fenretinide (4-HPR) have been shown to decrease plasma IGF-I levels. Fenretinide 42-53 haptoglobin-related protein Homo sapiens 57-60 10762020-5 2000 Both tamoxifen and the synthetic retinoid fenretinide (4-HPR) have been shown to decrease plasma IGF-I levels. Fenretinide 42-53 insulin like growth factor 1 Homo sapiens 97-102 10570055-0 1999 c-Jun N-terminal kinase mediates apoptotic signaling induced by N-(4-hydroxyphenyl)retinamide. Fenretinide 64-93 mitogen-activated protein kinase 8 Homo sapiens 0-23 10570055-1 1999 N-(4-Hydroxyphenyl)retinamide (4-HPR), a retinoic acid analog, induces apoptosis in several cell types. Fenretinide 0-29 haptoglobin-related protein Homo sapiens 33-36 10572559-17 1999 Based on experimental studies, the vitamin A analogue 4-hydroxyphenyl retinamide (4-HPR) was shown to delay and reduce carcinogen-induced breast cancer. Fenretinide 54-80 haptoglobin-related protein Homo sapiens 84-87 10427131-3 1999 Four glioma cell lines (C6, 9L, Med3 and U87) were treated with fenretinide. Fenretinide 64-75 small nucleolar RNA, C/D box 87 Homo sapiens 41-44 10448131-3 1999 All-trans-N-(4-hydroxyphenyl)retinamide (4HPR or fenretinide) is a synthetic retinoid that is reported to have fewer side-effects compared to naturally occurring retinoids such as all-trans retinoic acid (ATRA) and 9-cis retinoic acid. Fenretinide 49-60 haptoglobin-related protein Homo sapiens 42-45 10432011-0 1999 Modulation of the malignant phenotype of human prostate cancer cells by N-(4-hydroxyphenyl)retinamide (4-HPR). Fenretinide 72-101 haptoglobin-related protein Homo sapiens 105-108 10357800-0 1999 Retinoic acid and 4-hydroxyphenylretinamide induce growth inhibition and tissue transglutaminase through different signal transduction pathways in mouse fibroblasts (NIH 3T3 cells). Fenretinide 18-43 transglutaminase 2, C polypeptide Mus musculus 73-96 10357800-1 1999 4-Hydroxyphenylretinamide (4-HPR) is a synthetic retinoid with minimal toxicity and favorable pharmacokinetics during long-term administration to patients in clinical trials. Fenretinide 0-25 haptoglobin-related protein Homo sapiens 29-32 10328240-0 1999 Role of retinoic acid receptor overexpression in sensitivity to fenretinide and tumorigenicity of human ovarian carcinoma cells. Fenretinide 64-75 retinoic acid receptor alpha Homo sapiens 8-30 10328240-1 1999 The role of retinoic acid receptor (RAR) expression in sensitivity to N-(4-hydroxyphenyl)retinamide (4HPR or fenretinide) as well as on the tumorigenicity of human ovarian carcinoma cells was examined. Fenretinide 70-99 retinoic acid receptor alpha Homo sapiens 36-39 10328240-1 1999 The role of retinoic acid receptor (RAR) expression in sensitivity to N-(4-hydroxyphenyl)retinamide (4HPR or fenretinide) as well as on the tumorigenicity of human ovarian carcinoma cells was examined. Fenretinide 101-105 retinoic acid receptor alpha Homo sapiens 12-34 10328240-1 1999 The role of retinoic acid receptor (RAR) expression in sensitivity to N-(4-hydroxyphenyl)retinamide (4HPR or fenretinide) as well as on the tumorigenicity of human ovarian carcinoma cells was examined. Fenretinide 101-105 retinoic acid receptor alpha Homo sapiens 36-39 10328240-1 1999 The role of retinoic acid receptor (RAR) expression in sensitivity to N-(4-hydroxyphenyl)retinamide (4HPR or fenretinide) as well as on the tumorigenicity of human ovarian carcinoma cells was examined. Fenretinide 109-120 retinoic acid receptor alpha Homo sapiens 12-34 10328240-1 1999 The role of retinoic acid receptor (RAR) expression in sensitivity to N-(4-hydroxyphenyl)retinamide (4HPR or fenretinide) as well as on the tumorigenicity of human ovarian carcinoma cells was examined. Fenretinide 109-120 retinoic acid receptor alpha Homo sapiens 36-39 10473110-10 1999 Fenretinide treatment resulted in cleavage of poly ADP-ribose polymerase, indicating an activation of the caspase 3. Fenretinide 0-11 poly(ADP-ribose) polymerase 1 Homo sapiens 46-72 10473110-10 1999 Fenretinide treatment resulted in cleavage of poly ADP-ribose polymerase, indicating an activation of the caspase 3. Fenretinide 0-11 caspase 3 Homo sapiens 106-115 10393722-1 1999 BACKGROUND: The synthetic retinoid N-(4-hydroxyphenyl)retinamide (4-HPR or fenretinide) is toxic to myeloid leukemia and cervical carcinoma cell lines, probably in part due to its ability to increase levels of reactive oxygen species (ROS). Fenretinide 35-64 haptoglobin-related protein Homo sapiens 68-71 10393722-1 1999 BACKGROUND: The synthetic retinoid N-(4-hydroxyphenyl)retinamide (4-HPR or fenretinide) is toxic to myeloid leukemia and cervical carcinoma cell lines, probably in part due to its ability to increase levels of reactive oxygen species (ROS). Fenretinide 75-86 haptoglobin-related protein Homo sapiens 68-71 10334206-2 1999 In this study, we used cell proliferation and telomerase in MNU (N-methyl-N-nitrosourea)-induced mammary carcinomas as targets for assessing the response of tumor cells to 4-(hydroxyphenyl)retinamide (4-HPR), a known inhibitor of mammary carcinogenesis in animal models and premenopausal women. Fenretinide 172-199 haptoglobin-related protein Homo sapiens 203-206 10432011-4 1999 This study focuses on the ability of N-(4-hydroxyphenyl)-retinamide (4-HPR), a synthetic retinoid, to reverse malignant characteristics towards a normal phenotype, using the human prostate carcinoma cell line DU-145. Fenretinide 37-67 haptoglobin-related protein Homo sapiens 71-74 10188730-4 1999 Previously, we reported that the synthetic retinoid fenretinide (HPR) is an inducer of apoptosis in neuroblastoma (NB) cells, sharing the neuroectodermal origin with melanoma cells. Fenretinide 52-63 haptoglobin-related protein Homo sapiens 65-68 10325501-1 1999 An NCI-sponsored, phase II trial of N-(4-hydroxyphenyl)- retinamide (4-HPR) in patients with organ-confined prostate cancer in the period prior to radical prostatectomy was carried out. Fenretinide 36-67 haptoglobin-related protein Homo sapiens 71-74 10069458-0 1999 Inhibition of aberrant proliferation and induction of apoptosis in HER-2/neu oncogene transformed human mammary epithelial cells by N-(4-hydroxyphenyl)retinamide. Fenretinide 132-161 erb-b2 receptor tyrosine kinase 2 Homo sapiens 67-76 9920792-0 1999 Carcinoma cell lines resistant for growth inhibition and apoptosis to retinoic acid are responsive to 4-hydroxy-phenyl-retinamide: correlation with tissue transglutaminase. Fenretinide 102-129 transglutaminase 2 Homo sapiens 148-171 10709674-3 1999 As discussed above, all six potential chemopreventive agents, aspirin, 2-CPR, DFMO, 4-HPR, piroxicam, and 9-cis-retinoic acid, decreased the level of AOM-induced ACF. Fenretinide 84-89 ACF Homo sapiens 162-165 9923820-1 1999 PURPOSE: Fenretinide [N-(4-hydroxyphenyl)retinamide, 4HPR], a synthetic retinoid, is a potent inducer of apoptosis in small-cell lung cancer (SCLC) cell lines that may act through the generation of reactive oxygen species, suggesting that it may enhance the activity of other cytotoxic agents. Fenretinide 9-20 haptoglobin-related protein Homo sapiens 54-57 9923820-1 1999 PURPOSE: Fenretinide [N-(4-hydroxyphenyl)retinamide, 4HPR], a synthetic retinoid, is a potent inducer of apoptosis in small-cell lung cancer (SCLC) cell lines that may act through the generation of reactive oxygen species, suggesting that it may enhance the activity of other cytotoxic agents. Fenretinide 22-51 haptoglobin-related protein Homo sapiens 54-57 9395213-0 1997 Inhibition by N-(4-hydroxyphenyl)retinamide and all-trans-retinoic acid of exogenous and endogenous development of putative preneoplastic, glutathione S-transferase placental form-positive lesions in the livers of rats. Fenretinide 14-43 hematopoietic prostaglandin D synthase Rattus norvegicus 139-164 10578478-0 1999 N-(4-hydroxyphenyl)retinamide activation of transforming growth factor-beta and induction of apoptosis in human breast cancer cells. Fenretinide 0-29 transforming growth factor beta 1 Homo sapiens 44-75 10578478-1 1999 N-(4 hydroxyphenyl)retinamide (4-HPR), a synthetic derivative of all-trans-retinoic acid, induces DNA synthesis arrest and apoptosis in human breast cancer cells in a dose- and time-dependent manner. Fenretinide 0-29 haptoglobin-related protein Homo sapiens 33-36 9724094-4 1998 Cyclin D1 and cdk4 expression and Rb phosphorylation were significantly reduced, by 40-75%, after 24 hr of treatment with RA or 4HPR; these decreases were either transient, e.g., only at 24 hr for cdk4, or sustained for 72 hr. Fenretinide 128-132 cyclin D1 Homo sapiens 0-9 9724094-4 1998 Cyclin D1 and cdk4 expression and Rb phosphorylation were significantly reduced, by 40-75%, after 24 hr of treatment with RA or 4HPR; these decreases were either transient, e.g., only at 24 hr for cdk4, or sustained for 72 hr. Fenretinide 128-132 cyclin dependent kinase 4 Homo sapiens 14-18 9724094-4 1998 Cyclin D1 and cdk4 expression and Rb phosphorylation were significantly reduced, by 40-75%, after 24 hr of treatment with RA or 4HPR; these decreases were either transient, e.g., only at 24 hr for cdk4, or sustained for 72 hr. Fenretinide 128-132 cyclin dependent kinase 4 Homo sapiens 197-201 9724094-10 1998 Simultaneous treatment of cells with 4HPR and 25-100 microM fumonisin B1 resulted in a dose-dependent reduction in the elevation in ceramide, the extent of PARP cleavage, and induction of apoptosis. Fenretinide 37-41 poly(ADP-ribose) polymerase 1 Homo sapiens 156-160 9662255-0 1998 Effects of retinoic acid and fenretinide on the c-erbB-2 expression, growth and cisplatin sensitivity of breast cancer cells. Fenretinide 29-40 erb-b2 receptor tyrosine kinase 2 Homo sapiens 48-56 9662255-1 1998 We investigated the effects of all-trans retinoic acid (ATRA) and fenretinide (4-HPR) on c-erbB-2 expression in SK-BR-3, BT-474 and MCF-7 breast cancer cells and on the growth, differentiation, apoptosis and cisplatin (CDDP) sensitivity of SK-BR-3 cells. Fenretinide 66-77 erb-b2 receptor tyrosine kinase 2 Homo sapiens 89-97 9626341-0 1998 Effect of fenretinide on plasma IGF-I and IGFBP-3 in early breast cancer patients. Fenretinide 10-21 insulin like growth factor 1 Homo sapiens 32-37 9626341-0 1998 Effect of fenretinide on plasma IGF-I and IGFBP-3 in early breast cancer patients. Fenretinide 10-21 insulin like growth factor binding protein 3 Homo sapiens 42-49 9797705-0 1998 Metabolism of N-[4-hydroxyphenyl]retinamide (4-HPR) to N-[4-methoxyphenyl]retinamide (4-MPR) may serve as a biomarker for its efficacy against human breast cancer and melanoma cells. Fenretinide 14-43 haptoglobin-related protein Homo sapiens 47-50 9797705-0 1998 Metabolism of N-[4-hydroxyphenyl]retinamide (4-HPR) to N-[4-methoxyphenyl]retinamide (4-MPR) may serve as a biomarker for its efficacy against human breast cancer and melanoma cells. Fenretinide 14-43 progesterone receptor membrane component 1 Homo sapiens 88-91 9797705-1 1998 A clinical trial of N-[4-hydroxyphenyl]retinamide (4-HPR) has been in progress for the past 4 years to evaluate its role in chemoprevention of breast cancer. Fenretinide 20-49 haptoglobin-related protein Homo sapiens 53-56 9554442-0 1998 Retinoic acid receptor beta expression and growth inhibition of gynecologic cancer cells by the synthetic retinoid N-(4-hydroxyphenyl) retinamide. Fenretinide 115-145 retinoic acid receptor beta Homo sapiens 0-27 9554442-1 1998 BACKGROUND: The synthetic retinoid N-(4-hydroxyphenyl) retinamide (4HPR) can inhibit the growth of tumor cells. Fenretinide 35-65 haptoglobin-related protein Homo sapiens 68-71 9560096-3 1998 The synthetic retinoid fenretinide (N-(4-hydroxyphenyl)retinamide) (HPR) has significant antiproliferative activity against a number of animal and human malignancies and has been used in clinical trials as a chemopreventive agent in patients with breast and prostate cancer and oral leukoplakia. Fenretinide 23-34 haptoglobin-related protein Homo sapiens 68-71 9560096-3 1998 The synthetic retinoid fenretinide (N-(4-hydroxyphenyl)retinamide) (HPR) has significant antiproliferative activity against a number of animal and human malignancies and has been used in clinical trials as a chemopreventive agent in patients with breast and prostate cancer and oral leukoplakia. Fenretinide 36-65 haptoglobin-related protein Homo sapiens 68-71 9639444-0 1998 Effects of fenretinide (4-HPR) on dark adaptation. Fenretinide 11-22 haptoglobin-related protein Homo sapiens 26-29 9639444-1 1998 OBJECTIVES: To assess the alterations in dark adaptation induced by low (200 mg/d) doses of fenretinide (4-HPR), to assess whether these effects were cumulative and whether they were reversible, and to attempt to elucidate the mechanism underlying the changes in night vision. Fenretinide 92-103 haptoglobin-related protein Homo sapiens 107-110 9649125-0 1998 Modulation of the insulin-like growth factor-I system by N-(4-hydroxyphenyl)-retinamide in human breast cancer cell lines. Fenretinide 57-87 insulin like growth factor 1 Homo sapiens 18-46 9649125-2 1998 We studied and compared the effects of N-(4-hydroxyphenyl)-retinamide (4-HPR) in terms of growth inhibition and modulation of the IGF-I system in ER+ (MCF-7) and oestrogen receptor-negative (ER-) (MDA-MB231) breast cancer cell lines. Fenretinide 39-69 insulin like growth factor 1 Homo sapiens 130-135 9649125-6 1998 Immunoreactive IGF-I-like protein concentration in conditioned medium was reduced by 38% in MCF-7 and by 90% in MDA-MB231 cell lines following treatment for 48 h with 5 microM 4-HPR. Fenretinide 176-181 insulin like growth factor 1 Homo sapiens 15-20 9649125-8 1998 Exposure to 5 microM 4-HPR for 48 h inhibited [125I]IGF-I binding and Scatchard analysis revealed a decrease of more than 50% in maximum binding capacity (Bmax) and a reduced receptor number/cell in both cancer cell lines. Fenretinide 21-26 insulin like growth factor 1 Homo sapiens 52-57 9649125-10 1998 We conclude that 4-HPR induces a significant down-regulation of the IGF-I system in both ER+ (MCF-7) and ER- (MDA-MB231) breast cancer cell lines. Fenretinide 17-22 insulin like growth factor 1 Homo sapiens 68-73 8857507-0 1996 N-(4-hydroxyphenyl)-retinamide increases lecithin:retinol acyltransferase activity in rat liver. Fenretinide 0-30 lecithin retinol acyltransferase Rattus norvegicus 41-73 9316650-1 1997 BACKGROUND: Although fenretinide (4-HPR) is currently being evaluated in a phase II clinical study for the chemoprevention of prostate cancer [Greenwald et al. Fenretinide 21-32 haptoglobin-related protein Homo sapiens 36-39 9125196-7 1997 We found that 4-hydroxyphenylretinamide (4-HPR) inhibited the catabolism of RA. Fenretinide 14-39 haptoglobin-related protein Homo sapiens 43-46 9526578-2 1997 The Fenretinide (4-HPR) Breast Cancer Study is a randomized multicenter clinical trial originally designed and conducted by the investigators of the Istituto Nazionale Tumori of Milan. Fenretinide 4-15 haptoglobin-related protein Homo sapiens 19-22 9388681-0 1997 Fenretinide inhibits phorbol ester-induced cyclooxygenase-2 expression in human colon adenocarcinoma cells. Fenretinide 0-11 prostaglandin-endoperoxide synthase 2 Homo sapiens 43-59 9168437-1 1997 We demonstrate that N-(4-hydroxyphenyl)-all-trans-retinamide (4-HPR), a synthetic retinoic acid (RA) derivative, is a potent and selective inducer of apoptosis in malignant T lymphoid cells, but has little effect on normal lymphoid cells of the thymus or spleen. Fenretinide 20-60 haptoglobin-related protein Homo sapiens 64-67 8993839-0 1997 Fenretinide: induction of apoptosis and endogenous transforming growth factor beta in PC-3 prostate cancer cells. Fenretinide 0-11 transforming growth factor beta 1 Homo sapiens 51-82 8993839-1 1997 N-(4-Hydroxyphenyl)retinamide (4-HPR, Fenretinide) is a retinoid derivative with antineoplastic activity in various tumor types including prostate carcinoma. Fenretinide 0-29 haptoglobin-related protein Homo sapiens 33-36 8993839-1 1997 N-(4-Hydroxyphenyl)retinamide (4-HPR, Fenretinide) is a retinoid derivative with antineoplastic activity in various tumor types including prostate carcinoma. Fenretinide 38-49 haptoglobin-related protein Homo sapiens 33-36 9018085-0 1996 Effects of topical treatment with fenretinide (4-HPR) and plasma vitamin A levels in patients with actinic keratoses. Fenretinide 34-45 haptoglobin-related protein Homo sapiens 49-52 9018085-1 1996 Eighteen patients with facial actinic keratoses were treated with the retinoid fenretinide (4-HPR), applied topically twice-daily for 3 months. Fenretinide 79-90 haptoglobin-related protein Homo sapiens 94-97 8948605-1 1996 Both retinoic acid (RA) and the synthetic retinoid N-(4-hydroxyphenyl)retinamide (4HPR) have shown efficacy in head and neck cancer chemoprevention trials. Fenretinide 51-80 haptoglobin-related protein Homo sapiens 83-86 8857507-3 1996 We have conducted studies to examine the effects of 4-HPR on the activity of the enzyme lecithin:retinol acyltransferase (LRAT). Fenretinide 52-57 lecithin retinol acyltransferase Rattus norvegicus 88-120 8857507-3 1996 We have conducted studies to examine the effects of 4-HPR on the activity of the enzyme lecithin:retinol acyltransferase (LRAT). Fenretinide 52-57 lecithin retinol acyltransferase Rattus norvegicus 122-126 8857507-5 1996 To determine whether 4-HPR, like retinoic acid, is able to induce liver LRAT activity, vitamin A-deficient rats having negligible liver LRAT activity were treated with single doses of 4-HPR (0.02-2.5 mg) and liver homogenates were assayed for LRAT activity using 3H-retinol bound to the cellular-retinol binding protein, CRBP, as substrate. Fenretinide 21-26 lecithin retinol acyltransferase Rattus norvegicus 72-76 8857507-6 1996 Treatment with 4-HPR resulted in a dose- and time-dependent increase in liver LRAT activity which reached a maximum at 24 h. The activity of LRAT assayed in vitro and of hepatic 3H-retinyl ester content determined after an in vivo pulse of 3H-retinol were highly correlated (r = 0.802, P < 0.0002). Fenretinide 15-20 lecithin retinol acyltransferase Rattus norvegicus 78-82 8857507-6 1996 Treatment with 4-HPR resulted in a dose- and time-dependent increase in liver LRAT activity which reached a maximum at 24 h. The activity of LRAT assayed in vitro and of hepatic 3H-retinyl ester content determined after an in vivo pulse of 3H-retinol were highly correlated (r = 0.802, P < 0.0002). Fenretinide 15-20 lecithin retinol acyltransferase Rattus norvegicus 141-145 8640761-1 1996 The activities of N-(4-hydroxyphenyl)retinamide [(4-HPR), Fenretinide] and all-trans-retinoic acid (RA) were determined for (a) the inhibition of cell proliferation; (b) the activation of human retinoid receptor-mediated target gene expression; (c) the inhibition of estradiol- and progesterone-induced gene activation in breast cancer cell lines; and (d) the regulation of the expression of tumor suppressor retinoblastoma protein. Fenretinide 18-47 haptoglobin-related protein Homo sapiens 52-55 9063520-0 1996 Chemoprevention of breast cancer with fenretinide (4-HPR): study of long-term visual and ophthalmologic tolerability. Fenretinide 38-49 haptoglobin-related protein Homo sapiens 53-56 9063520-1 1996 BACKGROUND: Fenretinide (4-HPR) is a synthetic retinoid being clinically tested in the chemoprevention of different tumors and precancerous lesions. Fenretinide 12-23 haptoglobin-related protein Homo sapiens 27-30 8645719-0 1996 Retinol-binding protein secretion from the liver of N-(4-hydroxyphenyl) retinamide-treated rats. Fenretinide 52-82 retinol binding protein 4 Rattus norvegicus 0-23 8645719-1 1996 N-(4-Hydroxyphenyl)retinamide (HPR; Fenretinide), a synthetic retinoid possessing antitumor activity, depresses plasma retinol and retinol-binding protein (RBP) concentrations. Fenretinide 0-29 retinol binding protein 4 Rattus norvegicus 131-154 8645719-1 1996 N-(4-Hydroxyphenyl)retinamide (HPR; Fenretinide), a synthetic retinoid possessing antitumor activity, depresses plasma retinol and retinol-binding protein (RBP) concentrations. Fenretinide 0-29 retinol binding protein 4 Rattus norvegicus 156-159 8645719-1 1996 N-(4-Hydroxyphenyl)retinamide (HPR; Fenretinide), a synthetic retinoid possessing antitumor activity, depresses plasma retinol and retinol-binding protein (RBP) concentrations. Fenretinide 36-47 retinol binding protein 4 Rattus norvegicus 131-154 8645719-1 1996 N-(4-Hydroxyphenyl)retinamide (HPR; Fenretinide), a synthetic retinoid possessing antitumor activity, depresses plasma retinol and retinol-binding protein (RBP) concentrations. Fenretinide 36-47 retinol binding protein 4 Rattus norvegicus 156-159 8687125-1 1996 The O-glucuronide analog of N-(4-hydroxyphenyl)retinamide (4-HPROG) has shown a greater chemopreventive activity than the parent N-(4-hydroxyphenyl)retinamide (4-HPR). Fenretinide 28-57 haptoglobin-related protein Homo sapiens 61-64 8713132-0 1996 Differential effects of retinoic acid (RA) and N-(4-hydroxyphenyl) retinamide (4-HPR) on cell growth, induction of differentiation, and changes in p34cdc2, Bcl-2, and actin expression in the human promyelocytic HL-60 leukemic cells. Fenretinide 47-77 haptoglobin-related protein Homo sapiens 81-84 8651965-1 1996 OBJECTIVE: To evaluate the efficacy and tolerability of N-[4 hydroxyphenyl] retinamide (4-HPR), a synthetic retinoid, in the treatment of rheumatoid arthritis (RA). Fenretinide 56-86 haptoglobin-related protein Homo sapiens 90-93 21552953-0 1995 N-(4-hydroxyphenyl)retinamide induces apoptosis in human leukemia hl-60 cells and mediates vimentin down-regulation. Fenretinide 0-29 vimentin Homo sapiens 91-99 8621233-0 1996 Induction of apoptosis by fenretinide (4HPR) in human ovarian carcinoma cells and its association with retinoic acid receptor expression. Fenretinide 26-37 haptoglobin-related protein Homo sapiens 40-43 7473815-3 1995 Fenretinide [N-(4-hydroxyphenyl)retinamide; HPR] is a synthetic retinoid with minimal toxicity and favorable pharmacokinetics during long-term administration to patients in clinical trials. Fenretinide 0-11 haptoglobin-related protein Homo sapiens 44-47 7473815-3 1995 Fenretinide [N-(4-hydroxyphenyl)retinamide; HPR] is a synthetic retinoid with minimal toxicity and favorable pharmacokinetics during long-term administration to patients in clinical trials. Fenretinide 13-42 haptoglobin-related protein Homo sapiens 44-47 9816026-1 1995 The synthetic retinoid, N-(4-hydroxyphenyl)retinamide (4-HPR; Fenretinide), is a cancer chemopreventive and antiproliferative agent whose mechanism of action is unknown. Fenretinide 24-53 haptoglobin-related protein Homo sapiens 57-60 7586155-0 1995 N-(4-hydroxyphenyl)retinamide (4-HPR)-mediated biological actions involve retinoid receptor-independent pathways in human breast carcinoma. Fenretinide 0-29 haptoglobin-related protein Homo sapiens 33-36 7586155-2 1995 N-(4-hydroxyphenyl) retinamide (4-HPR), a derivative of all-trans-retinoic acid (RA) is currently in clinical trials as a chemopreventive agent for breast cancer. Fenretinide 0-30 haptoglobin-related protein Homo sapiens 34-37 7586162-8 1995 Although retinol and 4HPR cross-competed for RBP binding, N-phenylretinamide, in which the 4-hydroxyl group is absent, and N-(4-methoxyphenyl)retinamide, a major cellular metabolite of 4HPR, in which the hydroxyl group is blocked, did not show affinity for the binding protein. Fenretinide 21-25 retinol binding protein 4 Homo sapiens 45-48 8595390-0 1995 The synthetic retinoid N-(4-hydroxyphenyl) retinamide (4-HPR) exerts antiproliferative and apoptosis-inducing effects in the androgen-independent human prostatic JCA-1 cells. Fenretinide 23-53 haptoglobin-related protein Homo sapiens 57-60 21552857-2 1995 All-trans retinoic acid (ATRA) and N-(4-hydroxyphenyl)retinamide (4HPR) are currently being evaluated in clinical trials for their potential use in cancer chemoprevention and therapy. Fenretinide 35-64 haptoglobin-related protein Homo sapiens 67-70 7654032-2 1995 In this study, a series of experiments were performed to examine the effects of N-(4-hydroxyphenyl) retinamide (4-HPR, Fenretinide) on cellular adhesion, motility and proteolytic activity of established prostate cancer cell lines, TSU-PR 1 and PC-3. Fenretinide 112-117 proprotein convertase subtilisin/kexin type 1 Homo sapiens 231-248 7654032-4 1995 Radiomigration assay also demonstrated that 4-HPR concentration of 10(-6) M reduced the cellular motility by 29% in TSU-PR1 and 28% in PC-3 cells (p < 0.05). Fenretinide 44-49 transmembrane protein 37 Homo sapiens 120-123 9816026-1 1995 The synthetic retinoid, N-(4-hydroxyphenyl)retinamide (4-HPR; Fenretinide), is a cancer chemopreventive and antiproliferative agent whose mechanism of action is unknown. Fenretinide 62-73 haptoglobin-related protein Homo sapiens 57-60 7744071-12 1995 Additionally, the bulky end groups of fenretinide and N-ethyl retinamide replacing the retinol hydroxyl group have been found to prevent retinoid binding to CRBP. Fenretinide 38-49 retinol binding protein 1 Bos taurus 157-161 7538673-7 1995 We found that mac25 accumulates in senescent cells and is up-regulated in normal, growing mammary epithelial cells by all-trans-retinoic acid or the synthetic retinoid fenretinide. Fenretinide 168-179 insulin like growth factor binding protein 7 Homo sapiens 14-19 21607448-1 1994 Fenretinide (4-HPR), a synthetic amide derivative of retinoic acid, has proven effective in preventing chemically induced mammary carcinoma in rodents. Fenretinide 0-11 haptoglobin-related protein Homo sapiens 15-18 7961949-1 1994 The three-dimensional structures of complexes between bovine plasma retinol-binding protein (RBP) and three retinol analogs with different end groups (fenretinide, all-trans retinoic acid, and axerophthene) have been determined to 1.8-1.9-A resolution. Fenretinide 151-162 retinol binding protein 4 Bos taurus 93-96 8033144-1 1994 The efficacy of the retinoid N-(4-hydroxyphenyl)retinamide (4-HPR) has been demonstrated in the inhibition of cancers in a variety of tissues. Fenretinide 29-58 haptoglobin-related protein Homo sapiens 62-65 8149338-0 1994 Successful topical treatment of oral lichen planus and leukoplakias with fenretinide (4-HPR). Fenretinide 73-84 haptoglobin-related protein Homo sapiens 88-91 8208624-1 1994 The synthetic retinoid N-(4-hydroxyphenyl) retinamide (4-HPR) has been demonstrated to inhibit the development of primary and metastatic neoplasms in several animal models. Fenretinide 23-53 haptoglobin-related protein Homo sapiens 57-60 8180126-6 1994 RAR-beta gene expression is induced both by retinoic acid and by fenretinide in normal cells, but tumor cells fail to respond to either. Fenretinide 65-76 retinoic acid receptor beta Homo sapiens 0-8 8217599-1 1993 N-(4-hydroxyphenyl) retinamide (4-HPR) is a synthetic retinoid which reduces the incidence of experimental tumours in animals and has been chosen for its weak toxicity to be tested as a chemopreventive agent in humans. Fenretinide 0-30 haptoglobin-related protein Homo sapiens 34-37 7808027-6 1994 Autocrine stimulation of human breast cancer cell lines by tgf-alpha, insulin-like growth factors I and II is significantly abrogated by fenretinide. Fenretinide 137-148 transforming growth factor alpha Homo sapiens 59-68 7808027-6 1994 Autocrine stimulation of human breast cancer cell lines by tgf-alpha, insulin-like growth factors I and II is significantly abrogated by fenretinide. Fenretinide 137-148 insulin like growth factor 1 Homo sapiens 70-106 8221674-1 1993 Fenretinide or N-(4-hydroxyphenyl) retinamide (4HPR) is a synthetic retinoid currently being tested clinically, which can inhibit the development and the growth of breast and prostate cancers in rodents. Fenretinide 0-11 haptoglobin-related protein Homo sapiens 48-51 8221674-1 1993 Fenretinide or N-(4-hydroxyphenyl) retinamide (4HPR) is a synthetic retinoid currently being tested clinically, which can inhibit the development and the growth of breast and prostate cancers in rodents. Fenretinide 15-45 haptoglobin-related protein Homo sapiens 48-51 8319203-1 1993 Fenretinide (HPR), 13-cis-retinoic acid, and all-trans-retinoic acid are vitamin A derivatives used in the treatment of cancer and severe acne. Fenretinide 0-11 haptoglobin-related protein Homo sapiens 13-16 8402658-0 1993 The synthetic retinoid fenretinide lowers plasma insulin-like growth factor I levels in breast cancer patients. Fenretinide 23-34 insulin like growth factor 1 Homo sapiens 49-77 8402658-1 1993 We studied the effect of fenretinide [N-(4-hydroxyphenyl)retinamide (4-HPR)], a synthetic analogue of retinoic acid, on plasma insulin-like growth factor I (IGF-I) levels in a consecutive cohort of stage I breast cancer patients belonging to a randomized phase III trial of breast cancer chemoprevention. Fenretinide 25-36 insulin like growth factor 1 Homo sapiens 127-155 8402658-1 1993 We studied the effect of fenretinide [N-(4-hydroxyphenyl)retinamide (4-HPR)], a synthetic analogue of retinoic acid, on plasma insulin-like growth factor I (IGF-I) levels in a consecutive cohort of stage I breast cancer patients belonging to a randomized phase III trial of breast cancer chemoprevention. Fenretinide 25-36 insulin like growth factor 1 Homo sapiens 157-162 8402658-1 1993 We studied the effect of fenretinide [N-(4-hydroxyphenyl)retinamide (4-HPR)], a synthetic analogue of retinoic acid, on plasma insulin-like growth factor I (IGF-I) levels in a consecutive cohort of stage I breast cancer patients belonging to a randomized phase III trial of breast cancer chemoprevention. Fenretinide 38-67 insulin like growth factor 1 Homo sapiens 127-155 8360775-0 1993 N-(4-hydroxyphenyl)retinamide (fenretinide) induces retinol-binding protein secretion from liver and accumulation in the kidneys in rats. Fenretinide 0-29 retinol binding protein 4 Rattus norvegicus 52-75 8360775-0 1993 N-(4-hydroxyphenyl)retinamide (fenretinide) induces retinol-binding protein secretion from liver and accumulation in the kidneys in rats. Fenretinide 31-42 retinol binding protein 4 Rattus norvegicus 52-75 8360775-1 1993 The chemopreventive retinoid N-(4-hydroxyphenyl)retinamide (HPR) depresses serum retinol and retinol-binding protein (RBP) concentrations. Fenretinide 29-58 retinol binding protein 4 Rattus norvegicus 93-116 8360775-1 1993 The chemopreventive retinoid N-(4-hydroxyphenyl)retinamide (HPR) depresses serum retinol and retinol-binding protein (RBP) concentrations. Fenretinide 29-58 retinol binding protein 4 Rattus norvegicus 118-121 8360775-1 1993 The chemopreventive retinoid N-(4-hydroxyphenyl)retinamide (HPR) depresses serum retinol and retinol-binding protein (RBP) concentrations. Fenretinide 60-63 retinol binding protein 4 Rattus norvegicus 93-116 8360775-1 1993 The chemopreventive retinoid N-(4-hydroxyphenyl)retinamide (HPR) depresses serum retinol and retinol-binding protein (RBP) concentrations. Fenretinide 60-63 retinol binding protein 4 Rattus norvegicus 118-121 8297658-0 1993 Mammographic patterns in breast cancer chemoprevention with fenretinide (4-HPR). Fenretinide 60-71 haptoglobin-related protein Homo sapiens 75-78 7685988-1 1993 The in vitro anticlastogenic effects of a new synthetic retinoid derivative, N-(4-hydroxyphenyl)-retinamide (4-HPR), were studied in two human lymphoblastid cell lines (3640P and 4087P). Fenretinide 77-107 haptoglobin-related protein Homo sapiens 111-114 8007703-1 1993 Fenretinide (4-HPR), a synthetic derivative of retinoic acid, has proven effective at inhibiting in vitro breast cancer cell growth and preventing the progression of chemically induced mammary carcinoma in rodents. Fenretinide 0-11 haptoglobin-related protein Homo sapiens 15-18 8412202-0 1993 Fenretinide (4-HPR) in chemoprevention of oral leukoplakia. Fenretinide 0-11 haptoglobin-related protein Homo sapiens 15-18 8412202-2 1993 The goal of the trial is to evaluate the effectiveness of fenretinide (4-HPR) in preventing relapses, new localizations, and carcinomas in patients with benign postoperative diagnoses who have been surgically treated for oral leukoplakias. Fenretinide 58-69 haptoglobin-related protein Homo sapiens 73-76 8382073-3 1993 Attention is presently focused on two compounds, the synthetic retinoid fenretinide (4-HPR) and the antioestrogen tamoxifen, and their possible synergism. Fenretinide 72-83 haptoglobin-related protein Homo sapiens 87-90 1624219-4 1992 In similar experiments, two closely related retinamides, all-trans-(phenyl)retinamide (PR) and N-(4-hydroxyphenyl)retinamide (4-HPR), also potentiated murine splenocyte rIL-2 responses. Fenretinide 95-124 interleukin 2 Rattus norvegicus 169-174 1386578-3 1992 The remarkable early reduction in plasma retinol level induced by fenretinide administration may be associated with the high binding affinity of this retinoid to RBP and to its interference with the RBP-TTR complex formation. Fenretinide 66-77 retinol binding protein 4 Homo sapiens 162-165 1386578-3 1992 The remarkable early reduction in plasma retinol level induced by fenretinide administration may be associated with the high binding affinity of this retinoid to RBP and to its interference with the RBP-TTR complex formation. Fenretinide 66-77 retinol binding protein 4 Homo sapiens 199-202 1386578-3 1992 The remarkable early reduction in plasma retinol level induced by fenretinide administration may be associated with the high binding affinity of this retinoid to RBP and to its interference with the RBP-TTR complex formation. Fenretinide 66-77 transthyretin Homo sapiens 203-206 1306735-0 1992 Prevention of local relapses and new localisations of oral leukoplakias with the synthetic retinoid fenretinide (4-HPR). Fenretinide 100-111 haptoglobin-related protein Homo sapiens 115-118 1388202-0 1992 Secretion of vitamin A and retinol-binding protein into plasma is depressed in rats by N-(4-hydroxyphenyl)retinamide (fenretinide). Fenretinide 87-116 retinol binding protein 4 Rattus norvegicus 27-50 1388202-0 1992 Secretion of vitamin A and retinol-binding protein into plasma is depressed in rats by N-(4-hydroxyphenyl)retinamide (fenretinide). Fenretinide 118-129 retinol binding protein 4 Rattus norvegicus 27-50 1388202-1 1992 In clinical trials the cancer preventive drug N-(4-hydroxyphenyl)retinamide (HPR) markedly lowers plasma concentrations of retinol and retinol-binding protein (RBP). Fenretinide 46-75 retinol binding protein 4 Rattus norvegicus 135-158 1388202-1 1992 In clinical trials the cancer preventive drug N-(4-hydroxyphenyl)retinamide (HPR) markedly lowers plasma concentrations of retinol and retinol-binding protein (RBP). Fenretinide 46-75 retinol binding protein 4 Rattus norvegicus 160-163 1624219-4 1992 In similar experiments, two closely related retinamides, all-trans-(phenyl)retinamide (PR) and N-(4-hydroxyphenyl)retinamide (4-HPR), also potentiated murine splenocyte rIL-2 responses. Fenretinide 126-131 interleukin 2 Rattus norvegicus 169-174 1835622-0 1991 Long-term tolerability of fenretinide (4-HPR) in breast cancer patients. Fenretinide 26-37 haptoglobin-related protein Homo sapiens 41-44 1305675-4 1992 In vitro assays indicate that fenretinide (4-HPR), alpha-interferon, and 5-fluorouracil possess significant antitumoral activity in human transitional cell carcinoma (TCC) lines. Fenretinide 30-41 haptoglobin-related protein Homo sapiens 45-48 1305678-1 1992 The ability of the synthetic retinoid N-(4-hydroxyphenyl)retinamide (4-HPR) to affect the outcome of previously resected superficial bladder cancer was investigated in a pilot study using DNA content flow cytometry and conventional cytology as intermediate endpoints. Fenretinide 38-67 haptoglobin-related protein Homo sapiens 71-74 1535505-1 1992 Fenretinide [N-(4-hydroxyphenyl)retinamide, 4-HPR] is an effective agent for the inhibition of N-nitroso-N-methylurea-induced breast cancer in rats. Fenretinide 0-11 haptoglobin-related protein Homo sapiens 46-49 34742949-2 2022 However, fenretinide reduces plasma vitamin A levels by interacting with retinol-binding protein 4 (RBP4), which often results in reversible night blindness in patients. Fenretinide 9-20 retinol binding protein 4 Homo sapiens 73-98 33815111-6 2021 In contrast, EtOH + Fen-treated mice were resistant to the effects of EtOH on promoting intestinal permeability and had higher intestinal protein levels of claudin one and occludin. Fenretinide 20-23 occludin Mus musculus 172-180 33815111-7 2021 Also, EtOH + Fen-treated mice had significantly lower plasma levels of endotoxin, and reductions in expression of TNF-alpha and TLR4 positive macrophages, Kupffer cells, and hepatocytes in the intestine and liver. Fenretinide 13-16 tumor necrosis factor Mus musculus 114-123 33815111-7 2021 Also, EtOH + Fen-treated mice had significantly lower plasma levels of endotoxin, and reductions in expression of TNF-alpha and TLR4 positive macrophages, Kupffer cells, and hepatocytes in the intestine and liver. Fenretinide 13-16 toll-like receptor 4 Mus musculus 128-132 34742949-2 2022 However, fenretinide reduces plasma vitamin A levels by interacting with retinol-binding protein 4 (RBP4), which often results in reversible night blindness in patients. Fenretinide 9-20 retinol binding protein 4 Homo sapiens 100-104 34742949-3 2022 Cell culture and in vitro studies show that fenretinide binds and inhibits the activity of beta-carotene oxygenase 1 (BCO1), the enzyme responsible for endogenous vitamin A formation. Fenretinide 44-55 beta-carotene oxygenase 1 Mus musculus 91-116 34742949-3 2022 Cell culture and in vitro studies show that fenretinide binds and inhibits the activity of beta-carotene oxygenase 1 (BCO1), the enzyme responsible for endogenous vitamin A formation. Fenretinide 44-55 beta-carotene oxygenase 1 Homo sapiens 118-122 34742949-5 2022 The goal of this study was to determine if the inhibition of BCO1 by fenretinide affects vitamin A formation in mice fed beta-carotene. Fenretinide 69-80 beta-carotene oxygenase 1 Mus musculus 61-65 2527636-0 1989 Suppression of rat mammary cancer development by N-(4-hydroxyphenyl)retinamide (4-HPR) following surgical removal of first palpable tumor. Fenretinide 49-78 haptoglobin-related protein Homo sapiens 82-85 34106748-0 2021 N-(4-Hydroxyphenyl) retinamide suppresses SARS-CoV-2 spike protein-mediated cell-cell fusion by a dihydroceramide Delta4-desaturase 1-independent mechanism. Fenretinide 0-30 surface glycoprotein Severe acute respiratory syndrome coronavirus 2 53-58 34106748-0 2021 N-(4-Hydroxyphenyl) retinamide suppresses SARS-CoV-2 spike protein-mediated cell-cell fusion by a dihydroceramide Delta4-desaturase 1-independent mechanism. Fenretinide 0-30 delta like canonical Notch ligand 4 Homo sapiens 114-133 34106748-3 2021 N-(4-hydroxyphenyl) retinamide (4-HPR), an inhibitor of dihydroceramide Delta4-desaturase 1 (DES1), suppressed cell-cell fusion, and viral infection. Fenretinide 0-30 delta like canonical Notch ligand 4 Homo sapiens 72-91 34106748-3 2021 N-(4-hydroxyphenyl) retinamide (4-HPR), an inhibitor of dihydroceramide Delta4-desaturase 1 (DES1), suppressed cell-cell fusion, and viral infection. Fenretinide 0-30 delta 4-desaturase, sphingolipid 1 Homo sapiens 93-97 34106748-3 2021 N-(4-hydroxyphenyl) retinamide (4-HPR), an inhibitor of dihydroceramide Delta4-desaturase 1 (DES1), suppressed cell-cell fusion, and viral infection. Fenretinide 32-37 delta like canonical Notch ligand 4 Homo sapiens 72-91 34106748-3 2021 N-(4-hydroxyphenyl) retinamide (4-HPR), an inhibitor of dihydroceramide Delta4-desaturase 1 (DES1), suppressed cell-cell fusion, and viral infection. Fenretinide 32-37 delta 4-desaturase, sphingolipid 1 Homo sapiens 93-97 33380244-4 2021 Intraperitoneally administered 4-HPR particularly at dose of 100 mg/kg obviously alleviated UC symptoms and restrained the mRNA expression of colonic IL-1beta, IL-6, and TNF-alpha in dextran sulfate sodium (DSS)-induced mice. Fenretinide 31-36 interleukin 1 alpha Mus musculus 150-158 33380244-4 2021 Intraperitoneally administered 4-HPR particularly at dose of 100 mg/kg obviously alleviated UC symptoms and restrained the mRNA expression of colonic IL-1beta, IL-6, and TNF-alpha in dextran sulfate sodium (DSS)-induced mice. Fenretinide 31-36 interleukin 6 Mus musculus 160-164 33380244-4 2021 Intraperitoneally administered 4-HPR particularly at dose of 100 mg/kg obviously alleviated UC symptoms and restrained the mRNA expression of colonic IL-1beta, IL-6, and TNF-alpha in dextran sulfate sodium (DSS)-induced mice. Fenretinide 31-36 tumor necrosis factor Mus musculus 170-179 34363869-4 2021 We analyzed the effects of FEN on mutant SOD1 (mSOD1) toxicity in motoneuronal (NSC34) and a muscle (C2C12) cell lines and evaluated the impacts of chronic administration of a new nanomicellar fenretinide formulation (NanoMFen) on ALS disease progression in the SOD1G93A mouse model. Fenretinide 27-30 superoxide dismutase 1, soluble Mus musculus 47-52 34363869-5 2021 The results showed that FEN significantly prevents the toxicity of mSOD1 expression in NSC34 motor neuron; furthermore, FEN is able to partially overcome the toxic effect of mSOD1 on the myogenic program of C2C12 muscle cells. Fenretinide 24-27 superoxide dismutase 1, soluble Mus musculus 67-72 34363869-5 2021 The results showed that FEN significantly prevents the toxicity of mSOD1 expression in NSC34 motor neuron; furthermore, FEN is able to partially overcome the toxic effect of mSOD1 on the myogenic program of C2C12 muscle cells. Fenretinide 120-123 superoxide dismutase 1, soluble Mus musculus 67-72 34363869-5 2021 The results showed that FEN significantly prevents the toxicity of mSOD1 expression in NSC34 motor neuron; furthermore, FEN is able to partially overcome the toxic effect of mSOD1 on the myogenic program of C2C12 muscle cells. Fenretinide 120-123 superoxide dismutase 1, soluble Mus musculus 174-179 34106748-12 2021 4-HPR (also known as fenretinide) is an inhibitor of DES1 and it exhibits antitumor activity and suppresses cell-cell fusion and viral infection. Fenretinide 0-5 delta 4-desaturase, sphingolipid 1 Homo sapiens 53-57 34106748-12 2021 4-HPR (also known as fenretinide) is an inhibitor of DES1 and it exhibits antitumor activity and suppresses cell-cell fusion and viral infection. Fenretinide 21-32 delta 4-desaturase, sphingolipid 1 Homo sapiens 53-57 2527636-1 1989 A study was conducted to determine whether N-(4-hydroxyphenyl)retinamide (4-HPR) affects the development of new mammary tumors subsequent to the surgical removal of the first palpable tumor. Fenretinide 43-72 haptoglobin-related protein Homo sapiens 76-79 2694156-3 1989 N-(4-hydroxyphenyl)retinamide (4-HPR) currently appears to be the most efficacious retinoid against carcinogen-induced breast, urinary bladder, and lung cancer in rodents. Fenretinide 0-29 haptoglobin-related protein Homo sapiens 33-36 3621188-1 1987 Retinyl acetate, 13-cis-retinoic acid (13cisRA), and N-(4-hydroxyphenyl)-retinamide (4HPR) were assayed for their in vivo effects on hepatic levels of cytochrome P450, cytosolic glutathione-S-transferase, and quinone reductase. Fenretinide 53-83 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 151-203 2643696-3 1989 N-(4-hydroxyphenyl) retinamide (4-HPR) currently appears to be the most efficaceous retinoid against carcinogen-induced breast, urinary bladder, and lung cancer in rodents. Fenretinide 0-30 haptoglobin-related protein Homo sapiens 34-37 33423090-0 2021 A phase I study of intravenous fenretinide (4-HPR) for patients with malignant solid tumors. Fenretinide 31-42 haptoglobin-related protein Homo sapiens 46-49 33123895-11 2020 4-HPR-induced reactive oxygen species (ROS) production and NF-kappaB inhibition might be the reason of 4-HPR selectivity on FLT3 mutated AML cells. Fenretinide 103-108 fms related receptor tyrosine kinase 3 Homo sapiens 124-128 33439253-6 2021 Agents that target the viral component of the IMPalpha/beta1-virus interface include N-(4-hydroxyphenyl) retinamide (4-HPR), which specifically targets DENV/ZIKV/WNV non-structural protein 5 (NS5). Fenretinide 85-115 inositol monophosphatase 1 Homo sapiens 46-60 33439253-6 2021 Agents that target the viral component of the IMPalpha/beta1-virus interface include N-(4-hydroxyphenyl) retinamide (4-HPR), which specifically targets DENV/ZIKV/WNV non-structural protein 5 (NS5). Fenretinide 117-122 inositol monophosphatase 1 Homo sapiens 46-60 33079733-2 2021 The histone deacetylase (HDAC) inhibitors and the synthetic cytotoxic retinoid fenretinide have achieved durable clinical responses in T-cell lymphomas as single agents, and patients who failed prior HDAC inhibitor treatment have responded to fenretinide. Fenretinide 79-90 histone deacetylase 9 Homo sapiens 200-204 33079733-2 2021 The histone deacetylase (HDAC) inhibitors and the synthetic cytotoxic retinoid fenretinide have achieved durable clinical responses in T-cell lymphomas as single agents, and patients who failed prior HDAC inhibitor treatment have responded to fenretinide. Fenretinide 243-254 histone deacetylase 9 Homo sapiens 4-23 33079733-2 2021 The histone deacetylase (HDAC) inhibitors and the synthetic cytotoxic retinoid fenretinide have achieved durable clinical responses in T-cell lymphomas as single agents, and patients who failed prior HDAC inhibitor treatment have responded to fenretinide. Fenretinide 243-254 histone deacetylase 9 Homo sapiens 25-29 31927141-10 2020 Several retinoids, notably anhydroretinol and fenretinide, capable of displacing retinol from binding sites on PKCdelta, can co-activate PKCdelta signaling but, owing to their extended system of conjugated double bonds, are unable to silence PKCdelta in a timely manner. Fenretinide 46-57 protein kinase C delta Homo sapiens 111-119 31927141-10 2020 Several retinoids, notably anhydroretinol and fenretinide, capable of displacing retinol from binding sites on PKCdelta, can co-activate PKCdelta signaling but, owing to their extended system of conjugated double bonds, are unable to silence PKCdelta in a timely manner. Fenretinide 46-57 protein kinase C delta Homo sapiens 137-145 31927141-10 2020 Several retinoids, notably anhydroretinol and fenretinide, capable of displacing retinol from binding sites on PKCdelta, can co-activate PKCdelta signaling but, owing to their extended system of conjugated double bonds, are unable to silence PKCdelta in a timely manner. Fenretinide 46-57 protein kinase C delta Homo sapiens 137-145 33123895-0 2020 4-Hydroxyphenyl Retinamide Preferentially Targets FLT3 Mutated Acute Myeloid Leukemia via ROS Induction and NF-kappaB Inhibition. Fenretinide 0-26 fms related receptor tyrosine kinase 3 Homo sapiens 50-54 33123895-7 2020 CD34+ AML stem/progenitor cells separated from 32 AML samples were treated with 4-HPR. Fenretinide 80-85 CD34 molecule Homo sapiens 0-4 33123895-8 2020 Correlation analysis showed that AML cells with FLT3-ITD genetic alteration were more sensitive to 4-HPR treatment than those without FLT3-ITD. Fenretinide 99-104 fms related receptor tyrosine kinase 3 Homo sapiens 48-52 33123895-8 2020 Correlation analysis showed that AML cells with FLT3-ITD genetic alteration were more sensitive to 4-HPR treatment than those without FLT3-ITD. Fenretinide 99-104 fms related receptor tyrosine kinase 3 Homo sapiens 134-138 32982239-0 2020 Nanomicellar Lenalidomide-Fenretinide Combination Suppresses Tumor Growth in an MYCN Amplified Neuroblastoma Tumor. Fenretinide 26-37 MYCN proto-oncogene, bHLH transcription factor Homo sapiens 80-84 33123895-11 2020 4-HPR-induced reactive oxygen species (ROS) production and NF-kappaB inhibition might be the reason of 4-HPR selectivity on FLT3 mutated AML cells. Fenretinide 0-5 fms related receptor tyrosine kinase 3 Homo sapiens 124-128 32366203-1 2020 Fenretinide (4-HPR), a synthetic retinoid, has shown its antitumor activity in many tumor types with low cytotoxicity to normal cells and high clinical safety. Fenretinide 0-11 haptoglobin-related protein Homo sapiens 15-18 32982239-1 2020 Purpose: In a previous study, we demonstrated that the combination of fenretinide with lenalidomide, administered by a novel nanomicellar formulation (FLM), provided a strong antitumor effect in a neuroblastoma TrkB-expressing tumor. Fenretinide 70-81 neurotrophic receptor tyrosine kinase 2 Homo sapiens 211-215 32266855-1 2020 Fenretinide (4-HPR), as a semi-synthetic retinoid, has apoptosis-promoting effects as a single agent and chemotherapy synergist in vitro. Fenretinide 0-11 haptoglobin-related protein Homo sapiens 15-18 32266855-3 2020 In this research, a new TPGS-Soluplus mixed micelles were developed which encapsulation efficiencies of paclitaxel (PTX) and fenretinide (4-HPR) were as high as 98%, and the average diameter of the micelles was 66.26 nm. Fenretinide 126-137 haptoglobin-related protein Homo sapiens 141-144 32696214-0 2020 Chemosensitization by 4-hydroxyphenyl retinamide-induced NF-kappaB inhibition in acute myeloid leukemia cells. Fenretinide 22-48 nuclear factor kappa B subunit 1 Homo sapiens 57-66 32862802-0 2022 High-fat diet exacerbates cognitive and metabolic abnormalities in neuronal BACE1 knock-in mice - partial prevention by Fenretinide. Fenretinide 120-131 beta-site APP cleaving enzyme 1 Mus musculus 76-81 32497314-1 2020 Fenretinide (4-HPR) is a synthetic derivative of All-Trans-Retinoic Acid (ATRA) characterised by improved therapeutic properties and toxicological profile relative to ATRA. Fenretinide 0-11 haptoglobin-related protein Homo sapiens 15-18 32306138-2 2020 Our study aimed to investigate the age-dependent deterioration in lung function and the effects of treatment with Fenretinide formulation (LAU-7b) in Cftr knockout (KO) mice. Fenretinide 114-125 cystic fibrosis transmembrane conductance regulator Mus musculus 150-154 32144381-2 2020 Previously, we have identified fenretinide (retinoic acid p-hydroxyanilide) to affect PAX3-FOXO1 expression levels as well as FP-RMS cell viability. Fenretinide 31-42 paired box 3 Homo sapiens 86-90 32144381-2 2020 Previously, we have identified fenretinide (retinoic acid p-hydroxyanilide) to affect PAX3-FOXO1 expression levels as well as FP-RMS cell viability. Fenretinide 31-42 forkhead box O1 Homo sapiens 91-96 32144381-2 2020 Previously, we have identified fenretinide (retinoic acid p-hydroxyanilide) to affect PAX3-FOXO1 expression levels as well as FP-RMS cell viability. Fenretinide 44-74 paired box 3 Homo sapiens 86-90 32144381-2 2020 Previously, we have identified fenretinide (retinoic acid p-hydroxyanilide) to affect PAX3-FOXO1 expression levels as well as FP-RMS cell viability. Fenretinide 44-74 forkhead box O1 Homo sapiens 91-96 32273303-0 2020 Treatment of allergic asthma with Fenretinide formulation (LAU-7b) downregulates Ormdl3 expression and normalizes ceramides imbalance. Fenretinide 34-45 ORM1-like 3 (S. cerevisiae) Mus musculus 81-87 32273303-4 2020 We hypothesized that Fenretinide (FEN) can prevent the allergic asthma-induced augmentation of Ormdl3 gene expression, normalize aberrant levels of VLCCs and LCCs, and treat allergic asthma symptoms. Fenretinide 21-32 ORM1-like 3 (S. cerevisiae) Mus musculus 95-101 32273303-4 2020 We hypothesized that Fenretinide (FEN) can prevent the allergic asthma-induced augmentation of Ormdl3 gene expression, normalize aberrant levels of VLCCs and LCCs, and treat allergic asthma symptoms. Fenretinide 34-37 ORM1-like 3 (S. cerevisiae) Mus musculus 95-101 32238432-6 2020 The retinol-isomerase activities of Rpe65 and Des1 are inhibited by emixustat and fenretinide, respectively. Fenretinide 82-93 delta(4)-desaturase, sphingolipid 1 Danio rerio 46-50 32273303-12 2020 - The novel oral clinical formulation of Fenretinide (LAU-7b) effectively lowers the AHR, protects against inflammatory cell infiltration and mucus accumulation induced by HDM in both Zpbp2 KO and WT A/J mice. Fenretinide 41-52 zona pellucida binding protein 2 Mus musculus 184-189 31925643-0 2020 Fenretinide reduces angiogenesis by downregulating CDH5, FOXM1 and eNOS genes and suppressing microRNA-10b. Fenretinide 0-11 cadherin 5 Homo sapiens 51-55 32106372-0 2020 Fenretinide attenuates lipopolysaccharide (LPS)-induced blood-brain barrier (BBB) and depressive-like behavior in mice by targeting Nrf-2 signaling. Fenretinide 0-11 nuclear factor, erythroid derived 2, like 2 Mus musculus 132-137 32106372-5 2020 Results here indicated that Fen treatment markedly improved Nrf2 expression and nuclear translocation in mouse brain endothelial cell line bEnd.3 cells, and promoted Nrf2-antioxidant responsive element (ARE) transcription activity, as well as its down-streaming signals, which was Nrf2-dependent. Fenretinide 28-31 nuclear factor, erythroid derived 2, like 2 Mus musculus 60-64 32106372-5 2020 Results here indicated that Fen treatment markedly improved Nrf2 expression and nuclear translocation in mouse brain endothelial cell line bEnd.3 cells, and promoted Nrf2-antioxidant responsive element (ARE) transcription activity, as well as its down-streaming signals, which was Nrf2-dependent. Fenretinide 28-31 nuclear factor, erythroid derived 2, like 2 Mus musculus 166-170 32106372-5 2020 Results here indicated that Fen treatment markedly improved Nrf2 expression and nuclear translocation in mouse brain endothelial cell line bEnd.3 cells, and promoted Nrf2-antioxidant responsive element (ARE) transcription activity, as well as its down-streaming signals, which was Nrf2-dependent. Fenretinide 28-31 nuclear factor, erythroid derived 2, like 2 Mus musculus 166-170 31925643-0 2020 Fenretinide reduces angiogenesis by downregulating CDH5, FOXM1 and eNOS genes and suppressing microRNA-10b. Fenretinide 0-11 forkhead box M1 Homo sapiens 57-62 31925643-0 2020 Fenretinide reduces angiogenesis by downregulating CDH5, FOXM1 and eNOS genes and suppressing microRNA-10b. Fenretinide 0-11 nitric oxide synthase 3 Homo sapiens 67-71 31925643-0 2020 Fenretinide reduces angiogenesis by downregulating CDH5, FOXM1 and eNOS genes and suppressing microRNA-10b. Fenretinide 0-11 microRNA 10b Homo sapiens 94-106 31925643-6 2020 In this study, we aimed to investigate the effects of the fenretinide on some miRNAs involving in angiogenesis process and on the expression of CDH5, FOXM1 and eNOS genes upregulated in angiogenesis. Fenretinide 58-69 forkhead box M1 Homo sapiens 150-155 31925643-6 2020 In this study, we aimed to investigate the effects of the fenretinide on some miRNAs involving in angiogenesis process and on the expression of CDH5, FOXM1 and eNOS genes upregulated in angiogenesis. Fenretinide 58-69 nitric oxide synthase 3 Homo sapiens 160-164 31925643-13 2020 Our results have shown that fenretinide exhibited anti-angiogenic activity through the down-regulation of CDH5, FOXM1 and eNOS genes, and suppression of miR-10b. Fenretinide 28-39 cadherin 5 Homo sapiens 106-110 31925643-13 2020 Our results have shown that fenretinide exhibited anti-angiogenic activity through the down-regulation of CDH5, FOXM1 and eNOS genes, and suppression of miR-10b. Fenretinide 28-39 forkhead box M1 Homo sapiens 112-117 31925643-13 2020 Our results have shown that fenretinide exhibited anti-angiogenic activity through the down-regulation of CDH5, FOXM1 and eNOS genes, and suppression of miR-10b. Fenretinide 28-39 nitric oxide synthase 3 Homo sapiens 122-126 31925643-13 2020 Our results have shown that fenretinide exhibited anti-angiogenic activity through the down-regulation of CDH5, FOXM1 and eNOS genes, and suppression of miR-10b. Fenretinide 28-39 microRNA 10b Homo sapiens 153-160 31484706-10 2019 Thus, fenretinide + venetoclax is a synergistic combination that warrants clinical testing in high BCL-2-expressing neuroblastoma. Fenretinide 6-17 BCL2 apoptosis regulator Homo sapiens 99-104 31988387-3 2020 The aim here was to examine the possibility of cross-talk between AnxA8 and Wnt signalling, as both are down-regulated upon fenretinide (FR)-mediated RPE transdifferentiation. Fenretinide 124-135 annexin A8 Homo sapiens 66-71 31988387-3 2020 The aim here was to examine the possibility of cross-talk between AnxA8 and Wnt signalling, as both are down-regulated upon fenretinide (FR)-mediated RPE transdifferentiation. Fenretinide 137-139 annexin A8 Homo sapiens 66-71 31621898-0 2020 Fenretinide treatment accelerates atherosclerosis development in apoE-deficient mice in spite of beneficial metabolic effects. Fenretinide 0-11 apolipoprotein E Mus musculus 65-69 31515297-11 2019 Notably, combination fenretinide-tocilizumab-reparixin treatment significantly suppressed IL-6 and IL-8 release, stem cell gene expression, and invasion in these diverse CSCE populations. Fenretinide 21-32 interleukin 6 Homo sapiens 90-94 31515297-11 2019 Notably, combination fenretinide-tocilizumab-reparixin treatment significantly suppressed IL-6 and IL-8 release, stem cell gene expression, and invasion in these diverse CSCE populations. Fenretinide 21-32 C-X-C motif chemokine ligand 8 Homo sapiens 99-103 31678518-0 2020 Fenretinide favorably affects mucins (MUC5AC/MUC5B) and fatty acid imbalance in a manner mimicking CFTR-induced correction. Fenretinide 0-11 mucin 5AC, oligomeric mucus/gel-forming Homo sapiens 38-44 31678518-0 2020 Fenretinide favorably affects mucins (MUC5AC/MUC5B) and fatty acid imbalance in a manner mimicking CFTR-induced correction. Fenretinide 0-11 mucin 5B, oligomeric mucus/gel-forming Homo sapiens 45-50 31678518-0 2020 Fenretinide favorably affects mucins (MUC5AC/MUC5B) and fatty acid imbalance in a manner mimicking CFTR-induced correction. Fenretinide 0-11 CF transmembrane conductance regulator Homo sapiens 99-103 31678518-6 2020 Fenretinide prevented the lipopolysaccharide-induced increase of MUC5AC gene expression, without affecting the level of MUC5B, in a lung goblet cell line. Fenretinide 0-11 mucin 5AC, oligomeric mucus/gel-forming Homo sapiens 65-71 31880746-12 2020 The ALT and AST in 4-HPR group were significantly decreased compared with ACLF group. Fenretinide 19-24 glutamic-oxaloacetic transaminase 2 Rattus norvegicus 12-15 31484706-0 2019 Fenretinide via NOXA Induction, Enhanced Activity of the BCL-2 Inhibitor Venetoclax in High BCL-2-Expressing Neuroblastoma Preclinical Models. Fenretinide 0-11 phorbol-12-myristate-13-acetate-induced protein 1 Homo sapiens 16-20 31484706-0 2019 Fenretinide via NOXA Induction, Enhanced Activity of the BCL-2 Inhibitor Venetoclax in High BCL-2-Expressing Neuroblastoma Preclinical Models. Fenretinide 0-11 BCL2 apoptosis regulator Homo sapiens 57-62 31484706-2 2019 Fenretinide (4-HPR) is a cytotoxic retinoid with clinical activity in recurrent neuroblastoma and venetoclax (ABT-199) is a selective inhibitor of the antiapoptotic protein B-cell lymphoma-2 (BCL-2). Fenretinide 0-11 haptoglobin-related protein Homo sapiens 15-18 31484706-2 2019 Fenretinide (4-HPR) is a cytotoxic retinoid with clinical activity in recurrent neuroblastoma and venetoclax (ABT-199) is a selective inhibitor of the antiapoptotic protein B-cell lymphoma-2 (BCL-2). Fenretinide 0-11 BCL2 apoptosis regulator Homo sapiens 173-190 31484706-2 2019 Fenretinide (4-HPR) is a cytotoxic retinoid with clinical activity in recurrent neuroblastoma and venetoclax (ABT-199) is a selective inhibitor of the antiapoptotic protein B-cell lymphoma-2 (BCL-2). Fenretinide 0-11 BCL2 apoptosis regulator Homo sapiens 192-197 31839811-0 2019 Fenretinide-induced Apoptosis of Acute Myeloid Leukemia Cells via NR4A1 Translocation into Mitochondria and Bcl-2 Transformation. Fenretinide 0-11 nuclear receptor subfamily 4 group A member 1 Homo sapiens 66-71 31631013-4 2019 Inhibition of DEGS1 in hematopoietic stem and progenitor cells during the transition from quiescence to cellular activation with N-(4-hydroxyphenyl) retinamide activates coordinated stress pathways that coalesce on endoplasmic reticulum stress and autophagy programs to maintain immunophenotypic and functional HSCs. Fenretinide 129-159 delta 4-desaturase, sphingolipid 1 Homo sapiens 14-19 31839811-0 2019 Fenretinide-induced Apoptosis of Acute Myeloid Leukemia Cells via NR4A1 Translocation into Mitochondria and Bcl-2 Transformation. Fenretinide 0-11 BCL2 apoptosis regulator Homo sapiens 108-113 31839811-1 2019 OBJECTIVE: Fenretinide is reported to induce NR4A1-associated apoptosis in several types of cancer cells. Fenretinide 11-22 nuclear receptor subfamily 4 group A member 1 Homo sapiens 45-50 31839811-7 2019 In addition, the expression levels of NR4A1 in the nuclei and mitochondria of fenretinide-treated AML cells were also measured. Fenretinide 78-89 nuclear receptor subfamily 4 group A member 1 Homo sapiens 38-43 31839811-11 2019 Fenretinide induced the expression of NR4A1 and mitochondria-mediated apoptotic pathway-associated proteins in a time- and concentration-dependent manner. Fenretinide 0-11 nuclear receptor subfamily 4 group A member 1 Homo sapiens 38-43 31839811-13 2019 Under the action of fenretinide, the NR4A1 protein expression was down-regulated in nuclear extracts whereas up-regulated in mitochondrial extracts. Fenretinide 20-31 nuclear receptor subfamily 4 group A member 1 Homo sapiens 37-42 31839811-14 2019 At the same time, fenretinide promoted NR4A1 translocation from nuclei into mitochondria, and enhanced the interaction between NR4A1 and Bcl-2, thereby exposing the BH3 domain of Bcl-2 to exert the anti-apoptotic effect. Fenretinide 18-29 nuclear receptor subfamily 4 group A member 1 Homo sapiens 39-44 31839811-14 2019 At the same time, fenretinide promoted NR4A1 translocation from nuclei into mitochondria, and enhanced the interaction between NR4A1 and Bcl-2, thereby exposing the BH3 domain of Bcl-2 to exert the anti-apoptotic effect. Fenretinide 18-29 nuclear receptor subfamily 4 group A member 1 Homo sapiens 127-132 31839811-14 2019 At the same time, fenretinide promoted NR4A1 translocation from nuclei into mitochondria, and enhanced the interaction between NR4A1 and Bcl-2, thereby exposing the BH3 domain of Bcl-2 to exert the anti-apoptotic effect. Fenretinide 18-29 BCL2 apoptosis regulator Homo sapiens 137-142 31839811-14 2019 At the same time, fenretinide promoted NR4A1 translocation from nuclei into mitochondria, and enhanced the interaction between NR4A1 and Bcl-2, thereby exposing the BH3 domain of Bcl-2 to exert the anti-apoptotic effect. Fenretinide 18-29 BCL2 apoptosis regulator Homo sapiens 179-184 31839811-15 2019 Moreover, fenretinide also exhibited an anti-leukemic effect and induced NR4A1 expression in the AML mouse model. Fenretinide 10-21 nuclear receptor subfamily 4, group A, member 1 Mus musculus 73-78 31839811-17 2019 Besides, the NR4A1-mediated signaling pathway is highly involved in the fenretinide-induced apoptosis of AML cells. Fenretinide 72-83 nuclear receptor subfamily 4 group A member 1 Homo sapiens 13-18 31276572-0 2019 Combination of fenretinide and ABT-263 induces apoptosis through NOXA for head and neck squamous cell carcinoma treatment. Fenretinide 15-26 phorbol-12-myristate-13-acetate-induced protein 1 Homo sapiens 65-69 30939964-5 2019 By screening a library of nuclear-receptor-agonists for modifiers of the SQSTM1 aggregates, we identified 4-hydroxy(phenyl)retinamide (4HPR) as a potent modifier exerting detrimental effects on mutant-TBK1 motoneurons fitness exacerbating the autophagy overload. Fenretinide 106-133 sequestosome 1 Homo sapiens 73-79 30939964-5 2019 By screening a library of nuclear-receptor-agonists for modifiers of the SQSTM1 aggregates, we identified 4-hydroxy(phenyl)retinamide (4HPR) as a potent modifier exerting detrimental effects on mutant-TBK1 motoneurons fitness exacerbating the autophagy overload. Fenretinide 106-133 TANK binding kinase 1 Homo sapiens 201-205 30939964-5 2019 By screening a library of nuclear-receptor-agonists for modifiers of the SQSTM1 aggregates, we identified 4-hydroxy(phenyl)retinamide (4HPR) as a potent modifier exerting detrimental effects on mutant-TBK1 motoneurons fitness exacerbating the autophagy overload. Fenretinide 135-139 sequestosome 1 Homo sapiens 73-79 30939964-5 2019 By screening a library of nuclear-receptor-agonists for modifiers of the SQSTM1 aggregates, we identified 4-hydroxy(phenyl)retinamide (4HPR) as a potent modifier exerting detrimental effects on mutant-TBK1 motoneurons fitness exacerbating the autophagy overload. Fenretinide 135-139 TANK binding kinase 1 Homo sapiens 201-205 30939964-7 2019 4HPR-increased toxicity was associated with the upregulation of SQSTM1 in a context of strongly reduced ATG10, while rescue of ATG10 levels abolished 4HPR toxicity. Fenretinide 0-4 sequestosome 1 Homo sapiens 64-70 30939964-7 2019 4HPR-increased toxicity was associated with the upregulation of SQSTM1 in a context of strongly reduced ATG10, while rescue of ATG10 levels abolished 4HPR toxicity. Fenretinide 0-4 autophagy related 10 Homo sapiens 104-109 30939964-7 2019 4HPR-increased toxicity was associated with the upregulation of SQSTM1 in a context of strongly reduced ATG10, while rescue of ATG10 levels abolished 4HPR toxicity. Fenretinide 0-4 autophagy related 10 Homo sapiens 127-132 30939964-7 2019 4HPR-increased toxicity was associated with the upregulation of SQSTM1 in a context of strongly reduced ATG10, while rescue of ATG10 levels abolished 4HPR toxicity. Fenretinide 150-154 autophagy related 10 Homo sapiens 127-132 31103416-0 2019 Effects of Liraglutide and Fenretinide treatments on the diabetic phenotype of neuronal human BACE1 knock-in mice. Fenretinide 27-38 beta-secretase 1 Homo sapiens 94-99 31103416-5 2019 Liraglutide and Fenretinide treatments inhibited adiposity gain and decreased circulating serum triglyceride (with Liraglutide) and leptin (with Fenretinide) levels in PLB4 mice. Fenretinide 16-27 leptin Mus musculus 132-138 31103416-5 2019 Liraglutide and Fenretinide treatments inhibited adiposity gain and decreased circulating serum triglyceride (with Liraglutide) and leptin (with Fenretinide) levels in PLB4 mice. Fenretinide 145-156 leptin Mus musculus 132-138 31276572-7 2019 Furthermore, a retinoid derivative and an endoplasmic reticulum stress inducer, fenretinide, induced NOXA, and combination of fenretinide and ABT-263 strongly induced apoptosis in HNSCC cells regardless of the HPV or p53 statuses. Fenretinide 80-91 phorbol-12-myristate-13-acetate-induced protein 1 Homo sapiens 101-105 31276572-7 2019 Furthermore, a retinoid derivative and an endoplasmic reticulum stress inducer, fenretinide, induced NOXA, and combination of fenretinide and ABT-263 strongly induced apoptosis in HNSCC cells regardless of the HPV or p53 statuses. Fenretinide 80-91 tumor protein p53 Homo sapiens 217-220 31276572-7 2019 Furthermore, a retinoid derivative and an endoplasmic reticulum stress inducer, fenretinide, induced NOXA, and combination of fenretinide and ABT-263 strongly induced apoptosis in HNSCC cells regardless of the HPV or p53 statuses. Fenretinide 126-137 tumor protein p53 Homo sapiens 217-220 30061204-1 2018 BACKGROUND/AIM: N-(4-hydroxyphenyl)retinamide (4-HPR) is a synthetic retinoid, less toxic than the parent all-trans retinoic acid (RA). Fenretinide 16-45 haptoglobin-related protein Homo sapiens 49-52 30836314-9 2019 The validated method was successfully applied to the analyses of pharmacokinetic samples from patients treated with safingol and all-trans-N-(4-hydroxyphenyl)retinamide; (fenretinide, 4-HPR) in a current phase I clinical trial (SPOC-2010-002, ClinicalTrials.gov Identifier: NCT01553071). Fenretinide 129-168 haptoglobin-related protein Homo sapiens 186-189 30251395-2 2018 The synthetic retinoid fenretinide (4-HPR) achieved multiple complete responses in relapse/refractory neuroblastoma in early-phase clinical trials, has low systemic toxicity, and has been considered for maintenance therapy clinical trials. Fenretinide 23-34 haptoglobin-related protein Homo sapiens 38-41 30129371-7 2018 Fenretinide and emixustat are VCMs for dry AMD and STGD1 that failed to halt geographic atrophy progression or improve vision in trials for AMD. Fenretinide 0-11 ATP binding cassette subfamily A member 4 Homo sapiens 51-56 29767236-0 2018 Fenretinide inhibits the proliferation and migration of human liver cancer HepG2 cells by downregulating the activation of myosin light chain kinase through the p38-MAPK signaling pathway. Fenretinide 0-11 myosin light chain kinase Homo sapiens 123-148 30086303-7 2018 Importantly, ATF6 containing a luminal achromatopsia eye disease mutation, unresponsive to proteotoxic stress, can be activated by fenretinide, a drug that upregulates DHC, suggesting a potential therapy for this and other ATF6-related diseases including heart disease and stroke. Fenretinide 131-142 activating transcription factor 6 Homo sapiens 13-17 30086303-7 2018 Importantly, ATF6 containing a luminal achromatopsia eye disease mutation, unresponsive to proteotoxic stress, can be activated by fenretinide, a drug that upregulates DHC, suggesting a potential therapy for this and other ATF6-related diseases including heart disease and stroke. Fenretinide 131-142 activating transcription factor 6 Homo sapiens 223-227 29767236-0 2018 Fenretinide inhibits the proliferation and migration of human liver cancer HepG2 cells by downregulating the activation of myosin light chain kinase through the p38-MAPK signaling pathway. Fenretinide 0-11 mitogen-activated protein kinase 14 Homo sapiens 161-164 29767236-1 2018 N-(4-hydroxyphenyl)retinamide (4-HPR or fenretinide), which is a synthetic analog of all-trans retinoic acid (ATRA), effectively inhibits the growth of several types of tumor cells; however, its molecular mechanism remains unclear. Fenretinide 0-29 haptoglobin-related protein Homo sapiens 33-36 29767236-1 2018 N-(4-hydroxyphenyl)retinamide (4-HPR or fenretinide), which is a synthetic analog of all-trans retinoic acid (ATRA), effectively inhibits the growth of several types of tumor cells; however, its molecular mechanism remains unclear. Fenretinide 40-51 haptoglobin-related protein Homo sapiens 33-36 28988109-3 2017 We show here for the first time that ZIKV NS5 is recognized with high nanomolar affinity by the host cell importin alpha/beta1 heterodimer, and that this interaction can be blocked by the novel DENV NS5 targeting inhibitor N-(4-hydroxyphenyl) retinamide (4-HPR). Fenretinide 223-253 haptoglobin-related protein Homo sapiens 257-260 29500274-8 2018 Complementary experiments in these animal models using the DES1 inhibitor fenretinide show more modest effects on cone recovery. Fenretinide 74-85 delta 4-desaturase, sphingolipid 1 Homo sapiens 59-63 28692043-13 2017 Treatment of breast cancer xenografts in vivo with the retinoid fenretinide reduced the accumulation of CK5+ cells during estrogen depletion. Fenretinide 64-75 keratin 5 Homo sapiens 104-107 28256636-0 2017 Elevated Fibroblast growth factor 21 (FGF21) in obese, insulin resistant states is normalised by the synthetic retinoid Fenretinide in mice. Fenretinide 120-131 fibroblast growth factor 21 Mus musculus 9-36 28695226-0 2017 Fenretinide differentially modulates the levels of long- and very long-chain ceramides by downregulating Cers5 enzyme: evidence from bench to bedside. Fenretinide 0-11 ceramide synthase 5 Homo sapiens 105-110 28695226-13 2017 Fenretinide downregulates the levels of LCCs and upregulates the levels of VLCCs. Fenretinide 0-11 GLE1 RNA export mediator Homo sapiens 40-44 28695226-14 2017 Fenretinide changes the balance of LCCs and VLCCs by downregulating Cers5 enzyme. Fenretinide 0-11 GLE1 RNA export mediator Homo sapiens 35-39 28695226-14 2017 Fenretinide changes the balance of LCCs and VLCCs by downregulating Cers5 enzyme. Fenretinide 0-11 ceramide synthase 5 Homo sapiens 68-73 28448568-0 2017 Inhibitory effects of fenretinide metabolites N-[4-methoxyphenyl]retinamide (MPR) and 4-oxo-N-(4-hydroxyphenyl)retinamide (3-keto-HPR) on fenretinide molecular targets beta-carotene oxygenase 1, stearoyl-CoA desaturase 1 and dihydroceramide Delta4-desaturase 1. Fenretinide 22-33 haptoglobin-related protein Homo sapiens 130-133 28448568-0 2017 Inhibitory effects of fenretinide metabolites N-[4-methoxyphenyl]retinamide (MPR) and 4-oxo-N-(4-hydroxyphenyl)retinamide (3-keto-HPR) on fenretinide molecular targets beta-carotene oxygenase 1, stearoyl-CoA desaturase 1 and dihydroceramide Delta4-desaturase 1. Fenretinide 22-33 beta-carotene oxygenase 1 Homo sapiens 168-193 28448568-0 2017 Inhibitory effects of fenretinide metabolites N-[4-methoxyphenyl]retinamide (MPR) and 4-oxo-N-(4-hydroxyphenyl)retinamide (3-keto-HPR) on fenretinide molecular targets beta-carotene oxygenase 1, stearoyl-CoA desaturase 1 and dihydroceramide Delta4-desaturase 1. Fenretinide 22-33 stearoyl-CoA desaturase Homo sapiens 195-220 28448568-0 2017 Inhibitory effects of fenretinide metabolites N-[4-methoxyphenyl]retinamide (MPR) and 4-oxo-N-(4-hydroxyphenyl)retinamide (3-keto-HPR) on fenretinide molecular targets beta-carotene oxygenase 1, stearoyl-CoA desaturase 1 and dihydroceramide Delta4-desaturase 1. Fenretinide 22-33 delta 4-desaturase, sphingolipid 1 Homo sapiens 247-260 28448568-1 2017 The therapeutic capacity of fenretinide (N-[4-hydroxyphenyl] retinamide; 4-HPR) has been demonstrated for several conditions, including cancer, obesity, diabetes, and ocular disease. Fenretinide 28-39 haptoglobin-related protein Homo sapiens 75-78 28448568-1 2017 The therapeutic capacity of fenretinide (N-[4-hydroxyphenyl] retinamide; 4-HPR) has been demonstrated for several conditions, including cancer, obesity, diabetes, and ocular disease. Fenretinide 41-71 haptoglobin-related protein Homo sapiens 75-78 28448568-3 2017 We hypothesized that investigation of two of the major physiological metabolites of fenretinide, N-[4-methoxyphenyl]retinamide (MPR) and 4-oxo-N-(4-hydroxyphenyl)retinamide (3-keto-HPR), might begin to resolve the multifaceted effects of this synthetic retinoid. Fenretinide 84-95 haptoglobin-related protein Homo sapiens 181-184 28448568-6 2017 The data demonstrate that while fenretinide is an inhibitor of the activities of these three enzymes, that 3-keto-HPR is a more potent inhibitor of all three enzymes, potentially mediating most of the in vivo beneficial effects of fenretinide. Fenretinide 231-242 haptoglobin-related protein Homo sapiens 114-117 28275870-1 2017 PURPOSE: By a scaffold shortening strategy, a small series of retinoidal amides fenretinide (4-HPR) analogs have been synthesized from alpha, beta-ionones and tested for their antiproliferative and differentiating activities, and antioxidant effect. Fenretinide 80-91 haptoglobin-related protein Homo sapiens 95-98 28119491-6 2017 Fenretinide + romidepsin caused a reactive oxygen species (ROS)-dependent increase in proapoptotic proteins (Bim, tBid, Bax, and Bak), apoptosis, and inhibition of HDAC enzymatic activity, which achieved a synergistic increase in histone acetylation. Fenretinide 0-11 BCL2 like 11 Homo sapiens 109-112 28119491-6 2017 Fenretinide + romidepsin caused a reactive oxygen species (ROS)-dependent increase in proapoptotic proteins (Bim, tBid, Bax, and Bak), apoptosis, and inhibition of HDAC enzymatic activity, which achieved a synergistic increase in histone acetylation. Fenretinide 0-11 BCL2 associated X, apoptosis regulator Homo sapiens 120-123 28119491-6 2017 Fenretinide + romidepsin caused a reactive oxygen species (ROS)-dependent increase in proapoptotic proteins (Bim, tBid, Bax, and Bak), apoptosis, and inhibition of HDAC enzymatic activity, which achieved a synergistic increase in histone acetylation. Fenretinide 0-11 BCL2 antagonist/killer 1 Homo sapiens 129-132 28119491-8 2017 Romidepsin + fenretinide activated p38 and JNK via ROS, and knockdown of p38 and JNK1 significantly decreased the synergistic cytotoxicity. Fenretinide 13-24 mitogen-activated protein kinase 14 Homo sapiens 35-38 28119491-8 2017 Romidepsin + fenretinide activated p38 and JNK via ROS, and knockdown of p38 and JNK1 significantly decreased the synergistic cytotoxicity. Fenretinide 13-24 mitogen-activated protein kinase 8 Homo sapiens 43-46 28340497-0 2017 P450 inhibitor ketoconazole increased the intratumor drug levels and antitumor activity of fenretinide in human neuroblastoma xenograft models. Fenretinide 91-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 28340497-1 2017 We previously reported that concurrent ketoconazole, an oral anti-fungal agent and P450 enzyme inhibitor, increased plasma levels of the cytotoxic retinoid, fenretinide (4-HPR) in mice. Fenretinide 157-168 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 83-87 28340497-1 2017 We previously reported that concurrent ketoconazole, an oral anti-fungal agent and P450 enzyme inhibitor, increased plasma levels of the cytotoxic retinoid, fenretinide (4-HPR) in mice. Fenretinide 157-168 haptoglobin-related protein Homo sapiens 172-175 28429653-1 2017 Fenretinide (4-HPR) is a synthetic retinoid that has cytotoxic activity against cancer cells. Fenretinide 0-11 haptoglobin-related protein Homo sapiens 15-18 28256636-0 2017 Elevated Fibroblast growth factor 21 (FGF21) in obese, insulin resistant states is normalised by the synthetic retinoid Fenretinide in mice. Fenretinide 120-131 fibroblast growth factor 21 Mus musculus 38-43 28256636-0 2017 Elevated Fibroblast growth factor 21 (FGF21) in obese, insulin resistant states is normalised by the synthetic retinoid Fenretinide in mice. Fenretinide 120-131 insulin Homo sapiens 55-62 28256636-5 2017 Fenretinide normalised elevated levels of FGF21 in both high-fat diet-induced obese mice and in genetically obese-diabetic Leprdbmice. Fenretinide 0-11 fibroblast growth factor 21 Mus musculus 42-47 28256636-6 2017 Moreover, Fenretinide-mediated suppression of FGF21 was independent of body weight loss or improved hepatic insulin sensitivity and importantly does not induce unhealthy metabolic complications. Fenretinide 10-21 fibroblast growth factor 21 Mus musculus 46-51 28256636-8 2017 The repression of FGF21 levels by Fenretinide occurs by reduced binding of RARalpha and Pol-II at the Fgf21 promoter. Fenretinide 34-45 fibroblast growth factor 21 Mus musculus 18-23 28256636-8 2017 The repression of FGF21 levels by Fenretinide occurs by reduced binding of RARalpha and Pol-II at the Fgf21 promoter. Fenretinide 34-45 retinoic acid receptor, alpha Mus musculus 75-83 28256636-8 2017 The repression of FGF21 levels by Fenretinide occurs by reduced binding of RARalpha and Pol-II at the Fgf21 promoter. Fenretinide 34-45 fibroblast growth factor 21 Mus musculus 102-107 28256636-9 2017 We therefore establish Fgf21 as a novel gene target of Fenretinide signalling via a retinoid-dependent mechanism. Fenretinide 55-66 fibroblast growth factor 21 Mus musculus 23-28 28179290-1 2017 BACKGROUND/AIM: Combining an anticancer agent fenretinide (HPR) or C6-pyridinium ceramide (LCL29) with Foscan-mediated photodynamic therapy (FoscanPDT) is expected to augment anticancer benefits of each substance. Fenretinide 46-57 haptoglobin-related protein Homo sapiens 59-62 27843299-0 2016 Fenretinide inhibits macrophage inflammatory mediators and controls hypertension in spontaneously hypertensive rats via the peroxisome proliferator-activated receptor gamma pathway. Fenretinide 0-11 peroxisome proliferator-activated receptor gamma Rattus norvegicus 124-172 27701038-1 2017 A simple and accurate high-performance liquid chromatography-tandem mass spectrometry (LC-MS/MS) method was developed for the determination of N-(4-hydroxyphenyl)retinamide (fenretinide, 4-HPR) and its metabolites, 4-oxo-N-(4-hydroxyphenyl)retinamide (4-oxo-4-HPR) and N-(4-methoxyphenyl)retinamide (4-MPR), in human plasma. Fenretinide 143-172 haptoglobin-related protein Homo sapiens 189-192 27701038-1 2017 A simple and accurate high-performance liquid chromatography-tandem mass spectrometry (LC-MS/MS) method was developed for the determination of N-(4-hydroxyphenyl)retinamide (fenretinide, 4-HPR) and its metabolites, 4-oxo-N-(4-hydroxyphenyl)retinamide (4-oxo-4-HPR) and N-(4-methoxyphenyl)retinamide (4-MPR), in human plasma. Fenretinide 143-172 haptoglobin-related protein Homo sapiens 260-263 27843299-6 2016 These effects were blocked by the pretreatment with PPARgamma antagonist in a dose-dependent manner, indicating fenretinide activated PPARgamma to exert anti-inflammation activity. Fenretinide 112-123 peroxisome proliferator-activated receptor gamma Rattus norvegicus 52-61 27843299-6 2016 These effects were blocked by the pretreatment with PPARgamma antagonist in a dose-dependent manner, indicating fenretinide activated PPARgamma to exert anti-inflammation activity. Fenretinide 112-123 peroxisome proliferator-activated receptor gamma Rattus norvegicus 134-143 27843299-8 2016 Taken together, these results indicate that fenretinide might be a potent antihypertensive agent that works by suppressing inflammation via activating PPARgamma. Fenretinide 44-55 peroxisome proliferator-activated receptor gamma Rattus norvegicus 151-160 27843299-3 2016 In this study, we have shown that treatment with lipopolysaccharide (LPS) decreased the expression of peroxisome proliferator-activated receptor gamma (PPARgamma) in RAW264.7 macrophages, and pretreatment with fenretinide reversed the effect of LPS on PPARgamma expression. Fenretinide 210-221 peroxisome proliferator-activated receptor gamma Rattus norvegicus 102-150 27843299-3 2016 In this study, we have shown that treatment with lipopolysaccharide (LPS) decreased the expression of peroxisome proliferator-activated receptor gamma (PPARgamma) in RAW264.7 macrophages, and pretreatment with fenretinide reversed the effect of LPS on PPARgamma expression. Fenretinide 210-221 peroxisome proliferator-activated receptor gamma Rattus norvegicus 152-161 27843299-3 2016 In this study, we have shown that treatment with lipopolysaccharide (LPS) decreased the expression of peroxisome proliferator-activated receptor gamma (PPARgamma) in RAW264.7 macrophages, and pretreatment with fenretinide reversed the effect of LPS on PPARgamma expression. Fenretinide 210-221 peroxisome proliferator-activated receptor gamma Rattus norvegicus 252-261 27843299-4 2016 In addition, LPS-induced pro-inflammatory cytokine production, including tumor necrosis factor-alpha, interleukin 6, and monocyte chemoattractant protein 1 were dose-dependently reversed by fenretinide, and the effects of fenretinide on LPS-induced pro-inflammatory cytokine production were blocked by treatment with PPARgamma antagonist. Fenretinide 190-201 tumor necrosis factor Rattus norvegicus 73-100 27843299-4 2016 In addition, LPS-induced pro-inflammatory cytokine production, including tumor necrosis factor-alpha, interleukin 6, and monocyte chemoattractant protein 1 were dose-dependently reversed by fenretinide, and the effects of fenretinide on LPS-induced pro-inflammatory cytokine production were blocked by treatment with PPARgamma antagonist. Fenretinide 190-201 interleukin 6 Rattus norvegicus 102-115 27843299-4 2016 In addition, LPS-induced pro-inflammatory cytokine production, including tumor necrosis factor-alpha, interleukin 6, and monocyte chemoattractant protein 1 were dose-dependently reversed by fenretinide, and the effects of fenretinide on LPS-induced pro-inflammatory cytokine production were blocked by treatment with PPARgamma antagonist. Fenretinide 190-201 C-C motif chemokine ligand 2 Rattus norvegicus 121-155 27843299-4 2016 In addition, LPS-induced pro-inflammatory cytokine production, including tumor necrosis factor-alpha, interleukin 6, and monocyte chemoattractant protein 1 were dose-dependently reversed by fenretinide, and the effects of fenretinide on LPS-induced pro-inflammatory cytokine production were blocked by treatment with PPARgamma antagonist. Fenretinide 190-201 peroxisome proliferator-activated receptor gamma Rattus norvegicus 317-326 27843299-4 2016 In addition, LPS-induced pro-inflammatory cytokine production, including tumor necrosis factor-alpha, interleukin 6, and monocyte chemoattractant protein 1 were dose-dependently reversed by fenretinide, and the effects of fenretinide on LPS-induced pro-inflammatory cytokine production were blocked by treatment with PPARgamma antagonist. Fenretinide 222-233 interleukin 6 Rattus norvegicus 102-115 27843299-4 2016 In addition, LPS-induced pro-inflammatory cytokine production, including tumor necrosis factor-alpha, interleukin 6, and monocyte chemoattractant protein 1 were dose-dependently reversed by fenretinide, and the effects of fenretinide on LPS-induced pro-inflammatory cytokine production were blocked by treatment with PPARgamma antagonist. Fenretinide 222-233 C-C motif chemokine ligand 2 Rattus norvegicus 121-155 27628049-6 2016 Furthermore, we show that fenretinide or the ER stress inhibitor 4-PBA decreased curcumin-induced Glucose-regulated protein 78 (GRP78) upregulation, and produced a similar enhanced cytotoxic effect. Fenretinide 26-37 heat shock protein family A (Hsp70) member 5 Homo sapiens 98-126 27530131-2 2016 Fenretinide (4-HPR) is a synthetic retinoid. Fenretinide 0-11 haptoglobin-related protein Homo sapiens 15-18 27628049-6 2016 Furthermore, we show that fenretinide or the ER stress inhibitor 4-PBA decreased curcumin-induced Glucose-regulated protein 78 (GRP78) upregulation, and produced a similar enhanced cytotoxic effect. Fenretinide 26-37 heat shock protein family A (Hsp70) member 5 Homo sapiens 128-133 27367907-0 2016 Fenretinide (4-HPR) Targets Caspase-9, ERK 1/2 and the Wnt3a/beta-Catenin Pathway in Medulloblastoma Cells and Medulloblastoma Cell Spheroids. Fenretinide 0-11 haptoglobin-related protein Homo sapiens 15-18 27367907-0 2016 Fenretinide (4-HPR) Targets Caspase-9, ERK 1/2 and the Wnt3a/beta-Catenin Pathway in Medulloblastoma Cells and Medulloblastoma Cell Spheroids. Fenretinide 0-11 mitogen-activated protein kinase 3 Homo sapiens 39-46 26919975-6 2016 Quantitative reverse transcription-polymerase chain reaction from endometriotic cells treated with fenretinide was used to examine expression of genes involved in RA signaling including stimulated by RA 6 (STRA6), cellular RA binding protein 2 (CRABP2), and fatty acid binding protein 5 (FABP5). Fenretinide 99-110 fatty acid binding protein 5, epidermal Mus musculus 258-286 26919975-6 2016 Quantitative reverse transcription-polymerase chain reaction from endometriotic cells treated with fenretinide was used to examine expression of genes involved in RA signaling including stimulated by RA 6 (STRA6), cellular RA binding protein 2 (CRABP2), and fatty acid binding protein 5 (FABP5). Fenretinide 99-110 fatty acid binding protein 5, epidermal Mus musculus 288-293 26919975-13 2016 CONCLUSIONS: Fenretinide increases STRA6 expression thereby potentially reversing the pathological loss of retinoid availability. Fenretinide 13-24 stimulated by retinoic acid gene 6 Mus musculus 35-40 27367907-0 2016 Fenretinide (4-HPR) Targets Caspase-9, ERK 1/2 and the Wnt3a/beta-Catenin Pathway in Medulloblastoma Cells and Medulloblastoma Cell Spheroids. Fenretinide 0-11 Wnt family member 3A Homo sapiens 55-60 27367907-0 2016 Fenretinide (4-HPR) Targets Caspase-9, ERK 1/2 and the Wnt3a/beta-Catenin Pathway in Medulloblastoma Cells and Medulloblastoma Cell Spheroids. Fenretinide 0-11 caspase 9 Homo sapiens 28-37 27367907-0 2016 Fenretinide (4-HPR) Targets Caspase-9, ERK 1/2 and the Wnt3a/beta-Catenin Pathway in Medulloblastoma Cells and Medulloblastoma Cell Spheroids. Fenretinide 0-11 catenin beta 1 Homo sapiens 61-73 27367907-3 2016 N-(4-Hydroxyphenyl) retinamide (4-HPR, fenretinide), a synthetic analogue of all-trans retinoic acid, has emerged as a promising and well-tolerated cancer chemopreventive and chemotherapeutic agent for various neoplasms, from breast cancer to neuroblastoma. Fenretinide 0-30 haptoglobin-related protein Homo sapiens 34-37 27367907-3 2016 N-(4-Hydroxyphenyl) retinamide (4-HPR, fenretinide), a synthetic analogue of all-trans retinoic acid, has emerged as a promising and well-tolerated cancer chemopreventive and chemotherapeutic agent for various neoplasms, from breast cancer to neuroblastoma. Fenretinide 39-50 haptoglobin-related protein Homo sapiens 34-37 27045034-0 2016 Prognostic Significance of VEGF after Twenty-Year Follow-up in a Randomized Trial of Fenretinide in Non-Muscle-Invasive Bladder Cancer. Fenretinide 85-96 vascular endothelial growth factor A Homo sapiens 27-31 26934645-5 2016 Furthermore, SKi or ABC294640 or fenretinide increase the expression of the senescence markers, p53 and p21 in LNCaP-AI prostate cancer cells. Fenretinide 33-44 tumor protein p53 Homo sapiens 96-99 27085050-3 2016 In this study we tested the hypothesis that combining Foscan-mediated PDT with desaturase inhibitor fenretinide (HPR) enhances cancer cell killing. Fenretinide 100-111 haptoglobin-related protein Homo sapiens 113-116 26934645-3 2016 These effects are recapitulated by the dihydroceramide desaturase (Des1) inhibitor, fenretinide. Fenretinide 84-95 delta 4-desaturase, sphingolipid 1 Homo sapiens 67-71 26934645-5 2016 Furthermore, SKi or ABC294640 or fenretinide increase the expression of the senescence markers, p53 and p21 in LNCaP-AI prostate cancer cells. Fenretinide 33-44 H3 histone pseudogene 16 Homo sapiens 104-107 25739041-2 2015 The clinically-relevant fenretinide [N-(4-hydroxyphenyl) retinamide; 4HPR], was combined with the silicon phthalocyanine photosensitizer Pc4-mediated PDT to test for their potential to enhance killing of SCC17B cells, a clinically-relevant model of human head and neck squamous cell carcinoma. Fenretinide 69-73 keratin 6B Homo sapiens 137-140 26592777-11 2016 FEN and 4-OXO (but not RA) treatment additionally led to the activation of p38-MAPK, peIF2alpha and autophagy markers in adipocytes. Fenretinide 0-3 mitogen-activated protein kinase 14 Mus musculus 75-78 26843908-3 2016 The p75 neurotrophin receptor (p75NTR) enhances the antineuroblastoma cell efficacy of fenretinide in vitro. Fenretinide 87-98 nerve growth factor receptor Homo sapiens 4-29 26843908-3 2016 The p75 neurotrophin receptor (p75NTR) enhances the antineuroblastoma cell efficacy of fenretinide in vitro. Fenretinide 87-98 nerve growth factor receptor Homo sapiens 31-37 26843908-4 2016 We examined the role of the retinoid binding protein, CRABP1, in p75NTR-mediated potentiation of the efficacy of fenretinide. Fenretinide 113-124 cellular retinoic acid binding protein 1 Homo sapiens 54-60 26843908-4 2016 We examined the role of the retinoid binding protein, CRABP1, in p75NTR-mediated potentiation of the efficacy of fenretinide. Fenretinide 113-124 nerve growth factor receptor Homo sapiens 65-71 26843908-11 2016 Potentiation of the antineuroblastoma cell effects of fenretinide by p75NTR is not mediated solely through CRABP1. Fenretinide 54-65 nerve growth factor receptor Homo sapiens 69-75 26237500-0 2015 Bortezomib and fenretinide induce synergistic cytotoxicity in mantle cell lymphoma through apoptosis, cell-cycle dysregulation, and IkappaBalpha kinase downregulation. Fenretinide 15-26 NFKB inhibitor alpha Homo sapiens 132-144 26220867-1 2015 Fenretinide, N-(4-hydroxyphenyl)retinamide, (4-HPR), a synthetic retinoid, owes its cancer-toxic effects in part to the generation of ceramide, a potent tumor-suppressing sphingolipid. Fenretinide 0-11 haptoglobin-related protein Homo sapiens 47-50 26220867-1 2015 Fenretinide, N-(4-hydroxyphenyl)retinamide, (4-HPR), a synthetic retinoid, owes its cancer-toxic effects in part to the generation of ceramide, a potent tumor-suppressing sphingolipid. Fenretinide 13-42 haptoglobin-related protein Homo sapiens 47-50 26389809-8 2016 CONCLUSIONS: The expression of AQP4 and AQP6 is downregulated during fenretinide-induced transdifferentiation. Fenretinide 69-80 aquaporin 4 Homo sapiens 31-35 26389809-8 2016 CONCLUSIONS: The expression of AQP4 and AQP6 is downregulated during fenretinide-induced transdifferentiation. Fenretinide 69-80 aquaporin 6 Homo sapiens 40-44 26640617-6 2016 We therefore hypothesized that the fenretinide-potentiating effects of p75NTR are the result of transcriptional enrichment of Complex II by p75ICD. Fenretinide 35-46 nerve growth factor receptor Homo sapiens 71-77 26640617-6 2016 We therefore hypothesized that the fenretinide-potentiating effects of p75NTR are the result of transcriptional enrichment of Complex II by p75ICD. Fenretinide 35-46 nerve growth factor receptor Homo sapiens 140-146 25963741-0 2015 Thymosin-beta4 is a determinant of drug sensitivity for Fenretinide and Vorinostat combination therapy in neuroblastoma. Fenretinide 56-67 thymosin beta 4 X-linked Homo sapiens 0-14 25963741-5 2015 The 4-HPR + SAHA combination induced caspase-dependent apoptosis through activation of caspase 3, reduced colony formation and cell migration in vitro, and tumorigenicity in vivo. Fenretinide 4-9 caspase 3 Homo sapiens 87-96 25712051-0 2015 Fenretinide Perturbs Focal Adhesion Kinase in Premalignant and Malignant Human Oral Keratinocytes. Fenretinide 0-11 protein tyrosine kinase 2 Homo sapiens 21-42 25712051-1 2015 Fenretinide"s Chemopreventive Mechanisms Include ECM Interactions. Fenretinide 0-11 multimerin 1 Homo sapiens 49-52 25712051-5 2015 The cancer chemopreventive and synthetic vitamin A derivative, fenretinide, has demonstrated protein-binding capacities, for example, mTOR- and retinol-binding protein interactions. Fenretinide 63-74 mechanistic target of rapamycin kinase Homo sapiens 134-138 25712051-6 2015 These studies used a continuum of human oral keratinocytes (normal-HPV E6/E7-transduced-OSCC) to assess potential fenretinide-FAK drug protein interactions and functional consequences on cellular growth regulation and motility. Fenretinide 114-125 protein tyrosine kinase 2 Homo sapiens 126-129 25712051-7 2015 Molecular modeling studies demonstrated that fenretinide has approximately 200-fold greater binding affinity relative to the natural ligand (ATP) at FAK"s kinase domain. Fenretinide 45-56 protein tyrosine kinase 2 Homo sapiens 149-152 25712051-8 2015 Fenretinide also shows intermediate binding at FAK"s FERM domain and interacts at the ATP-binding site of the closest FAK analogue, PYK2. Fenretinide 0-11 protein tyrosine kinase 2 Homo sapiens 47-50 25712051-8 2015 Fenretinide also shows intermediate binding at FAK"s FERM domain and interacts at the ATP-binding site of the closest FAK analogue, PYK2. Fenretinide 0-11 protein tyrosine kinase 2 Homo sapiens 118-121 25712051-8 2015 Fenretinide also shows intermediate binding at FAK"s FERM domain and interacts at the ATP-binding site of the closest FAK analogue, PYK2. Fenretinide 0-11 protein tyrosine kinase 2 beta Homo sapiens 132-136 25712051-14 2015 Our data imply that fenretinide is uniquely capable of disrupting FAK"s and PYK2"s prosurvival and mobility-enhancing effects and further extend fenretinide"s chemopreventive contributions beyond induction of apoptosis and differentiation. Fenretinide 20-31 protein tyrosine kinase 2 Homo sapiens 66-69 25712051-14 2015 Our data imply that fenretinide is uniquely capable of disrupting FAK"s and PYK2"s prosurvival and mobility-enhancing effects and further extend fenretinide"s chemopreventive contributions beyond induction of apoptosis and differentiation. Fenretinide 20-31 protein tyrosine kinase 2 beta Homo sapiens 76-80 25313218-4 2015 We screened a library of bioactive lipids and modulators of lipid metabolism and identified 4-hydroxyphenyl retinamide (4-HPR) (fenretinide) as an inhibitor of DENV in cell culture. Fenretinide 92-118 haptoglobin-related protein Homo sapiens 122-125 25576804-4 2015 In the current study, N-(4-hydroxyphenyl)retinamide (4-HPR), a synthetic retinoid, was used as a drug model of ROS induction to investigate its synergistic effect with AgNPs. Fenretinide 22-51 haptoglobin-related protein Homo sapiens 55-58 25461293-3 2015 The high expression of caveolin-1 on the A549 cell surface further enhanced the antitumor activity of the nanoencapsulated fenretinide. Fenretinide 123-134 caveolin 1 Homo sapiens 23-33 25461293-4 2015 Caveolin-1 favored albumin uptake and improved the efficacy of the fenretinide-loaded albumin nanocapsules, especially in 3-D cultures where the densely packed 3-D structures impaired drug diffusibility and severely reduced the activity of the free drug. Fenretinide 67-78 caveolin 1 Homo sapiens 0-10 25313218-4 2015 We screened a library of bioactive lipids and modulators of lipid metabolism and identified 4-hydroxyphenyl retinamide (4-HPR) (fenretinide) as an inhibitor of DENV in cell culture. Fenretinide 128-139 haptoglobin-related protein Homo sapiens 122-125 24885263-7 2014 Our data demonstrate that treatment of allergen-sensitized mice with fenretinide before allergen challenge prevents ovalbumin-induced changes in the AA/DHA ratio. Fenretinide 69-80 serine (or cysteine) peptidase inhibitor, clade B, member 1, pseudogene Mus musculus 116-125 24885263-8 2014 The levels of several metabolites, such as serotonin, and markers of cellular stress, which are increased after ovalbumin challenge, are also controlled by fenretinide treatment. Fenretinide 156-167 serine (or cysteine) peptidase inhibitor, clade B, member 1, pseudogene Mus musculus 112-121 24885263-9 2014 We observed the protective effect of fenretinide against ovalbumin-induced airway hyperresponsiveness and inflammation in the lungs, illustrated by a complete block in the infiltration of inflammatory cells to the airways and dramatically diminished goblet cell proliferation, even though IgE remained high. Fenretinide 37-48 serine (or cysteine) peptidase inhibitor, clade B, member 1, pseudogene Mus musculus 57-66 25414197-10 2014 CONCLUSIONS: These findings improve our understanding of the mechanism of cell death in Prominin-1-related disease and provide evidence that fenretinide may be worth studying in human disease. Fenretinide 141-152 prominin 1 Homo sapiens 88-98 25340777-6 2014 Further, we have shown that this induction of apoptosis by fenretinide may be caused by increased retinol uptake via STRA6. Fenretinide 59-70 signaling receptor and transporter of retinol STRA6 Homo sapiens 117-122 24392637-4 2014 RESULTS: Increasing concentrations of N-(4-hydroxyphenyl) retinamide (4-HPR)-treatment pushed autophagy down to apoptosis in a dose-dependent manner, and 4-HPR-induced ROS contribute to this process. Fenretinide 38-68 haptoglobin-related protein Homo sapiens 72-75 24392637-4 2014 RESULTS: Increasing concentrations of N-(4-hydroxyphenyl) retinamide (4-HPR)-treatment pushed autophagy down to apoptosis in a dose-dependent manner, and 4-HPR-induced ROS contribute to this process. Fenretinide 38-68 haptoglobin-related protein Homo sapiens 156-159 24357007-0 2014 Fenretinide induces ubiquitin-dependent proteasomal degradation of stearoyl-CoA desaturase in human retinal pigment epithelial cells. Fenretinide 0-11 stearoyl-CoA desaturase Homo sapiens 67-90 25069047-7 2014 This review covers the current knowledge about Fenretinide"s use as a therapy for cancer and potential to treat obesity, insulin resistance and glucose intolerance. Fenretinide 47-58 insulin Homo sapiens 121-128 24934240-5 2014 (N-(4-Hydroxyphenyl) retinamide), a potent anti-angiogenic agent, could not only down-regulate the expression of vascular epithelial growth factor, but also decrease endocan transcription and expression in NB4 cells, a human acute promyelocytic leukemia cell line. Fenretinide 1-32 endothelial cell specific molecule 1 Homo sapiens 167-174 25246272-3 2014 The antioxidant N-acetylcysteine decreased GSK3beta phosphorylation and poly(ADP-ribose) polymerase cleavage induced by 4HPR, As2O3, and PEITC, implicating oxidative stress in these effects. Fenretinide 120-124 glycogen synthase kinase 3 beta Homo sapiens 43-51 24357007-3 2014 We examined whether fenretinide, (N-(4-hydroxyphenyl)retinamide, 4HPR), which induces apoptosis in cancer cells and recently shown to improve insulin sensitivity, can modulate the expression of SCD. Fenretinide 20-31 stearoyl-CoA desaturase Homo sapiens 194-197 24357007-3 2014 We examined whether fenretinide, (N-(4-hydroxyphenyl)retinamide, 4HPR), which induces apoptosis in cancer cells and recently shown to improve insulin sensitivity, can modulate the expression of SCD. Fenretinide 33-63 stearoyl-CoA desaturase Homo sapiens 194-197 24357007-3 2014 We examined whether fenretinide, (N-(4-hydroxyphenyl)retinamide, 4HPR), which induces apoptosis in cancer cells and recently shown to improve insulin sensitivity, can modulate the expression of SCD. Fenretinide 65-69 stearoyl-CoA desaturase Homo sapiens 194-197 24357007-4 2014 We observed that fenretinide decreased SCD protein and enzymatic activity in the ARPE-19 human retinal pigment epithelial cell line. Fenretinide 17-28 stearoyl-CoA desaturase Homo sapiens 39-42 24357007-8 2014 In addition, PYR41, an inhibitor of ubiquitin activating enzyme E1, blocked the fenretinide-mediated decrease in SCD. Fenretinide 80-91 stearoyl-CoA desaturase Homo sapiens 113-116 24357007-9 2014 Immunoprecipitation analysis using anti-ubiquitin and anti-SCD antibodies and the blocking of SCD loss by PYR41 inhibition of ubiquitination further corroborate that fenretinide mediates the degradation of SCD in human RPE cells via the ubiquitin-proteasome dependent pathway. Fenretinide 166-177 stearoyl-CoA desaturase Homo sapiens 59-62 24357007-9 2014 Immunoprecipitation analysis using anti-ubiquitin and anti-SCD antibodies and the blocking of SCD loss by PYR41 inhibition of ubiquitination further corroborate that fenretinide mediates the degradation of SCD in human RPE cells via the ubiquitin-proteasome dependent pathway. Fenretinide 166-177 stearoyl-CoA desaturase Homo sapiens 94-97 24357007-10 2014 Therefore, the effect of fenretinide on SCD should be considered in its potential therapeutic role against cancer, type-2 diabetes, and retinal diseases. Fenretinide 25-36 stearoyl-CoA desaturase Homo sapiens 40-43 24835984-4 2014 Inhibitor co-crystal X-ray structures revealed unique disruptions of RBP4-TTR interactions by our compounds through induced loop conformational changes instead of steric hindrance exemplified by fenretinide. Fenretinide 195-206 retinol binding protein 4, plasma Mus musculus 69-73 25015569-0 2014 Anti-tumor activity of fenretinide complexed with human serum albumin in lung cancer xenograft mouse model. Fenretinide 23-34 albumin Mus musculus 62-69 24835984-4 2014 Inhibitor co-crystal X-ray structures revealed unique disruptions of RBP4-TTR interactions by our compounds through induced loop conformational changes instead of steric hindrance exemplified by fenretinide. Fenretinide 195-206 transthyretin Mus musculus 74-77 23764045-8 2013 Targeting these activated pro-proliferative genes by combined approaches of HDAC-inhibitors plus fenretinide, which inhibits cyclinD1, exhibit strong synergistic effects on induction of apoptosis. Fenretinide 97-108 cyclin D1 Homo sapiens 125-133 24755119-2 2014 The combination of fenretinide (4-HPR), a synthetic retinoid inducing apoptosis by ROS generation, and TRAIL, a cell death ligand inducing caspase-dependent apoptosis, might result in more powerful cytotoxic activity. Fenretinide 19-30 haptoglobin-related protein Homo sapiens 34-37 24755119-8 2014 Confirming apoptosis, active caspase-3 and cleaved PARP were increased by fenretinide or TRAIL in both western blotting and ELISA. Fenretinide 74-85 caspase 3 Homo sapiens 29-38 24755119-8 2014 Confirming apoptosis, active caspase-3 and cleaved PARP were increased by fenretinide or TRAIL in both western blotting and ELISA. Fenretinide 74-85 poly(ADP-ribose) polymerase 1 Homo sapiens 51-55 24021153-1 2014 AIMS: We have recently shown that fenretinide preferentially targets CD34(+) cells of acute myeloid leukemia (AML), and here, we test whether this agent exerts the effect on CD34(+) cells of chronic myeloid leukemia (CML), which are refractory to imatinib. Fenretinide 34-45 CD34 molecule Homo sapiens 69-73 24021153-2 2014 RESULTS: As tested by colony-forming cell assays using clinical specimens, both number and size of total colonies derived from CD34(+) CML cells were significantly reduced by fenretinide, and by combining fenretinide with imatinib. Fenretinide 175-186 CD34 molecule Homo sapiens 127-131 24021153-2 2014 RESULTS: As tested by colony-forming cell assays using clinical specimens, both number and size of total colonies derived from CD34(+) CML cells were significantly reduced by fenretinide, and by combining fenretinide with imatinib. Fenretinide 205-216 CD34 molecule Homo sapiens 127-131 24021153-4 2014 Accordantly, fenretinide appeared to induce apoptosis in CD34(+) CML cells, particularly with regard to the cells in the subpopulation of CD34(+)CD38(-). Fenretinide 13-24 CD34 molecule Homo sapiens 57-61 24021153-4 2014 Accordantly, fenretinide appeared to induce apoptosis in CD34(+) CML cells, particularly with regard to the cells in the subpopulation of CD34(+)CD38(-). Fenretinide 13-24 CD34 molecule Homo sapiens 138-142 24021153-4 2014 Accordantly, fenretinide appeared to induce apoptosis in CD34(+) CML cells, particularly with regard to the cells in the subpopulation of CD34(+)CD38(-). Fenretinide 13-24 CD38 molecule Homo sapiens 145-149 24021153-5 2014 Through cell quiescent assays, including Ki-67 negativity test, we added evidence that nonproliferative CD34(+) CML cells were largely eliminated by fenretinide. Fenretinide 149-160 CD34 molecule Homo sapiens 104-108 24021153-7 2014 INNOVATION AND CONCLUSION: As compared with CD34(+) AML cells, the apoptotic effects of fenretinide on CD34(+) CML cells were more prominent whereas less varied among the samples of different patients, and also various stress-responsive events appeared to be more robust in fenretinide-treated CD34(+) CML cells. Fenretinide 88-99 CD34 molecule Homo sapiens 103-107 24021153-7 2014 INNOVATION AND CONCLUSION: As compared with CD34(+) AML cells, the apoptotic effects of fenretinide on CD34(+) CML cells were more prominent whereas less varied among the samples of different patients, and also various stress-responsive events appeared to be more robust in fenretinide-treated CD34(+) CML cells. Fenretinide 88-99 CD34 molecule Homo sapiens 103-107 24192821-3 2013 The caspase3/9-dependent pathway was triggered significantly in combination treatment, and moreover, the AMPK pathway also mediated the apoptosis induction in fenretinide and selenite combination. Fenretinide 159-170 caspase 3 Homo sapiens 4-12 23813912-1 2013 BACKGROUND: A phase I study was conducted to determine the maximum-tolerated dose, dose-limiting toxicities (DLTs), and pharmacokinetics of fenretinide (4-HPR) delivered in an oral powderized lipid complex (LXS) in patients with relapsed/refractory neuroblastoma. Fenretinide 140-151 haptoglobin-related protein Homo sapiens 155-158 23792118-5 2013 Among them, fenretinide (HPR) has been considered one of the most promising anti-tumor agent but it is partially efficacious due to both poor aqueous solubility and rapid metabolism. Fenretinide 12-23 haptoglobin-related protein Homo sapiens 25-28 24040340-1 2013 We previously reported that fenretinide (4-HPR) was cytotoxic to acute lymphoblastic leukemia (ALL) cell lines in vitro in association with increased levels of de novo synthesized dihydroceramides, the immediate precursors of ceramides. Fenretinide 28-39 haptoglobin-related protein Homo sapiens 43-46 23764045-0 2013 The histone deacetylase inhibitor SAHA acts in synergism with fenretinide and doxorubicin to control growth of rhabdoid tumor cells. Fenretinide 62-73 histone deacetylase 9 Homo sapiens 4-23 24253178-2 2014 The retinoic acid analogue, fenretinide (4-hydroxyphenyl retinamide; 4-HPR), induces apoptosis in neuroblastoma cells in vitro and is currently in clinical trials for children with refractory neuroblastoma. Fenretinide 28-39 haptoglobin-related protein Homo sapiens 71-74 24253178-2 2014 The retinoic acid analogue, fenretinide (4-hydroxyphenyl retinamide; 4-HPR), induces apoptosis in neuroblastoma cells in vitro and is currently in clinical trials for children with refractory neuroblastoma. Fenretinide 41-67 haptoglobin-related protein Homo sapiens 71-74 24253178-6 2014 Specific inhibitors of JNK phosphorylation and scavengers of mitochondrial reactive oxygen species were used to demonstrate the roles of these phenomena in the enhancement of fenretinide efficacy. Fenretinide 175-186 mitogen-activated protein kinase 8 Homo sapiens 23-26 24069363-0 2013 Fenretinide corrects the imbalance between omega-6 to omega-3 polyunsaturated fatty acids and inhibits macrophage inflammatory mediators via the ERK pathway. Fenretinide 0-11 mitogen-activated protein kinase 1 Mus musculus 145-148 24069363-5 2013 Indeed, our results demonstrate that 4HPR inhibited the excessive production of inflammatory mediators, including TNF, IL-6, CCL2 and CCL-5 in LPS-stimulated FXR1-KO macrophages, by selectively inhibiting phosphorylation of ERK1/2, which is naturally more phosphorylated in FXR1-KO cells. Fenretinide 37-41 tumor necrosis factor Mus musculus 114-117 24069363-5 2013 Indeed, our results demonstrate that 4HPR inhibited the excessive production of inflammatory mediators, including TNF, IL-6, CCL2 and CCL-5 in LPS-stimulated FXR1-KO macrophages, by selectively inhibiting phosphorylation of ERK1/2, which is naturally more phosphorylated in FXR1-KO cells. Fenretinide 37-41 interleukin 6 Mus musculus 119-123 24069363-5 2013 Indeed, our results demonstrate that 4HPR inhibited the excessive production of inflammatory mediators, including TNF, IL-6, CCL2 and CCL-5 in LPS-stimulated FXR1-KO macrophages, by selectively inhibiting phosphorylation of ERK1/2, which is naturally more phosphorylated in FXR1-KO cells. Fenretinide 37-41 chemokine (C-C motif) ligand 2 Mus musculus 125-129 24069363-5 2013 Indeed, our results demonstrate that 4HPR inhibited the excessive production of inflammatory mediators, including TNF, IL-6, CCL2 and CCL-5 in LPS-stimulated FXR1-KO macrophages, by selectively inhibiting phosphorylation of ERK1/2, which is naturally more phosphorylated in FXR1-KO cells. Fenretinide 37-41 chemokine (C-C motif) ligand 5 Mus musculus 134-139 24069363-5 2013 Indeed, our results demonstrate that 4HPR inhibited the excessive production of inflammatory mediators, including TNF, IL-6, CCL2 and CCL-5 in LPS-stimulated FXR1-KO macrophages, by selectively inhibiting phosphorylation of ERK1/2, which is naturally more phosphorylated in FXR1-KO cells. Fenretinide 37-41 FMR1 autosomal homolog 1 Mus musculus 158-162 24069363-5 2013 Indeed, our results demonstrate that 4HPR inhibited the excessive production of inflammatory mediators, including TNF, IL-6, CCL2 and CCL-5 in LPS-stimulated FXR1-KO macrophages, by selectively inhibiting phosphorylation of ERK1/2, which is naturally more phosphorylated in FXR1-KO cells. Fenretinide 37-41 mitogen-activated protein kinase 3 Mus musculus 224-230 24069363-5 2013 Indeed, our results demonstrate that 4HPR inhibited the excessive production of inflammatory mediators, including TNF, IL-6, CCL2 and CCL-5 in LPS-stimulated FXR1-KO macrophages, by selectively inhibiting phosphorylation of ERK1/2, which is naturally more phosphorylated in FXR1-KO cells. Fenretinide 37-41 FMR1 autosomal homolog 1 Mus musculus 274-278 24069363-7 2013 Interestingly, treatment with 4HPR was associated with the normalization of arachidonic acid/docosahexaenoic acid ratio in macrophages, which we found to impact phosphorylation of ERK1/2. Fenretinide 30-34 mitogen-activated protein kinase 3 Mus musculus 180-186 24069363-8 2013 Overall, this study shows for the first time that 4HPR modulates inflammatory cytokine expression in macrophages by correcting a phospholipid-bound fatty acid imbalance that impacts the phosphorylation of ERK1/2. Fenretinide 50-54 mitogen-activated protein kinase 3 Mus musculus 205-211 23396089-8 2013 In addition, fenretinide inhibited the expression of the genes involved in RAS/RAF/ERK-mediated survival pathway. Fenretinide 13-24 mitogen-activated protein kinase 1 Homo sapiens 83-86 23396089-3 2013 Fenretinide and ATRA-induced gene expressions and DNA bindings were profiled using microarray and chromatin immunoprecipitation with anti-RXRalpha antibody. Fenretinide 0-11 retinoid X receptor alpha Homo sapiens 138-146 23396089-10 2013 Most genes regulated by fenretinide and ATRA were bound by RXRalpha, suggesting a direct effect. Fenretinide 24-35 retinoid X receptor alpha Homo sapiens 59-67 23396089-7 2013 However, fenretinide specifically induced Fas/TNFalpha-mediated apoptosis by increasing the expression of pro-apoptotic genes i.e., DEDD2, CASP8, CASP4, and HSPA1A/B; whereas, ATRA induced the expression of BIRC3 and TNFAIP3, which inhibit apoptosis by interacting with TRAF2. Fenretinide 9-20 tumor necrosis factor Homo sapiens 46-54 23396089-7 2013 However, fenretinide specifically induced Fas/TNFalpha-mediated apoptosis by increasing the expression of pro-apoptotic genes i.e., DEDD2, CASP8, CASP4, and HSPA1A/B; whereas, ATRA induced the expression of BIRC3 and TNFAIP3, which inhibit apoptosis by interacting with TRAF2. Fenretinide 9-20 death effector domain containing 2 Homo sapiens 132-137 23396089-7 2013 However, fenretinide specifically induced Fas/TNFalpha-mediated apoptosis by increasing the expression of pro-apoptotic genes i.e., DEDD2, CASP8, CASP4, and HSPA1A/B; whereas, ATRA induced the expression of BIRC3 and TNFAIP3, which inhibit apoptosis by interacting with TRAF2. Fenretinide 9-20 caspase 8 Homo sapiens 139-144 23396089-12 2013 The data also suggested that fenretinide induces apoptosis via death receptor effector and by inhibiting the RAS/RAF/ERK pathway. Fenretinide 29-40 zinc fingers and homeoboxes 2 Homo sapiens 113-116 23396089-12 2013 The data also suggested that fenretinide induces apoptosis via death receptor effector and by inhibiting the RAS/RAF/ERK pathway. Fenretinide 29-40 mitogen-activated protein kinase 1 Homo sapiens 117-120 23314735-0 2013 p75NTR: an enhancer of fenretinide toxicity in neuroblastoma. Fenretinide 23-34 nerve growth factor receptor Homo sapiens 0-6 23396089-7 2013 However, fenretinide specifically induced Fas/TNFalpha-mediated apoptosis by increasing the expression of pro-apoptotic genes i.e., DEDD2, CASP8, CASP4, and HSPA1A/B; whereas, ATRA induced the expression of BIRC3 and TNFAIP3, which inhibit apoptosis by interacting with TRAF2. Fenretinide 9-20 caspase 4 Homo sapiens 146-151 23396089-7 2013 However, fenretinide specifically induced Fas/TNFalpha-mediated apoptosis by increasing the expression of pro-apoptotic genes i.e., DEDD2, CASP8, CASP4, and HSPA1A/B; whereas, ATRA induced the expression of BIRC3 and TNFAIP3, which inhibit apoptosis by interacting with TRAF2. Fenretinide 9-20 heat shock protein family A (Hsp70) member 1A Homo sapiens 157-165 23396089-7 2013 However, fenretinide specifically induced Fas/TNFalpha-mediated apoptosis by increasing the expression of pro-apoptotic genes i.e., DEDD2, CASP8, CASP4, and HSPA1A/B; whereas, ATRA induced the expression of BIRC3 and TNFAIP3, which inhibit apoptosis by interacting with TRAF2. Fenretinide 9-20 baculoviral IAP repeat containing 3 Homo sapiens 207-212 23396089-7 2013 However, fenretinide specifically induced Fas/TNFalpha-mediated apoptosis by increasing the expression of pro-apoptotic genes i.e., DEDD2, CASP8, CASP4, and HSPA1A/B; whereas, ATRA induced the expression of BIRC3 and TNFAIP3, which inhibit apoptosis by interacting with TRAF2. Fenretinide 9-20 TNF alpha induced protein 3 Homo sapiens 217-224 23396089-7 2013 However, fenretinide specifically induced Fas/TNFalpha-mediated apoptosis by increasing the expression of pro-apoptotic genes i.e., DEDD2, CASP8, CASP4, and HSPA1A/B; whereas, ATRA induced the expression of BIRC3 and TNFAIP3, which inhibit apoptosis by interacting with TRAF2. Fenretinide 9-20 TNF receptor associated factor 2 Homo sapiens 270-275 23396089-8 2013 In addition, fenretinide inhibited the expression of the genes involved in RAS/RAF/ERK-mediated survival pathway. Fenretinide 13-24 zinc fingers and homeoboxes 2 Homo sapiens 79-82 23052481-3 2013 Fenretinide (4-HPR) is a synthetic retinoid that has shown clinical activity in recurrent NB and is cytotoxic to a variety of cancer cells, in part via the accumulation of dihydroceramides, which are precursors of GD2. Fenretinide 0-11 haptoglobin-related protein Homo sapiens 15-18 23513221-4 2013 Fenretinide exerted a selective cytotoxic effect on primary AML CD34(+) cells, especially the LSC-enriched CD34(+)CD38(-) subpopulation, whereas no significant effect was observed on normal counterparts. Fenretinide 0-11 CD34 molecule Homo sapiens 64-68 23513221-4 2013 Fenretinide exerted a selective cytotoxic effect on primary AML CD34(+) cells, especially the LSC-enriched CD34(+)CD38(-) subpopulation, whereas no significant effect was observed on normal counterparts. Fenretinide 0-11 CD34 molecule Homo sapiens 107-111 23513221-4 2013 Fenretinide exerted a selective cytotoxic effect on primary AML CD34(+) cells, especially the LSC-enriched CD34(+)CD38(-) subpopulation, whereas no significant effect was observed on normal counterparts. Fenretinide 0-11 CD38 molecule Homo sapiens 114-118 23513221-5 2013 Methylcellulose colony formation assays further showed that fenretinide significantly suppressed the formation of colonies derived from AML CD34(+) cells but not those from normal CD34(+) cells. Fenretinide 60-71 CD34 molecule Homo sapiens 140-144 23052481-5 2013 Here, we demonstrate for the first time that pretreatment of fenretinide-resistant NB cells with 4-HPR significantly enhanced ch14.18/CHO-mediated CDC and ADCC and AICC by both human natural killer cells and peripheral blood mononuclear cells. Fenretinide 61-72 haptoglobin-related protein Homo sapiens 99-102 23314735-4 2013 We, therefore, hypothesized that p75NTR expression level would influence the effects of the redox-active chemotherapeutic drug fenretinide on neuroblastoma cells. Fenretinide 127-138 nerve growth factor receptor Homo sapiens 33-39 23314735-10 2013 RESULTS: Knockdown of p75NTR attenuates fenretinide-induced accumulation of mitochondrial superoxide and apoptosis. Fenretinide 40-51 nerve growth factor receptor Homo sapiens 22-28 23314735-12 2013 Pretreatment of cells with 2-thenoyltrifluoroacetone or dehydroascorbic acid uniquely prevents mitochondrial superoxide accumulation and cell death after fenretinide treatment, indicating that mitochondrial complex II is the likely site of fenretinide-induced superoxide generation and p75NTR-induced potentiation of these phenomena. Fenretinide 154-165 nerve growth factor receptor Homo sapiens 286-292 23314735-12 2013 Pretreatment of cells with 2-thenoyltrifluoroacetone or dehydroascorbic acid uniquely prevents mitochondrial superoxide accumulation and cell death after fenretinide treatment, indicating that mitochondrial complex II is the likely site of fenretinide-induced superoxide generation and p75NTR-induced potentiation of these phenomena. Fenretinide 240-251 nerve growth factor receptor Homo sapiens 286-292 23314735-13 2013 CONCLUSION: Modification of expression of p75NTR in a particular neuroblastoma cell line modifies its susceptibility to fenretinide. Fenretinide 120-131 nerve growth factor receptor Homo sapiens 42-48 23023528-2 2013 Fenretinide, an orally available drug that reduces retinol delivery to the eye through antagonism of serum retinol-binding protein (RBP), was used in a 2-year trial to determine whether retinol reduction would be effective in the management of geographic atrophy. Fenretinide 0-11 retinol binding protein 4 Homo sapiens 132-135 23023528-4 2013 RESULTS: Fenretinide treatment produced dose-dependent reversible reductions in serum RBP-retinol that were associated with trends in reduced lesion growth rates. Fenretinide 9-20 retinol binding protein 4 Homo sapiens 86-89 23372815-8 2013 Moreover, fenretinide induced reactive oxygen species and apoptosis as shown by caspase 9 and PARP cleavage and upregulated miR-9. Fenretinide 10-21 caspase 9 Homo sapiens 80-89 23139430-4 2013 Moreover, fenretinide attenuated interleukin (IL)-1beta, IL-6, and cyclooxygenase-2 mRNA expression induced by A. actinomycetemcomitans. Fenretinide 10-21 interleukin 6 Mus musculus 57-61 23139430-5 2013 Fenretinide also decreased IL-1beta, IL-6, and prostaglandin E2 proinflammatory cytokine levels in Raw 264.7 cells induced by A. actinomycetemcomitans. Fenretinide 0-11 interleukin 1 beta Mus musculus 27-35 23139430-5 2013 Fenretinide also decreased IL-1beta, IL-6, and prostaglandin E2 proinflammatory cytokine levels in Raw 264.7 cells induced by A. actinomycetemcomitans. Fenretinide 0-11 interleukin 6 Mus musculus 37-41 23372815-8 2013 Moreover, fenretinide induced reactive oxygen species and apoptosis as shown by caspase 9 and PARP cleavage and upregulated miR-9. Fenretinide 10-21 collagen type XI alpha 2 chain Homo sapiens 94-98 22475870-2 2012 4HPR promoted the intrinsic apoptotic pathway by reactive oxygen species (ROS) generation and was accompanied by drop of Mcl-1 protein expression. Fenretinide 0-4 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 121-126 23326540-8 2013 Chronic intraperitoneal injection of fenretinide increased the alveolar airspace surface area and increased the number of caspase-3 positive cells in rat lungs. Fenretinide 37-48 caspase 3 Rattus norvegicus 122-131 23326540-9 2013 Fenretinide also suppressed HIF-1alpha and VEGF protein expression in rat lungs. Fenretinide 0-11 hypoxia inducible factor 1 subunit alpha Rattus norvegicus 28-38 23326540-9 2013 Fenretinide also suppressed HIF-1alpha and VEGF protein expression in rat lungs. Fenretinide 0-11 vascular endothelial growth factor A Rattus norvegicus 43-47 23326540-14 2013 Concomitant treatment with S1P normalizes the ceramide-S1P balance in the rat lungs and increases HIF-1alpha protein expression via activation of sphingosine kinase 1; as a consequence, S1P salvages fenretinide induced emphysema in rat lungs. Fenretinide 199-210 sphingosine kinase 1 Rattus norvegicus 146-166 22498172-2 2012 We explored efficacy of N-(4-hydroxyphenyl) retinamide (4-HPR) and (-)-epigallocatechin-3-gallate (EGCG) in altering expression of oncogenic microRNAs (OGmiRs) and tumor suppressor miRs (TSmiRs) for controlling growth of human malignant neuroblastoma SK-N-BE2 and IMR-32 cells. Fenretinide 24-54 haptoglobin-related protein Homo sapiens 58-61 22573912-7 2012 We observed that the synthetic retinoid fenretinide substituted for the retinol cofactor function but, on chronic use, distorted this signal balance, leading to predominance of PKCepsilon over PKCdelta. Fenretinide 40-51 protein kinase C epsilon Homo sapiens 177-187 22573912-7 2012 We observed that the synthetic retinoid fenretinide substituted for the retinol cofactor function but, on chronic use, distorted this signal balance, leading to predominance of PKCepsilon over PKCdelta. Fenretinide 40-51 protein kinase C delta Homo sapiens 193-201 22637576-7 2012 Finally, we show that the drug fenretinide, used clinically to presumably lower blood RBP levels and thus decrease circulating retinol, targets the functional coupling of STRA6 and LRAT to increase cellular vitamin A uptake in peripheral tissues. Fenretinide 31-42 retinol binding protein 4, plasma Mus musculus 86-89 22637576-7 2012 Finally, we show that the drug fenretinide, used clinically to presumably lower blood RBP levels and thus decrease circulating retinol, targets the functional coupling of STRA6 and LRAT to increase cellular vitamin A uptake in peripheral tissues. Fenretinide 31-42 stimulated by retinoic acid gene 6 Mus musculus 171-176 22474281-2 2012 Recent in vitro data indicate that fenretinide inhibits dihydroceramide desaturase, an enzyme involved in the biosynthesis of lipotoxic ceramides that antagonize insulin action. Fenretinide 35-46 delta(4)-desaturase, sphingolipid 1 Mus musculus 56-82 22474281-9 2012 These data suggest that fenretinide improves insulin sensitivity, at least in part, by inhibiting Des1 and suggest that therapeutics targeting this enzyme may be a viable therapeutic means for normalizing glucose homeostasis in the overweight and diabetic. Fenretinide 24-35 delta(4)-desaturase, sphingolipid 1 Mus musculus 98-102 22798378-0 2012 Identification of mammalian target of rapamycin as a direct target of fenretinide both in vitro and in vivo. Fenretinide 70-81 mechanistic target of rapamycin kinase Homo sapiens 18-47 22488409-0 2012 N-(4-hydroxyphenyl)retinamide inhibits breast cancer cell invasion through suppressing NF-KB activation and inhibiting matrix metalloproteinase-9 expression. Fenretinide 0-29 matrix metallopeptidase 9 Homo sapiens 119-145 22488409-1 2012 Synthetic retinoid N-(4-hydroxyphenyl)retinamide (4-HPR) has been reported to exhibit anti-invasive and anti-metastatic activities by suppressing the enzymatic activity of matrix metalloproteinase (MMP)-9, but the underlying mechanism remains unclear. Fenretinide 19-48 haptoglobin-related protein Homo sapiens 52-55 22488409-1 2012 Synthetic retinoid N-(4-hydroxyphenyl)retinamide (4-HPR) has been reported to exhibit anti-invasive and anti-metastatic activities by suppressing the enzymatic activity of matrix metalloproteinase (MMP)-9, but the underlying mechanism remains unclear. Fenretinide 19-48 matrix metallopeptidase 9 Homo sapiens 172-204 22532987-1 2012 Fenretinide (4-HPR) cytotoxicity relative to glutathione levels in pediatric acute lymphoblastic leukemia cell lines cultured at bone marrow level hypoxia (5% O2) is evaluated. Fenretinide 0-11 haptoglobin-related protein Homo sapiens 15-18 22427354-9 2012 Human oral mucosal correlative studies showed substantial interdonor variations in levels of the enzyme (cytochrome P450 3A4-CYP3A4) responsible for conversion of fenretinide to its highly active metabolite, 4-oxo-4-HPR. Fenretinide 163-174 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 105-131 22427354-9 2012 Human oral mucosal correlative studies showed substantial interdonor variations in levels of the enzyme (cytochrome P450 3A4-CYP3A4) responsible for conversion of fenretinide to its highly active metabolite, 4-oxo-4-HPR. Fenretinide 163-174 haptoglobin-related protein Homo sapiens 216-219 22427354-6 2012 LC-MS/MS analyses of post-treatment samples revealed a delivery gradient with highest fenretinide levels achieved at the patch-mucosal interface (no metabolites), pharmacologically active levels in fenretinide-treated oral mucosa (mean: 5.65 muM; trace amounts of 4-oxo-4-HPR) and undetectable sera levels. Fenretinide 198-209 haptoglobin-related protein Homo sapiens 272-275 22428532-1 2012 4-(Hydroxyphenyl)retinamide (4-HPR) is a synthetic retinoid with a strong apoptotic effect towards different cancer cell lines in vitro, and it is currently tested in clinical trials. Fenretinide 0-27 haptoglobin-related protein Homo sapiens 31-34 22427354-7 2012 Epithelial markers for cell proliferation (Ki-67), terminal differentiation (transglutaminase 1-TGase1) and glucuronidation (UDP-glucuronosyltransferase1A1-UGT1A1) exhibited fenretinide concentration-specific relationships (elevated TGase1 and UGT1A1 levels <5 muM, reduced Ki-67 indices >5 muM) relative to blank-treated epithelium. Fenretinide 174-185 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-155 22183330-0 2012 The mechanism of fenretinide (4-HPR) inhibition of beta-carotene monooxygenase 1. Fenretinide 17-28 haptoglobin-related protein Homo sapiens 32-35 22387538-0 2012 Combined treatment with fenretinide and indomethacin induces AIF-mediated, non-classical cell death in human acute T-cell leukemia Jurkat cells. Fenretinide 24-35 apoptosis inducing factor mitochondria associated 1 Homo sapiens 61-64 22387538-8 2012 Taken together these results indicate, that Jurkat cells after treatment with a combination of fenretinide and indomethacin undergo AIF-mediated programmed cell death. Fenretinide 95-106 apoptosis inducing factor mitochondria associated 1 Homo sapiens 132-135 22221098-2 2012 METHODS: We synthesised an amphiphilic dextrin derivative (DX-OL) able to host fenretinide (4-HPR) by complexation. Fenretinide 79-90 haptoglobin-related protein Homo sapiens 94-97 22162577-0 2012 Prognostic effect of circulating adiponectin in a randomized 2 x 2 trial of low-dose tamoxifen and fenretinide in premenopausal women at risk for breast cancer. Fenretinide 99-110 adiponectin, C1Q and collagen domain containing Homo sapiens 33-44 22350416-4 2012 In this study, we developed a mathematical model that integrates temporal patterns of drug exposure, receptor occupancy, and signal transduction to predict the effects of the CD20 agonist rituximab in combination with rhApo2L/TNF-related apoptosis inducing ligand or fenretinide, a cytotoxic retinoid, upon growth kinetics in non-Hodgkin lymphoma xenografts. Fenretinide 267-278 keratin 20 Homo sapiens 175-179 22382323-1 2012 Fenretinide (FEN), a ligand of retinol binding protein 4 (RBP4), has been suggested as a measure to reduce insulin resistance and its associated disorders such as obesity, and fatty liver by reducing serum RBP4. Fenretinide 0-11 retinol binding protein 4, plasma Mus musculus 31-56 22382323-1 2012 Fenretinide (FEN), a ligand of retinol binding protein 4 (RBP4), has been suggested as a measure to reduce insulin resistance and its associated disorders such as obesity, and fatty liver by reducing serum RBP4. Fenretinide 0-11 retinol binding protein 4, plasma Mus musculus 58-62 22382323-1 2012 Fenretinide (FEN), a ligand of retinol binding protein 4 (RBP4), has been suggested as a measure to reduce insulin resistance and its associated disorders such as obesity, and fatty liver by reducing serum RBP4. Fenretinide 0-11 retinol binding protein 4, plasma Mus musculus 206-210 22382323-1 2012 Fenretinide (FEN), a ligand of retinol binding protein 4 (RBP4), has been suggested as a measure to reduce insulin resistance and its associated disorders such as obesity, and fatty liver by reducing serum RBP4. Fenretinide 13-16 retinol binding protein 4, plasma Mus musculus 31-56 22382323-1 2012 Fenretinide (FEN), a ligand of retinol binding protein 4 (RBP4), has been suggested as a measure to reduce insulin resistance and its associated disorders such as obesity, and fatty liver by reducing serum RBP4. Fenretinide 13-16 retinol binding protein 4, plasma Mus musculus 58-62 22382323-1 2012 Fenretinide (FEN), a ligand of retinol binding protein 4 (RBP4), has been suggested as a measure to reduce insulin resistance and its associated disorders such as obesity, and fatty liver by reducing serum RBP4. Fenretinide 13-16 retinol binding protein 4, plasma Mus musculus 206-210 22382323-9 2012 The mechanism by which fenretinide prevents fatty liver may be explained by an increased plasma adiponectin level, increased activation of hepatic AMP-activated protein kinase, and the expression of peroxisome proliferator-activated protein-alpha and peroxisomal acyl-CoA oxidase, which promote fat oxidation. Fenretinide 23-34 adiponectin, C1Q and collagen domain containing Mus musculus 96-107 22500077-0 2012 Fenretinide (4-HPR): a preventive chance for women at genetic and familial risk? Fenretinide 0-11 haptoglobin-related protein Homo sapiens 15-18 22500077-4 2012 Fenretinide (4-HPR) is the most studied retinoid in breast cancer chemoprevention clinical trials due to its selective accumulation in breast tissue and its favorable toxicological profile. Fenretinide 0-11 haptoglobin-related protein Homo sapiens 15-18 22761939-1 2012 BACKGROUND: Fenretinide (4-HPR) is a synthetic retinoid that exhibits potent antitumor and chemopreventive activities against different malignancies, including ovarian tumors. Fenretinide 12-23 haptoglobin-related protein Homo sapiens 27-30 21964808-2 2012 A retinoid analogue, fenretinide [N-(4-hydroxyphenyl) retinamide; 4-HPR] can induce apoptosis in neuroblastoma cell lines and could have clinical use after therapy with 13cisRA. Fenretinide 21-32 haptoglobin-related protein Homo sapiens 80-83 21964808-2 2012 A retinoid analogue, fenretinide [N-(4-hydroxyphenyl) retinamide; 4-HPR] can induce apoptosis in neuroblastoma cell lines and could have clinical use after therapy with 13cisRA. Fenretinide 34-64 haptoglobin-related protein Homo sapiens 80-83 21908574-0 2011 Phase II study of oral capsular 4-hydroxyphenylretinamide (4-HPR/fenretinide) in pediatric patients with refractory or recurrent neuroblastoma: a report from the Children"s Oncology Group. Fenretinide 32-57 haptoglobin-related protein Homo sapiens 61-64 21933888-1 2011 PURPOSE: Fenretinide (4-HPR) is a cytotoxic retinoid with minimal systemic toxicity that has shown clinical activity against recurrent high-risk neuroblastoma. Fenretinide 9-20 haptoglobin-related protein Homo sapiens 24-27 22061047-1 2011 BACKGROUND: N-(4-hydroxyphenyl)retinamide (4-HPR, fenretinide) is a synthetic retinoid with potent pro-apoptotic activity against several types of cancer, but little is known regarding mechanisms leading to chemoresistance. Fenretinide 12-41 haptoglobin-related protein Homo sapiens 45-48 22061047-1 2011 BACKGROUND: N-(4-hydroxyphenyl)retinamide (4-HPR, fenretinide) is a synthetic retinoid with potent pro-apoptotic activity against several types of cancer, but little is known regarding mechanisms leading to chemoresistance. Fenretinide 50-61 haptoglobin-related protein Homo sapiens 45-48 21557271-0 2011 Inhibition of acid ceramidase by a 2-substituted aminoethanol amide synergistically sensitizes prostate cancer cells to N-(4-hydroxyphenyl) retinamide. Fenretinide 120-150 N-acylsphingosine amidohydrolase 1 Homo sapiens 14-29 21622882-1 2011 Apoptosis via the intrinsic caspase 9 pathway can be induced by oxidative stressors hydrogen peroxide (H2O2) and N-(4 hydroxyphenol) rentinamide (fenretinide), a synthetic retinoid. Fenretinide 146-157 caspase-9 Ovis aries 28-37 21951911-2 2011 N-(4-hydroxy phenyl) retinamide (4-HPR or fenretinide) is a potential chemotherapeutic for RTs with activity correlated to its ability to down-modulate Cyclin D1. Fenretinide 0-31 haptoglobin-related protein Homo sapiens 35-38 21951911-2 2011 N-(4-hydroxy phenyl) retinamide (4-HPR or fenretinide) is a potential chemotherapeutic for RTs with activity correlated to its ability to down-modulate Cyclin D1. Fenretinide 0-31 cyclin D1 Homo sapiens 152-161 21951911-2 2011 N-(4-hydroxy phenyl) retinamide (4-HPR or fenretinide) is a potential chemotherapeutic for RTs with activity correlated to its ability to down-modulate Cyclin D1. Fenretinide 42-53 haptoglobin-related protein Homo sapiens 35-38 21951911-2 2011 N-(4-hydroxy phenyl) retinamide (4-HPR or fenretinide) is a potential chemotherapeutic for RTs with activity correlated to its ability to down-modulate Cyclin D1. Fenretinide 42-53 cyclin D1 Homo sapiens 152-161 21072519-1 2011 PURPOSE: Although fenretinide (4-HPR) has been studied in breast cancer and in neuroblastoma, little is known regarding its activity in pancreatic cancer, a neoplasm for which there are few therapeutic options. Fenretinide 18-29 haptoglobin-related protein Homo sapiens 33-36 21543327-2 2011 We previously demonstrated that fenretinide (4-HPR) inhibited DES activity in SMS-KCNR neuroblastoma cells. Fenretinide 32-43 haptoglobin-related protein Homo sapiens 47-50 21591606-0 2011 Fenretinide derivatives act as disrupters of interactions of serum retinol binding protein (sRBP) with transthyretin and the sRBP receptor. Fenretinide 0-11 sigma non-opioid intracellular receptor 1 Homo sapiens 61-90 21591606-0 2011 Fenretinide derivatives act as disrupters of interactions of serum retinol binding protein (sRBP) with transthyretin and the sRBP receptor. Fenretinide 0-11 sigma non-opioid intracellular receptor 1 Homo sapiens 92-96 21591606-0 2011 Fenretinide derivatives act as disrupters of interactions of serum retinol binding protein (sRBP) with transthyretin and the sRBP receptor. Fenretinide 0-11 transthyretin Homo sapiens 103-116 21591606-0 2011 Fenretinide derivatives act as disrupters of interactions of serum retinol binding protein (sRBP) with transthyretin and the sRBP receptor. Fenretinide 0-11 sigma non-opioid intracellular receptor 1 Homo sapiens 125-129 21591606-5 2011 Shortening the chain length of the FEN derivative substantially abolished binding to sRBP, indicating that the strength of the interaction lies in the polyene chain region. Fenretinide 35-38 sigma non-opioid intracellular receptor 1 Homo sapiens 85-89 21391977-8 2011 4-HPR was oxidized to 4-oxo-4-HPR, at least in part via human CYP3A4. Fenretinide 0-5 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 62-68 21723035-0 2011 TMEM14A inhibits N-(4-hydroxyphenyl)retinamide-induced apoptosis through the stabilization of mitochondrial membrane potential. Fenretinide 17-46 transmembrane protein 14A Homo sapiens 0-7 21723035-5 2011 Over-expression of TMEM14A in U87MG cells inhibited N-(4-hydroxyphenyl)retinamide (4-HPR)-induced apoptosis. Fenretinide 52-81 transmembrane protein 14A Homo sapiens 19-26 21723035-5 2011 Over-expression of TMEM14A in U87MG cells inhibited N-(4-hydroxyphenyl)retinamide (4-HPR)-induced apoptosis. Fenretinide 52-81 haptoglobin-related protein Homo sapiens 85-88 21557271-1 2011 BACKGROUND: The purpose of this study was to determine whether the therapeutic efficacy of fenretinide (4-HPR), a ceramide-generating anticancer agent, could be enhanced in prostate cancer cells by inclusion of a novel synthetic acid ceramidase (AC) inhibitor, DM102, a pivaloylamide of a 2-substituted aminoethanol. Fenretinide 91-102 haptoglobin-related protein Homo sapiens 106-109 21557271-1 2011 BACKGROUND: The purpose of this study was to determine whether the therapeutic efficacy of fenretinide (4-HPR), a ceramide-generating anticancer agent, could be enhanced in prostate cancer cells by inclusion of a novel synthetic acid ceramidase (AC) inhibitor, DM102, a pivaloylamide of a 2-substituted aminoethanol. Fenretinide 91-102 N-acylsphingosine amidohydrolase 1 Homo sapiens 229-244 21557271-1 2011 BACKGROUND: The purpose of this study was to determine whether the therapeutic efficacy of fenretinide (4-HPR), a ceramide-generating anticancer agent, could be enhanced in prostate cancer cells by inclusion of a novel synthetic acid ceramidase (AC) inhibitor, DM102, a pivaloylamide of a 2-substituted aminoethanol. Fenretinide 91-102 N-acylsphingosine amidohydrolase 1 Homo sapiens 1-3 22468231-4 2011 We evaluated synergistic efficacy of N-(4-hydroxyphenyl) retinamide (4-HPR) and genistein (GST) for induction of apoptosis in human malignant neuroblastoma SH-SY5Y and SK-N-BE2 cells in culture and activation of multiple pathways for increasing apoptosis in ectopic and orthotopic neuroblastoma xenografts in nude mice. Fenretinide 37-67 haptoglobin-related protein Homo sapiens 71-74 21365528-5 2011 Administration of fenretinide in SHR to increase urinary RBP4 excretion significantly decreased plasma RBP4 levels and improved IR. Fenretinide 18-29 retinol binding protein 4 Rattus norvegicus 57-61 21365528-5 2011 Administration of fenretinide in SHR to increase urinary RBP4 excretion significantly decreased plasma RBP4 levels and improved IR. Fenretinide 18-29 retinol binding protein 4 Rattus norvegicus 103-107 21270111-4 2011 RESULTS: Autophagy was activated in response to fenretinide or bortezomib in B-RAF wild-type cells, shown by increased conversion of LC3 to the autophagic vesicle-associated form (LC3-II) and redistribution to autophagosomes and autolysosomes, increased acidic vesicular organelle formation and autophagic vacuolization. Fenretinide 48-59 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 77-82 21270111-4 2011 RESULTS: Autophagy was activated in response to fenretinide or bortezomib in B-RAF wild-type cells, shown by increased conversion of LC3 to the autophagic vesicle-associated form (LC3-II) and redistribution to autophagosomes and autolysosomes, increased acidic vesicular organelle formation and autophagic vacuolization. Fenretinide 48-59 microtubule associated protein 1 light chain 3 alpha Homo sapiens 133-136 21270111-4 2011 RESULTS: Autophagy was activated in response to fenretinide or bortezomib in B-RAF wild-type cells, shown by increased conversion of LC3 to the autophagic vesicle-associated form (LC3-II) and redistribution to autophagosomes and autolysosomes, increased acidic vesicular organelle formation and autophagic vacuolization. Fenretinide 48-59 microtubule associated protein 1 light chain 3 alpha Homo sapiens 180-183 21270111-7 2011 Knockdown of Beclin-1 or ATG7 sensitized B-RAF wild-type cells to fenretinide- or bortezomib-induced cell death, demonstrating a pro-survival function of autophagy. Fenretinide 66-77 beclin 1 Homo sapiens 13-21 21270111-7 2011 Knockdown of Beclin-1 or ATG7 sensitized B-RAF wild-type cells to fenretinide- or bortezomib-induced cell death, demonstrating a pro-survival function of autophagy. Fenretinide 66-77 autophagy related 7 Homo sapiens 25-29 21270111-7 2011 Knockdown of Beclin-1 or ATG7 sensitized B-RAF wild-type cells to fenretinide- or bortezomib-induced cell death, demonstrating a pro-survival function of autophagy. Fenretinide 66-77 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 41-46 21270111-8 2011 In addition, autophagy was partially reactivated in B-RAF-mutated cells treated with the BH3 mimetic ABT737 in combination with fenretinide or bortezomib, suggesting autophagy resistance is partly mediated by abrogated Beclin-1 function. Fenretinide 128-139 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 52-57 21270111-8 2011 In addition, autophagy was partially reactivated in B-RAF-mutated cells treated with the BH3 mimetic ABT737 in combination with fenretinide or bortezomib, suggesting autophagy resistance is partly mediated by abrogated Beclin-1 function. Fenretinide 128-139 beclin 1 Homo sapiens 219-227 21241664-0 2011 ERK1/2 deactivation enhances cytoplasmic Nur77 expression level and improves the apoptotic effect of fenretinide in human liver cancer cells. Fenretinide 101-112 mitogen-activated protein kinase 3 Homo sapiens 0-6 21241664-3 2011 Previously, we have shown that fenretinide-induced apoptosis is Nur77 dependent, and the sensitivity of the cancer cells to fenretinide-induced apoptosis is positively associated with cytoplasmic enrichment of Nur77. Fenretinide 31-42 nuclear receptor subfamily 4 group A member 1 Homo sapiens 64-69 21241664-3 2011 Previously, we have shown that fenretinide-induced apoptosis is Nur77 dependent, and the sensitivity of the cancer cells to fenretinide-induced apoptosis is positively associated with cytoplasmic enrichment of Nur77. Fenretinide 31-42 nuclear receptor subfamily 4 group A member 1 Homo sapiens 210-215 21241664-3 2011 Previously, we have shown that fenretinide-induced apoptosis is Nur77 dependent, and the sensitivity of the cancer cells to fenretinide-induced apoptosis is positively associated with cytoplasmic enrichment of Nur77. Fenretinide 124-135 nuclear receptor subfamily 4 group A member 1 Homo sapiens 210-215 21241664-4 2011 The goal of current study was to identify means to modulate nuclear export of Nur77 in order to improve the efficacy of fenretinide. Fenretinide 120-131 nuclear receptor subfamily 4 group A member 1 Homo sapiens 78-83 21241664-7 2011 In fenretinide sensitive Huh7 cells, activation of ERK1/2 by epidermal growth factor (EGF) prevented fenretinide-induced cell death and caspase 3/7 induction. Fenretinide 3-14 epidermal growth factor Homo sapiens 61-84 21241664-5 2011 Fenretinide treatment deactivated ERK1/2 in Huh7 cells, but activated ERK1/2 in HepG2 cells, which was positively associated with the sensitivity of cells to the apoptotic effect of fenretinide. Fenretinide 0-11 mitogen-activated protein kinase 3 Homo sapiens 34-40 21241664-7 2011 In fenretinide sensitive Huh7 cells, activation of ERK1/2 by epidermal growth factor (EGF) prevented fenretinide-induced cell death and caspase 3/7 induction. Fenretinide 3-14 epidermal growth factor Homo sapiens 86-89 21241664-5 2011 Fenretinide treatment deactivated ERK1/2 in Huh7 cells, but activated ERK1/2 in HepG2 cells, which was positively associated with the sensitivity of cells to the apoptotic effect of fenretinide. Fenretinide 0-11 mitogen-activated protein kinase 3 Homo sapiens 70-76 21241664-5 2011 Fenretinide treatment deactivated ERK1/2 in Huh7 cells, but activated ERK1/2 in HepG2 cells, which was positively associated with the sensitivity of cells to the apoptotic effect of fenretinide. Fenretinide 182-193 mitogen-activated protein kinase 3 Homo sapiens 70-76 21241664-7 2011 In fenretinide sensitive Huh7 cells, activation of ERK1/2 by epidermal growth factor (EGF) prevented fenretinide-induced cell death and caspase 3/7 induction. Fenretinide 3-14 caspase 3 Homo sapiens 136-145 21241664-7 2011 In fenretinide sensitive Huh7 cells, activation of ERK1/2 by epidermal growth factor (EGF) prevented fenretinide-induced cell death and caspase 3/7 induction. Fenretinide 101-112 mitogen-activated protein kinase 3 Homo sapiens 51-57 21241664-7 2011 In fenretinide sensitive Huh7 cells, activation of ERK1/2 by epidermal growth factor (EGF) prevented fenretinide-induced cell death and caspase 3/7 induction. Fenretinide 101-112 epidermal growth factor Homo sapiens 61-84 21241664-7 2011 In fenretinide sensitive Huh7 cells, activation of ERK1/2 by epidermal growth factor (EGF) prevented fenretinide-induced cell death and caspase 3/7 induction. Fenretinide 101-112 epidermal growth factor Homo sapiens 86-89 21241664-9 2011 Fenretinide/PD98059-induced cell death of HepG2 cell was positively associated with induction and cytoplasmic location as well as mitochondria enrichment of Nur77. Fenretinide 0-11 nuclear receptor subfamily 4 group A member 1 Homo sapiens 157-162 21241664-7 2011 In fenretinide sensitive Huh7 cells, activation of ERK1/2 by epidermal growth factor (EGF) prevented fenretinide-induced cell death and caspase 3/7 induction. Fenretinide 3-14 mitogen-activated protein kinase 3 Homo sapiens 51-57 21241664-11 2011 Taken together, the current study demonstrates that ERK1/2-modulated Nur77 intracellular location dictates the efficacy of fenretinide-induced apoptosis. Fenretinide 123-134 mitogen-activated protein kinase 3 Homo sapiens 52-58 21498721-1 2011 BACKGROUND: The synthetic retinoid fenretinide (N-(4-hydroxyphenyl)retinamide, 4-HPR) has shown promising anticancer activity in preclinical studies, but its limited oral bioavailability has hindered clinical assessment. Fenretinide 35-46 haptoglobin-related protein Homo sapiens 81-84 21241664-11 2011 Taken together, the current study demonstrates that ERK1/2-modulated Nur77 intracellular location dictates the efficacy of fenretinide-induced apoptosis. Fenretinide 123-134 nuclear receptor subfamily 4 group A member 1 Homo sapiens 69-74 21822457-3 2011 We examined for the first time the efficacy of N-(4-hydroxyphenyl) retinamide (4-HPR) and genistein (GST) alone and also in combination for controlling growth of human Ewing"s sarcoma SK-N-MC and RD-ES xenografts. Fenretinide 47-77 haptoglobin-related protein Homo sapiens 81-84 21498721-1 2011 BACKGROUND: The synthetic retinoid fenretinide (N-(4-hydroxyphenyl)retinamide, 4-HPR) has shown promising anticancer activity in preclinical studies, but its limited oral bioavailability has hindered clinical assessment. Fenretinide 48-77 haptoglobin-related protein Homo sapiens 81-84 21738400-9 2011 Fenretinide induced the expression of the cone long wavelength sensitive opsin (OPN1lw) but not rhodopsin (RHO), while decreasing the expression of RPE cell markers. Fenretinide 0-11 opsin 1, long wave sensitive Homo sapiens 80-86 21319187-0 2011 Enrichment of Nur77 mediated by retinoic acid receptor beta leads to apoptosis of human hepatocellular carcinoma cells induced by fenretinide and histone deacetylase inhibitors. Fenretinide 130-141 nuclear receptor subfamily 4 group A member 1 Homo sapiens 14-19 21319187-0 2011 Enrichment of Nur77 mediated by retinoic acid receptor beta leads to apoptosis of human hepatocellular carcinoma cells induced by fenretinide and histone deacetylase inhibitors. Fenretinide 130-141 retinoic acid receptor beta Homo sapiens 32-59 21319187-2 2011 Previously, we have shown that fenretinide induces apoptosis of Huh7 cells, but HepG2 cells are relatively resistant to fenretinide-induced apoptosis. Fenretinide 31-42 MIR7-3 host gene Homo sapiens 64-68 21319187-4 2011 Trichostatin A and scriptaid can either enhance fenretinide-induced apoptosis in the fenretinide sensitive HCC cells (Huh7 and Hep3B) or sensitize the fenretinide resistant cells (HepG2) to become sensitive to the apoptotic effect of fenretinide in a cancer cell-specific manner. Fenretinide 48-59 MIR7-3 host gene Homo sapiens 118-122 21319187-7 2011 Upon fenretinide and HDACi treatment, the expression of RARbeta and Nur77 were induced and colocalized in the cytosol. Fenretinide 5-16 retinoic acid receptor beta Homo sapiens 56-63 21319187-7 2011 Upon fenretinide and HDACi treatment, the expression of RARbeta and Nur77 were induced and colocalized in the cytosol. Fenretinide 5-16 nuclear receptor subfamily 4 group A member 1 Homo sapiens 68-73 21319187-10 2011 Nur77 was essential for fenretinide-induced and HDACi-induced apoptosis of Huh7 cells. Fenretinide 24-35 nuclear receptor subfamily 4 group A member 1 Homo sapiens 0-5 21319187-10 2011 Nur77 was essential for fenretinide-induced and HDACi-induced apoptosis of Huh7 cells. Fenretinide 24-35 MIR7-3 host gene Homo sapiens 75-79 21319187-11 2011 Induction of the expression, the interaction, and the nuclear export of RARbeta and Nur77 mediate fenretinide-induced and HDACi-induced apoptosis. Fenretinide 98-109 retinoic acid receptor beta Homo sapiens 72-79 21319187-11 2011 Induction of the expression, the interaction, and the nuclear export of RARbeta and Nur77 mediate fenretinide-induced and HDACi-induced apoptosis. Fenretinide 98-109 nuclear receptor subfamily 4 group A member 1 Homo sapiens 84-89 21119025-3 2011 Infection of RCCs with a tropism-modified serotype 5/3 adenovirus expressing MDA-7/IL-24 (Ad.5/3-mda-7) caused plasma membrane clustering of CD95 and CD95 association with pro-caspase 8, effects that were enhanced by combined exposure to 17-N-allylamino-17-demethoxygeldanamycin (17AAG), As(2)O(3), or fenretinide and that correlated with enhanced cell killing. Fenretinide 302-313 interleukin 24 Homo sapiens 77-82 21119025-3 2011 Infection of RCCs with a tropism-modified serotype 5/3 adenovirus expressing MDA-7/IL-24 (Ad.5/3-mda-7) caused plasma membrane clustering of CD95 and CD95 association with pro-caspase 8, effects that were enhanced by combined exposure to 17-N-allylamino-17-demethoxygeldanamycin (17AAG), As(2)O(3), or fenretinide and that correlated with enhanced cell killing. Fenretinide 302-313 interleukin 24 Homo sapiens 83-88 21119025-3 2011 Infection of RCCs with a tropism-modified serotype 5/3 adenovirus expressing MDA-7/IL-24 (Ad.5/3-mda-7) caused plasma membrane clustering of CD95 and CD95 association with pro-caspase 8, effects that were enhanced by combined exposure to 17-N-allylamino-17-demethoxygeldanamycin (17AAG), As(2)O(3), or fenretinide and that correlated with enhanced cell killing. Fenretinide 302-313 interleukin 24 Homo sapiens 97-102 21119025-3 2011 Infection of RCCs with a tropism-modified serotype 5/3 adenovirus expressing MDA-7/IL-24 (Ad.5/3-mda-7) caused plasma membrane clustering of CD95 and CD95 association with pro-caspase 8, effects that were enhanced by combined exposure to 17-N-allylamino-17-demethoxygeldanamycin (17AAG), As(2)O(3), or fenretinide and that correlated with enhanced cell killing. Fenretinide 302-313 Fas cell surface death receptor Homo sapiens 141-145 21054342-1 2011 BACKGROUND AND PURPOSE: Fenretinide (4-HPR) is a retinoic acid analogue, currently used in clinical trials in oncology. Fenretinide 24-35 haptoglobin-related protein Homo sapiens 39-42 20679253-2 2010 Treatment of cancer cells with N-(4-hydroxyphenyl) retinamide (4-HPR) induces apoptosis through destabilization of mitochondrial membrane and activation of caspase-mediated apoptotic pathways. Fenretinide 31-61 haptoglobin-related protein Homo sapiens 65-68 22174837-6 2011 RNAi-mediated knockdown of key GSH regulatory enzymes gamma-glutamylcysteine synthetase or glutathione disulfide reductase partially reversed the hypoxia-induced resistance to fenretinide, and increasing GSH levels using N-acetylcysteine augmented the hypoxia-induced resistance in a cell line-specific manner. Fenretinide 176-187 glutamate-cysteine ligase catalytic subunit Homo sapiens 54-87 20935456-2 2010 Many anticancer chemotherapeutics (anthracyclines, Vinca alkaloids, paclitaxel, and fenretinide), as well as physiological stimuli such as tumor necrosis factor alpha (TNFalpha), stimulate ceramide accumulation and increase oxidative stress in malignant cells. Fenretinide 84-95 tumor necrosis factor Homo sapiens 139-166 20532509-1 2010 PURPOSE: Fenretinide (4-HPR), a synthetic retinoid currently used in clinic for cancer therapy and prevention, markedly lowers plasma retinol levels, an effect associated with nyctalopia. Fenretinide 9-20 haptoglobin-related protein Homo sapiens 24-27 21117610-1 2010 Intravenous fenretinide (4-HPR), a cytotoxic retinoid, is being evaluated as part of a phase I clinical trial for patients with hematologic malignancies. Fenretinide 12-23 haptoglobin-related protein Homo sapiens 27-30 20976277-1 2010 BACKGROUND: The retinoid 4-oxo-N-(4-hydroxyphenyl)retinamide (4-oxo-4-HPR) is a polar metabolite of fenretinide (4-HPR) very effective in killing cancer cells of different histotypes, able to inhibit 4-HPR-resistant cell growth and to act synergistically in combination with the parent drug. Fenretinide 100-111 haptoglobin-related protein Homo sapiens 70-73 20976277-1 2010 BACKGROUND: The retinoid 4-oxo-N-(4-hydroxyphenyl)retinamide (4-oxo-4-HPR) is a polar metabolite of fenretinide (4-HPR) very effective in killing cancer cells of different histotypes, able to inhibit 4-HPR-resistant cell growth and to act synergistically in combination with the parent drug. Fenretinide 100-111 haptoglobin-related protein Homo sapiens 115-118 20976277-1 2010 BACKGROUND: The retinoid 4-oxo-N-(4-hydroxyphenyl)retinamide (4-oxo-4-HPR) is a polar metabolite of fenretinide (4-HPR) very effective in killing cancer cells of different histotypes, able to inhibit 4-HPR-resistant cell growth and to act synergistically in combination with the parent drug. Fenretinide 100-111 haptoglobin-related protein Homo sapiens 115-118 20637870-0 2010 HIF-1alpha-dependent autophagy protects HeLa cells from fenretinide (4-HPR)-induced apoptosis in hypoxia. Fenretinide 56-67 hypoxia inducible factor 1 subunit alpha Homo sapiens 0-10 20637870-0 2010 HIF-1alpha-dependent autophagy protects HeLa cells from fenretinide (4-HPR)-induced apoptosis in hypoxia. Fenretinide 56-67 haptoglobin-related protein Homo sapiens 71-74 20637870-2 2010 N-(4-Hydroxyphenyl)retinamide (4-HPR) exhibits potent anticancer and chemopreventive activities, but its inefficiency under hypoxia, through undetermined mechanisms, may contribute to its lack of activity in clinical trials. Fenretinide 0-29 haptoglobin-related protein Homo sapiens 33-36 20877355-0 2010 Fenretinide-dependent upregulation of death receptors through ASK1 and p38alpha enhances death receptor ligand-induced cell death in Ewing"s sarcoma family of tumours. Fenretinide 0-11 mitogen-activated protein kinase kinase kinase 5 Homo sapiens 62-66 20877355-0 2010 Fenretinide-dependent upregulation of death receptors through ASK1 and p38alpha enhances death receptor ligand-induced cell death in Ewing"s sarcoma family of tumours. Fenretinide 0-11 mitogen-activated protein kinase 14 Homo sapiens 71-79 20877355-2 2010 As fenretinide induces ESFT death through sustained p38(MAPK) phosphorylation, we hypothesised that this may be effected through upregulation of death receptors (DRs) and that treatment of fenretinide plus DR ligands may enhance apoptosis. Fenretinide 3-14 mitogen-activated protein kinase 14 Homo sapiens 52-55 20877355-5 2010 RESULTS: Fenretinide upregulated cell surface expression of tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) receptors, FAS and p75(NTR), in an ASK1- and p38alpha-dependent manner. Fenretinide 9-20 TNF superfamily member 10 Homo sapiens 60-116 20877355-5 2010 RESULTS: Fenretinide upregulated cell surface expression of tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) receptors, FAS and p75(NTR), in an ASK1- and p38alpha-dependent manner. Fenretinide 9-20 TNF superfamily member 10 Homo sapiens 118-123 20877355-5 2010 RESULTS: Fenretinide upregulated cell surface expression of tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) receptors, FAS and p75(NTR), in an ASK1- and p38alpha-dependent manner. Fenretinide 9-20 neurotensin receptor 1 Homo sapiens 144-152 20877355-5 2010 RESULTS: Fenretinide upregulated cell surface expression of tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) receptors, FAS and p75(NTR), in an ASK1- and p38alpha-dependent manner. Fenretinide 9-20 mitogen-activated protein kinase kinase kinase 5 Homo sapiens 160-164 20877355-5 2010 RESULTS: Fenretinide upregulated cell surface expression of tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) receptors, FAS and p75(NTR), in an ASK1- and p38alpha-dependent manner. Fenretinide 9-20 mitogen-activated protein kinase 14 Homo sapiens 170-178 20877355-6 2010 Cotreatment with fenretinide and DR ligands resulted in synergistic death compared with either agent alone; caspase-8 and Bid were cleaved in a time-dependent manner. Fenretinide 17-28 caspase 8 Homo sapiens 108-117 20877355-6 2010 Cotreatment with fenretinide and DR ligands resulted in synergistic death compared with either agent alone; caspase-8 and Bid were cleaved in a time-dependent manner. Fenretinide 17-28 BH3 interacting domain death agonist Homo sapiens 122-125 20687958-0 2010 Brg-1 mediates the constitutive and fenretinide-induced expression of SPARC in mammary carcinoma cells via its interaction with transcription factor Sp1. Fenretinide 36-47 SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily a, member 4 Homo sapiens 0-5 20628055-0 2010 Alkaline ceramidase 2 (ACER2) and its product dihydrosphingosine mediate the cytotoxicity of N-(4-hydroxyphenyl)retinamide in tumor cells. Fenretinide 93-122 alkaline ceramidase 2 Homo sapiens 0-21 20628055-0 2010 Alkaline ceramidase 2 (ACER2) and its product dihydrosphingosine mediate the cytotoxicity of N-(4-hydroxyphenyl)retinamide in tumor cells. Fenretinide 93-122 alkaline ceramidase 2 Homo sapiens 23-28 20628055-1 2010 Increased generation of dihydrosphingosine (DHS), a bioactive sphingolipid, has been implicated in the cytotoxicity of the synthetic retinoid N-(4-hydroxyphenyl)retinamide (4-HPR) in tumor cells. Fenretinide 142-171 haptoglobin-related protein Homo sapiens 175-178 20583135-0 2010 Decreased expression of insulin-like growth factor binding protein-5 during N-(4-hydroxyphenyl)retinamide-induced neuronal differentiation of ARPE-19 human retinal pigment epithelial cells: regulation by CCAAT/enhancer-binding protein. Fenretinide 76-105 insulin like growth factor binding protein 5 Homo sapiens 24-68 20583135-2 2010 We identified IGFBP5 by microarray analysis as a gene differentially regulated during N-(4-hydroxyphenyl)retinamide (4HPR)-induced neuronal differentiation of human retinal pigment epithelial (RPE) cells. Fenretinide 86-115 insulin like growth factor binding protein 5 Homo sapiens 14-20 20583135-2 2010 We identified IGFBP5 by microarray analysis as a gene differentially regulated during N-(4-hydroxyphenyl)retinamide (4HPR)-induced neuronal differentiation of human retinal pigment epithelial (RPE) cells. Fenretinide 117-121 insulin like growth factor binding protein 5 Homo sapiens 14-20 20520630-4 2010 Short hairpin RNA interference of XIAP was used to analyze the effect of XIAP expression on ER stress-induced apoptosis in response to fenretinide or bortezomib in vitro. Fenretinide 135-146 X-linked inhibitor of apoptosis Homo sapiens 73-77 20520630-7 2010 The correlation of XIAP expression with disease stage, as well as data showing that XIAP knockdown significantly increases fenretinide and bortezomib-induced apoptosis of metastatic melanoma cells, suggests that XIAP may prove to be an effective therapeutic target for melanoma therapy. Fenretinide 123-134 X-linked inhibitor of apoptosis Homo sapiens 84-88 20520630-7 2010 The correlation of XIAP expression with disease stage, as well as data showing that XIAP knockdown significantly increases fenretinide and bortezomib-induced apoptosis of metastatic melanoma cells, suggests that XIAP may prove to be an effective therapeutic target for melanoma therapy. Fenretinide 123-134 X-linked inhibitor of apoptosis Homo sapiens 84-88 20546536-4 2010 Down-regulation of GRP78 by small-interfering RNA increased fenretinide- or bortezomib-induced apoptosis. Fenretinide 60-71 heat shock protein family A (Hsp70) member 5 Homo sapiens 19-24 20546536-5 2010 Treatment of cells with a GRP78-specific subtilase toxin produced a synergistic enhancement with fenretinide or bortezomib. Fenretinide 97-108 heat shock protein family A (Hsp70) member 5 Homo sapiens 26-31 20546536-5 2010 Treatment of cells with a GRP78-specific subtilase toxin produced a synergistic enhancement with fenretinide or bortezomib. Fenretinide 97-108 proprotein convertase subtilisin/kexin type 5 Homo sapiens 41-50 20687958-9 2010 CONCLUSIONS: Overall, our results reveal a novel cooperative role of Brg-1 and Sp1 in mediating the constitutive and fenretinide-induced expression of SPARC, and provide new insights for the understanding of the anti-cancer effects of fenretinide. Fenretinide 117-128 SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily a, member 4 Homo sapiens 69-74 20687958-9 2010 CONCLUSIONS: Overall, our results reveal a novel cooperative role of Brg-1 and Sp1 in mediating the constitutive and fenretinide-induced expression of SPARC, and provide new insights for the understanding of the anti-cancer effects of fenretinide. Fenretinide 117-128 secreted protein acidic and cysteine rich Homo sapiens 151-156 20687958-9 2010 CONCLUSIONS: Overall, our results reveal a novel cooperative role of Brg-1 and Sp1 in mediating the constitutive and fenretinide-induced expression of SPARC, and provide new insights for the understanding of the anti-cancer effects of fenretinide. Fenretinide 235-246 SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily a, member 4 Homo sapiens 69-74 20687958-9 2010 CONCLUSIONS: Overall, our results reveal a novel cooperative role of Brg-1 and Sp1 in mediating the constitutive and fenretinide-induced expression of SPARC, and provide new insights for the understanding of the anti-cancer effects of fenretinide. Fenretinide 235-246 secreted protein acidic and cysteine rich Homo sapiens 151-156 20687958-0 2010 Brg-1 mediates the constitutive and fenretinide-induced expression of SPARC in mammary carcinoma cells via its interaction with transcription factor Sp1. Fenretinide 36-47 secreted protein acidic and cysteine rich Homo sapiens 70-75 20687958-7 2010 Interestingly, fenretinide [N-4(hydroxyphenyl) retinamide, 4-HPR], a synthetic retinoid with anti-cancer properties, was found to up-regulate the transcription, expression and secretion of SPARC via induction of the Brg-1 in a dose-dependent manner. Fenretinide 15-26 haptoglobin-related protein Homo sapiens 61-64 20687958-7 2010 Interestingly, fenretinide [N-4(hydroxyphenyl) retinamide, 4-HPR], a synthetic retinoid with anti-cancer properties, was found to up-regulate the transcription, expression and secretion of SPARC via induction of the Brg-1 in a dose-dependent manner. Fenretinide 15-26 secreted protein acidic and cysteine rich Homo sapiens 189-194 20687958-7 2010 Interestingly, fenretinide [N-4(hydroxyphenyl) retinamide, 4-HPR], a synthetic retinoid with anti-cancer properties, was found to up-regulate the transcription, expression and secretion of SPARC via induction of the Brg-1 in a dose-dependent manner. Fenretinide 15-26 SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily a, member 4 Homo sapiens 216-221 20687958-7 2010 Interestingly, fenretinide [N-4(hydroxyphenyl) retinamide, 4-HPR], a synthetic retinoid with anti-cancer properties, was found to up-regulate the transcription, expression and secretion of SPARC via induction of the Brg-1 in a dose-dependent manner. Fenretinide 28-57 haptoglobin-related protein Homo sapiens 61-64 20687958-7 2010 Interestingly, fenretinide [N-4(hydroxyphenyl) retinamide, 4-HPR], a synthetic retinoid with anti-cancer properties, was found to up-regulate the transcription, expression and secretion of SPARC via induction of the Brg-1 in a dose-dependent manner. Fenretinide 28-57 secreted protein acidic and cysteine rich Homo sapiens 189-194 20687958-7 2010 Interestingly, fenretinide [N-4(hydroxyphenyl) retinamide, 4-HPR], a synthetic retinoid with anti-cancer properties, was found to up-regulate the transcription, expression and secretion of SPARC via induction of the Brg-1 in a dose-dependent manner. Fenretinide 28-57 SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily a, member 4 Homo sapiens 216-221 20687958-8 2010 Finally, our results demonstrated that fenretinide-induced expression of SPARC contributes significantly to a decreased invasion of mammary carcinoma cells. Fenretinide 39-50 secreted protein acidic and cysteine rich Homo sapiens 73-78 20537156-6 2010 4HPR was able to decrease AKT phosphorylation also when powerfully upregulated by IGF-1 and, consequently, to impair IGF-1-stimulated cell motility. Fenretinide 0-4 insulin like growth factor 1 Homo sapiens 117-122 20034809-1 2010 Clinical trials have revealed that N-(4-hydroxyphenyl) retinamide (4HPR; fenretinide), a synthetic retinoic acid derivative, is a highly active and promising therapeutic and chemopreventive agent. Fenretinide 35-65 haptoglobin-related protein Homo sapiens 68-71 20034809-1 2010 Clinical trials have revealed that N-(4-hydroxyphenyl) retinamide (4HPR; fenretinide), a synthetic retinoic acid derivative, is a highly active and promising therapeutic and chemopreventive agent. Fenretinide 73-84 haptoglobin-related protein Homo sapiens 68-71 20537156-6 2010 4HPR was able to decrease AKT phosphorylation also when powerfully upregulated by IGF-1 and, consequently, to impair IGF-1-stimulated cell motility. Fenretinide 0-4 AKT serine/threonine kinase 1 Homo sapiens 26-29 20537156-6 2010 4HPR was able to decrease AKT phosphorylation also when powerfully upregulated by IGF-1 and, consequently, to impair IGF-1-stimulated cell motility. Fenretinide 0-4 insulin like growth factor 1 Homo sapiens 82-87 20399851-6 2010 In premalignant prostate epithelial cells and malignant cutaneous keratinocytes the suppression of DHODH activity by the chemical inhibitor teriflunomide or the reduction in DHODH protein expression by RNA interference markedly reduced 4HPR-induced ROS generation and apoptosis. Fenretinide 236-240 dihydroorotate dehydrogenase (quinone) Homo sapiens 99-104 20399851-6 2010 In premalignant prostate epithelial cells and malignant cutaneous keratinocytes the suppression of DHODH activity by the chemical inhibitor teriflunomide or the reduction in DHODH protein expression by RNA interference markedly reduced 4HPR-induced ROS generation and apoptosis. Fenretinide 236-240 dihydroorotate dehydrogenase (quinone) Homo sapiens 174-179 20537156-7 2010 Conversely, the expression of constitutively active AKT (myr-AKT) overcame the effects of 4HPR and beta-catenin-silencing on cell migration. Fenretinide 90-94 AKT serine/threonine kinase 1 Homo sapiens 52-55 20537156-7 2010 Conversely, the expression of constitutively active AKT (myr-AKT) overcame the effects of 4HPR and beta-catenin-silencing on cell migration. Fenretinide 90-94 AKT serine/threonine kinase 1 Homo sapiens 61-64 20537156-9 2010 CONCLUSION: These data point to 4HPR as a negative regulator of AKT phosphorylation, effectively targeting the beta-catenin pathway and inducing a relatively benign phenotype in prostate cancer cells, limiting neoangiogenesis and cell invasion. Fenretinide 32-36 AKT serine/threonine kinase 1 Homo sapiens 64-67 20537156-9 2010 CONCLUSION: These data point to 4HPR as a negative regulator of AKT phosphorylation, effectively targeting the beta-catenin pathway and inducing a relatively benign phenotype in prostate cancer cells, limiting neoangiogenesis and cell invasion. Fenretinide 32-36 catenin beta 1 Homo sapiens 111-123 19912993-0 2010 Induction and intracellular localization of Nur77 dictate fenretinide-induced apoptosis of human liver cancer cells. Fenretinide 58-69 nuclear receptor subfamily 4 group A member 1 Homo sapiens 44-49 19912993-4 2010 The induction of Nur77 and the cytoplasmic distribution of Nur77 induced by fenretinide were positively correlated with the apoptotic effect of fenretinide in HCC cells. Fenretinide 76-87 nuclear receptor subfamily 4 group A member 1 Homo sapiens 17-22 19912993-4 2010 The induction of Nur77 and the cytoplasmic distribution of Nur77 induced by fenretinide were positively correlated with the apoptotic effect of fenretinide in HCC cells. Fenretinide 76-87 nuclear receptor subfamily 4 group A member 1 Homo sapiens 59-64 19912993-4 2010 The induction of Nur77 and the cytoplasmic distribution of Nur77 induced by fenretinide were positively correlated with the apoptotic effect of fenretinide in HCC cells. Fenretinide 144-155 nuclear receptor subfamily 4 group A member 1 Homo sapiens 17-22 19912993-4 2010 The induction of Nur77 and the cytoplasmic distribution of Nur77 induced by fenretinide were positively correlated with the apoptotic effect of fenretinide in HCC cells. Fenretinide 144-155 nuclear receptor subfamily 4 group A member 1 Homo sapiens 59-64 19912993-6 2010 The intracellular location of Nur77 was also associated with the differential capability of fenretinide-induced ROS generation in these two cell lines. Fenretinide 92-103 nuclear receptor subfamily 4 group A member 1 Homo sapiens 30-35 19912993-7 2010 In addition, the knockdown of Nur77 expression by siRNA greatly reduced fenretinide-induced apoptosis and cleaved caspase 3 in Huh-7 cells. Fenretinide 72-83 nuclear receptor subfamily 4 group A member 1 Homo sapiens 30-35 19912993-8 2010 Therefore, our findings demonstrate that fenretinide-induced apoptosis of HCC cells is Nur77 dependent and that the intracellular localization of Nur77 dictates the sensitivity of the HCC cells to fenretinide-induced apoptosis. Fenretinide 41-52 nuclear receptor subfamily 4 group A member 1 Homo sapiens 87-92 19912993-8 2010 Therefore, our findings demonstrate that fenretinide-induced apoptosis of HCC cells is Nur77 dependent and that the intracellular localization of Nur77 dictates the sensitivity of the HCC cells to fenretinide-induced apoptosis. Fenretinide 197-208 nuclear receptor subfamily 4 group A member 1 Homo sapiens 146-151 20022965-3 2010 The aim of this study was to test the hypothesis that the early events of ER stress signaling and response pathways induced by fenretinide and bortezomib are mediated by the eukaryotic initiation factor 2alpha (eIF2alpha)-ATF4 signaling pathway. Fenretinide 127-138 eukaryotic translation initiation factor 2A Homo sapiens 211-220 20203181-8 2010 In addition, in vitro studies demonstrated that fenretinide reduced TNF-alpha (tumor necrosis factor-alpha) expression by reactive microglia. Fenretinide 48-59 tumor necrosis factor Mus musculus 68-77 20203181-8 2010 In addition, in vitro studies demonstrated that fenretinide reduced TNF-alpha (tumor necrosis factor-alpha) expression by reactive microglia. Fenretinide 48-59 tumor necrosis factor Mus musculus 79-106 20022965-3 2010 The aim of this study was to test the hypothesis that the early events of ER stress signaling and response pathways induced by fenretinide and bortezomib are mediated by the eukaryotic initiation factor 2alpha (eIF2alpha)-ATF4 signaling pathway. Fenretinide 127-138 activating transcription factor 4 Homo sapiens 222-226 20022965-4 2010 Treatment of neuroblastoma and melanoma cell lines with fenretinide, bortezomib, or thapsigargin resulted in induction of eIF2alpha signaling, characterized by increased expression of phosphorylated eIF2alpha, ATF4, ATF3, and GADD34. Fenretinide 56-67 eukaryotic translation initiation factor 2A Homo sapiens 122-131 20022965-4 2010 Treatment of neuroblastoma and melanoma cell lines with fenretinide, bortezomib, or thapsigargin resulted in induction of eIF2alpha signaling, characterized by increased expression of phosphorylated eIF2alpha, ATF4, ATF3, and GADD34. Fenretinide 56-67 eukaryotic translation initiation factor 2A Homo sapiens 199-208 20022965-4 2010 Treatment of neuroblastoma and melanoma cell lines with fenretinide, bortezomib, or thapsigargin resulted in induction of eIF2alpha signaling, characterized by increased expression of phosphorylated eIF2alpha, ATF4, ATF3, and GADD34. Fenretinide 56-67 activating transcription factor 4 Homo sapiens 210-214 20022965-4 2010 Treatment of neuroblastoma and melanoma cell lines with fenretinide, bortezomib, or thapsigargin resulted in induction of eIF2alpha signaling, characterized by increased expression of phosphorylated eIF2alpha, ATF4, ATF3, and GADD34. Fenretinide 56-67 activating transcription factor 3 Homo sapiens 216-220 20022965-4 2010 Treatment of neuroblastoma and melanoma cell lines with fenretinide, bortezomib, or thapsigargin resulted in induction of eIF2alpha signaling, characterized by increased expression of phosphorylated eIF2alpha, ATF4, ATF3, and GADD34. Fenretinide 56-67 protein phosphatase 1 regulatory subunit 15A Homo sapiens 226-232 20022965-7 2010 Although PERK-dependent phosphorylation of eIF2alpha enhanced ATF4 protein levels during ER stress, cell death in response to fenretinide, bortezomib, or thapsigargin was not abrogated by inhibition of eIF2alpha phosphorylation through PERK knockdown or overexpression of wild-type eIF2alpha. Fenretinide 126-137 eukaryotic translation initiation factor 2A Homo sapiens 43-52 20844597-0 2010 Role for PKC delta in Fenretinide-Mediated Apoptosis in Lymphoid Leukemia Cells. Fenretinide 22-33 protein kinase C delta Homo sapiens 9-18 20022965-9 2010 These results demonstrate that ATF4 mediates ER stress-induced cell death of neuroectodermal tumor cells in response to fenretinide or bortezomib. Fenretinide 120-131 activating transcription factor 4 Homo sapiens 31-35 20844597-2 2010 In the current paper, the role of PKC delta was examined in fenretinide-induced apoptosis in lymphoid leukemia cells. Fenretinide 60-71 protein kinase C delta Homo sapiens 34-43 20844597-8 2010 Now we have found that fenretinide-induced MCL-1 degradation may involve PKC delta as cleavage of the kinase correlated with loss of MCL-1 even in cells when JNK was not activated. Fenretinide 23-34 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 43-48 20844597-8 2010 Now we have found that fenretinide-induced MCL-1 degradation may involve PKC delta as cleavage of the kinase correlated with loss of MCL-1 even in cells when JNK was not activated. Fenretinide 23-34 protein kinase C delta Homo sapiens 73-82 20844597-5 2010 The antioxidant Vitamin C prevented fenretinide-induced PKC delta cleavage and protected cells from fenretinide. Fenretinide 36-47 protein kinase C delta Homo sapiens 56-65 20844597-8 2010 Now we have found that fenretinide-induced MCL-1 degradation may involve PKC delta as cleavage of the kinase correlated with loss of MCL-1 even in cells when JNK was not activated. Fenretinide 23-34 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 133-138 20844597-6 2010 Suppression of PKC delta expression by shRNA sensitized cells to fenretinide-induced apoptosis possibly by a mechanism involving ROS production. Fenretinide 65-76 protein kinase C delta Homo sapiens 15-24 20844597-8 2010 Now we have found that fenretinide-induced MCL-1 degradation may involve PKC delta as cleavage of the kinase correlated with loss of MCL-1 even in cells when JNK was not activated. Fenretinide 23-34 mitogen-activated protein kinase 8 Homo sapiens 158-161 20844597-9 2010 These results suggest that PKC delta may play a complex role in fenretinide-induced apoptosis and may be targeted in antileukemia strategies that utilize fenretinide. Fenretinide 64-75 protein kinase C delta Homo sapiens 27-36 20844597-9 2010 These results suggest that PKC delta may play a complex role in fenretinide-induced apoptosis and may be targeted in antileukemia strategies that utilize fenretinide. Fenretinide 154-165 protein kinase C delta Homo sapiens 27-36 20844597-7 2010 A previous study demonstrated that fenretinide promotes degradation of antiapoptotic MCL-1 in ALL cells via JNK. Fenretinide 35-46 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 85-90 20844597-7 2010 A previous study demonstrated that fenretinide promotes degradation of antiapoptotic MCL-1 in ALL cells via JNK. Fenretinide 35-46 mitogen-activated protein kinase 8 Homo sapiens 108-111 19621427-3 2009 We present a novel case of a child who developed an LCH bone lesion while receiving a Phase I protocol therapy with oral fenretinide/Lym-X-Sorb (4-HPR/LXS) powder for neuroblastoma. Fenretinide 121-132 haptoglobin-related protein Homo sapiens 147-150 19826103-0 2009 Long-term Fenretinide treatment prevents high-fat diet-induced obesity, insulin resistance, and hepatic steatosis. Fenretinide 10-21 insulin Homo sapiens 72-79 19826103-1 2009 The synthetic retinoid Fenretinide (FEN) increases insulin sensitivity in obese rodents and is in early clinical trials for treatment of insulin resistance in obese humans with hepatic steatosis (46). Fenretinide 23-34 insulin Homo sapiens 51-58 19826103-1 2009 The synthetic retinoid Fenretinide (FEN) increases insulin sensitivity in obese rodents and is in early clinical trials for treatment of insulin resistance in obese humans with hepatic steatosis (46). Fenretinide 23-34 insulin Homo sapiens 137-144 19826103-1 2009 The synthetic retinoid Fenretinide (FEN) increases insulin sensitivity in obese rodents and is in early clinical trials for treatment of insulin resistance in obese humans with hepatic steatosis (46). Fenretinide 36-39 insulin Homo sapiens 51-58 19826103-1 2009 The synthetic retinoid Fenretinide (FEN) increases insulin sensitivity in obese rodents and is in early clinical trials for treatment of insulin resistance in obese humans with hepatic steatosis (46). Fenretinide 36-39 insulin Homo sapiens 137-144 19996280-1 2009 The retinoid 4-oxo-N-(4-hydroxyphenyl)retinamide (4-oxo-4-HPR), a metabolite of fenretinide (4-HPR) present in plasma of 4-HPR-treated patients, is very effective in inducing growth inhibition and apoptosis in several cancer cell lines. Fenretinide 80-91 haptoglobin-related protein Homo sapiens 58-61 19996280-1 2009 The retinoid 4-oxo-N-(4-hydroxyphenyl)retinamide (4-oxo-4-HPR), a metabolite of fenretinide (4-HPR) present in plasma of 4-HPR-treated patients, is very effective in inducing growth inhibition and apoptosis in several cancer cell lines. Fenretinide 80-91 haptoglobin-related protein Homo sapiens 95-98 19996280-1 2009 The retinoid 4-oxo-N-(4-hydroxyphenyl)retinamide (4-oxo-4-HPR), a metabolite of fenretinide (4-HPR) present in plasma of 4-HPR-treated patients, is very effective in inducing growth inhibition and apoptosis in several cancer cell lines. Fenretinide 80-91 haptoglobin-related protein Homo sapiens 95-98 19844581-0 2009 Converting redox signaling to apoptotic activities by stress-responsive regulators HSF1 and NRF2 in fenretinide treated cancer cells. Fenretinide 100-111 heat shock transcription factor 1 Homo sapiens 83-87 19446953-2 2009 Here we show that 4HPR induces apoptosis through increased level of ROS and activation of caspase-8, 9 and 3, and inhibits growth of several MM cell lines in a dose-dependent manner. Fenretinide 18-22 caspase 8 Homo sapiens 90-108 19844581-0 2009 Converting redox signaling to apoptotic activities by stress-responsive regulators HSF1 and NRF2 in fenretinide treated cancer cells. Fenretinide 100-111 NFE2 like bZIP transcription factor 2 Homo sapiens 92-96 19482004-4 2009 To enable the search for nonretinoid molecules with fenretinide-like activities we developed a HTS-compatible homogeneous TR-FRET assay monitoring the displacement of retinoic acid derivatives from RBP4 in high-density 384-well and 1536-well microtiter plate formats. Fenretinide 52-63 retinol binding protein 4, plasma Mus musculus 198-202 19482004-2 2009 The retinoic acid derivative fenretinide (N-(4-hydroxyphenyl) retinamide; HPR) exerts therapeutic effects in mouse models of obesity, diabetes, and Stargardt"s disease by targeting RBP4. Fenretinide 29-40 retinol binding protein 4, plasma Mus musculus 181-185 19482004-2 2009 The retinoic acid derivative fenretinide (N-(4-hydroxyphenyl) retinamide; HPR) exerts therapeutic effects in mouse models of obesity, diabetes, and Stargardt"s disease by targeting RBP4. Fenretinide 42-72 retinol binding protein 4, plasma Mus musculus 181-185 19482076-1 2009 N-(4-hydroxyphenyl) retinamide (4-HPR), as a synthetic retinoid, has been shown to inhibit carcinogenesis in a variety of cancers. Fenretinide 0-30 haptoglobin-related protein Homo sapiens 34-37 19482004-3 2009 Fenretinide competes with retinoids for RBP4 binding, disrupts RBP4-transthyretin (TTR) complexes, and results in urinary secretion of RBP4 and systemic depletion of retinol. Fenretinide 0-11 retinol binding protein 4, plasma Mus musculus 40-44 19482004-3 2009 Fenretinide competes with retinoids for RBP4 binding, disrupts RBP4-transthyretin (TTR) complexes, and results in urinary secretion of RBP4 and systemic depletion of retinol. Fenretinide 0-11 transthyretin Mus musculus 63-81 19482004-3 2009 Fenretinide competes with retinoids for RBP4 binding, disrupts RBP4-transthyretin (TTR) complexes, and results in urinary secretion of RBP4 and systemic depletion of retinol. Fenretinide 0-11 transthyretin Mus musculus 83-86 19482004-3 2009 Fenretinide competes with retinoids for RBP4 binding, disrupts RBP4-transthyretin (TTR) complexes, and results in urinary secretion of RBP4 and systemic depletion of retinol. Fenretinide 0-11 retinol binding protein 4, plasma Mus musculus 63-67 19421858-2 2009 The synthetic retinoid N-(4-hydroxyphenyl)retinamide (4HPR) inhibits prostate cancer development in vivo, and triggers reactive oxygen species (ROS)-dependent prostate cancer cell apoptosis in vitro. Fenretinide 23-52 haptoglobin-related protein Homo sapiens 55-58 19041174-0 2009 Identification of ADP-ribosylation factor 4 as a suppressor of N-(4-hydroxyphenyl)retinamide-induced cell death. Fenretinide 63-92 ADP ribosylation factor 4 Homo sapiens 18-43 19325135-0 2009 PLAB induction in fenretinide-induced apoptosis of ovarian cancer cells occurs via a ROS-dependent mechanism involving ER stress and JNK activation. Fenretinide 18-29 mitogen-activated protein kinase 8 Homo sapiens 133-136 19325135-2 2009 We showed previously that 4HPR upregulates the proapoptotic gene placental bone morphogenetic protein (PLAB), which is a mediator of 4HPR-induced apoptosis in ovarian cancer cells. Fenretinide 26-30 growth differentiation factor 15 Homo sapiens 65-101 19325135-2 2009 We showed previously that 4HPR upregulates the proapoptotic gene placental bone morphogenetic protein (PLAB), which is a mediator of 4HPR-induced apoptosis in ovarian cancer cells. Fenretinide 133-137 growth differentiation factor 15 Homo sapiens 65-101 19325135-9 2009 These data indicate that 4HPR-induced PLAB upregulation occurs downstream of a signaling cascade involving ROS generation, ER stress induction and JNK activation and that these steps are mediators of 4HPR-induced apoptosis. Fenretinide 25-29 mitogen-activated protein kinase 8 Homo sapiens 147-150 19285047-8 2009 Collectively, the combination of 4-HPR and PTX diminished the survival factors (e.g., rTERT, PCNA, and Bcl-2) to make glioblastoma cells highly prone to apoptosis with activation of cysteine proteases (e.g., calpain, cathepsins, caspase-8, caspase-3). Fenretinide 33-38 proliferating cell nuclear antigen Rattus norvegicus 93-97 19285047-8 2009 Collectively, the combination of 4-HPR and PTX diminished the survival factors (e.g., rTERT, PCNA, and Bcl-2) to make glioblastoma cells highly prone to apoptosis with activation of cysteine proteases (e.g., calpain, cathepsins, caspase-8, caspase-3). Fenretinide 33-38 BCL2, apoptosis regulator Rattus norvegicus 103-108 19285047-8 2009 Collectively, the combination of 4-HPR and PTX diminished the survival factors (e.g., rTERT, PCNA, and Bcl-2) to make glioblastoma cells highly prone to apoptosis with activation of cysteine proteases (e.g., calpain, cathepsins, caspase-8, caspase-3). Fenretinide 33-38 caspase 8 Rattus norvegicus 229-238 19285047-8 2009 Collectively, the combination of 4-HPR and PTX diminished the survival factors (e.g., rTERT, PCNA, and Bcl-2) to make glioblastoma cells highly prone to apoptosis with activation of cysteine proteases (e.g., calpain, cathepsins, caspase-8, caspase-3). Fenretinide 33-38 caspase 3 Rattus norvegicus 240-249 19041174-2 2009 Yeast or U373MG cells that overexpressed ARF4 exhibited reduced reactive oxygen species (ROS) generation in response to Bax or N-(4-hydroxyphenyl)retinamide (4-HPR), respectively, which suggests that ROS play a role in the inhibition of cell death by ARF4. Fenretinide 127-156 ADP ribosylation factor 4 Homo sapiens 41-45 19010927-0 2008 Effect of fenretinide and low-dose tamoxifen on insulin sensitivity in premenopausal women at high risk for breast cancer. Fenretinide 10-21 insulin Homo sapiens 48-55 19331667-0 2009 Fenretinide-induced caspase-8 activation and apoptosis in an established model of metastatic neuroblastoma. Fenretinide 0-11 caspase 8 Homo sapiens 20-29 19331667-2 2009 Patients are treated by maintenance CT. For some patients, an adjuvant retinoid therapy is proposed, such as the synthetic retinoid fenretinide (4-HPR), an apoptotic inducer. Fenretinide 132-143 haptoglobin-related protein Homo sapiens 147-150 19331667-6 2009 NB cell lines, i.e., IGR-N-91 and SH-EP, were treated with various doses of Fenretinide (4-HPR), then cytotoxicity was analyzed by MTS proliferation assay, apoptosis by the propidium staining method, gene or protein expressions by RT-PCR and immunoblotting and caspases activity by colorimetric protease assays. Fenretinide 76-87 haptoglobin-related protein Homo sapiens 91-94 18952041-3 2009 This can be accomplished by administration of small molecules, such as fenretinide, that compete with retinol for binding to RBP4 and disrupt the protein-protein interaction between RBP4 and transthyretin (TTR), another serum protein that protects RBP4 from renal clearance. Fenretinide 71-82 retinol binding protein 4 Homo sapiens 125-129 18952041-3 2009 This can be accomplished by administration of small molecules, such as fenretinide, that compete with retinol for binding to RBP4 and disrupt the protein-protein interaction between RBP4 and transthyretin (TTR), another serum protein that protects RBP4 from renal clearance. Fenretinide 71-82 retinol binding protein 4 Homo sapiens 182-186 18952041-3 2009 This can be accomplished by administration of small molecules, such as fenretinide, that compete with retinol for binding to RBP4 and disrupt the protein-protein interaction between RBP4 and transthyretin (TTR), another serum protein that protects RBP4 from renal clearance. Fenretinide 71-82 transthyretin Homo sapiens 191-204 18952041-3 2009 This can be accomplished by administration of small molecules, such as fenretinide, that compete with retinol for binding to RBP4 and disrupt the protein-protein interaction between RBP4 and transthyretin (TTR), another serum protein that protects RBP4 from renal clearance. Fenretinide 71-82 transthyretin Homo sapiens 206-209 18952041-3 2009 This can be accomplished by administration of small molecules, such as fenretinide, that compete with retinol for binding to RBP4 and disrupt the protein-protein interaction between RBP4 and transthyretin (TTR), another serum protein that protects RBP4 from renal clearance. Fenretinide 71-82 retinol binding protein 4 Homo sapiens 182-186 18952041-5 2009 We present an allosteric model that describes the pharmacology of interaction among RBP4, TTR, retinol, and fenretinide, and we show data that support the model. Fenretinide 108-119 retinol binding protein 4 Homo sapiens 84-88 18952041-5 2009 We present an allosteric model that describes the pharmacology of interaction among RBP4, TTR, retinol, and fenretinide, and we show data that support the model. Fenretinide 108-119 transthyretin Homo sapiens 90-93 18952041-6 2009 We show that retinol increases the affinity of RBP4 for TTR by a factor of 4 and determine the affinity constants of fenretinide and retinyl acetate. Fenretinide 117-128 retinol binding protein 4 Homo sapiens 47-51 19213559-4 2009 In this manuscript we use as an example the synthetic retinoid N-(4-hydroxyphenyl)retinamide (4HPR), a molecule with confirmed clinical applications in breast cancer adjuvant therapy to prevent cancer recurrence and under evaluation in neuroblastoma and glioblastoma treatment. Fenretinide 63-92 haptoglobin-related protein Homo sapiens 95-98 18515328-3 2008 Here, we show that the natural retinoid 9-cis retinoic acid (9cRA) and the synthetic retinoid fenretinide (4-HPR) specifically inhibit the functional up-regulation of CCR7 on maturing human DCs, without affecting early steps of DC maturation. Fenretinide 94-105 C-C motif chemokine receptor 7 Homo sapiens 167-171 18066548-1 2008 PURPOSE: Pharmacokinetic data on fenretinide (4-HPR) are scant, thus limiting the rational use of the drug. Fenretinide 33-44 haptoglobin-related protein Homo sapiens 48-51 18471963-1 2008 The role of NF-kappaB in the Ewing"s sarcoma family of tumours (ESFT) and their response to fenretinide has been investigated. Fenretinide 92-103 nuclear factor kappa B subunit 1 Homo sapiens 12-21 18515328-3 2008 Here, we show that the natural retinoid 9-cis retinoic acid (9cRA) and the synthetic retinoid fenretinide (4-HPR) specifically inhibit the functional up-regulation of CCR7 on maturing human DCs, without affecting early steps of DC maturation. Fenretinide 94-105 haptoglobin-related protein Homo sapiens 109-112 18549809-4 2008 4-HPR-induced mitochondrial translocation of Bax, release of mitochondrial cytochrome c, and activation of caspase-3 were markedly attenuated in U373MG cells that stably expressed COX6A1. Fenretinide 0-5 BCL2 associated X, apoptosis regulator Homo sapiens 45-48 18602901-1 2008 N-(4-Hydroxyphenyl) retinamide (4-HPR) is a synthetic retinoid that has shown biological activity against several malignant tumors and minimal side effects in humans. Fenretinide 0-30 haptoglobin-related protein Homo sapiens 34-37 18549809-2 2008 The overexpression of COX6A1 significantly suppressed Bax- and N-(4-hydroxyphenyl)retinamide (4-HPR)-induced apoptosis in yeast and human glioblastoma-derived U373MG cells, respectively. Fenretinide 63-92 cytochrome c oxidase subunit 6A1 Homo sapiens 22-28 18549809-4 2008 4-HPR-induced mitochondrial translocation of Bax, release of mitochondrial cytochrome c, and activation of caspase-3 were markedly attenuated in U373MG cells that stably expressed COX6A1. Fenretinide 0-5 cytochrome c, somatic Homo sapiens 75-87 18549809-2 2008 The overexpression of COX6A1 significantly suppressed Bax- and N-(4-hydroxyphenyl)retinamide (4-HPR)-induced apoptosis in yeast and human glioblastoma-derived U373MG cells, respectively. Fenretinide 94-99 cytochrome c oxidase subunit 6A1 Homo sapiens 22-28 18549809-4 2008 4-HPR-induced mitochondrial translocation of Bax, release of mitochondrial cytochrome c, and activation of caspase-3 were markedly attenuated in U373MG cells that stably expressed COX6A1. Fenretinide 0-5 caspase 3 Homo sapiens 107-116 18549809-4 2008 4-HPR-induced mitochondrial translocation of Bax, release of mitochondrial cytochrome c, and activation of caspase-3 were markedly attenuated in U373MG cells that stably expressed COX6A1. Fenretinide 0-5 cytochrome c oxidase subunit 6A1 Homo sapiens 180-186 18387645-2 2008 JWA, a novel retinoic acid-inducible gene, is known to be involved in apoptosis induced by various agents, for example, 12-O-tetradecanoylphorbol 13-acetate, N-4-hydroxy-phenyl-retinamide and arsenic trioxide. Fenretinide 158-187 ADP ribosylation factor like GTPase 6 interacting protein 5 Homo sapiens 0-3 18410500-5 2008 By blocking the upstream kinase MEK1/2 with specific inhibitor U0126 we abrogated the 4HPR-induced phosphorylation of ERK1/2 and SAPK/JNK, indicating that the neuronal differentiation occurs through a positive cross-talk between the ERK and the SAPK/JNK pathways. Fenretinide 86-90 mitogen-activated protein kinase kinase 1 Homo sapiens 32-38 18410500-5 2008 By blocking the upstream kinase MEK1/2 with specific inhibitor U0126 we abrogated the 4HPR-induced phosphorylation of ERK1/2 and SAPK/JNK, indicating that the neuronal differentiation occurs through a positive cross-talk between the ERK and the SAPK/JNK pathways. Fenretinide 86-90 mitogen-activated protein kinase 3 Homo sapiens 118-124 18410500-5 2008 By blocking the upstream kinase MEK1/2 with specific inhibitor U0126 we abrogated the 4HPR-induced phosphorylation of ERK1/2 and SAPK/JNK, indicating that the neuronal differentiation occurs through a positive cross-talk between the ERK and the SAPK/JNK pathways. Fenretinide 86-90 mitogen-activated protein kinase 9 Homo sapiens 129-133 18410500-5 2008 By blocking the upstream kinase MEK1/2 with specific inhibitor U0126 we abrogated the 4HPR-induced phosphorylation of ERK1/2 and SAPK/JNK, indicating that the neuronal differentiation occurs through a positive cross-talk between the ERK and the SAPK/JNK pathways. Fenretinide 86-90 mitogen-activated protein kinase 9 Homo sapiens 134-137 18410500-5 2008 By blocking the upstream kinase MEK1/2 with specific inhibitor U0126 we abrogated the 4HPR-induced phosphorylation of ERK1/2 and SAPK/JNK, indicating that the neuronal differentiation occurs through a positive cross-talk between the ERK and the SAPK/JNK pathways. Fenretinide 86-90 mitogen-activated protein kinase 1 Homo sapiens 118-121 18410500-5 2008 By blocking the upstream kinase MEK1/2 with specific inhibitor U0126 we abrogated the 4HPR-induced phosphorylation of ERK1/2 and SAPK/JNK, indicating that the neuronal differentiation occurs through a positive cross-talk between the ERK and the SAPK/JNK pathways. Fenretinide 86-90 mitogen-activated protein kinase 9 Homo sapiens 245-249 18410500-5 2008 By blocking the upstream kinase MEK1/2 with specific inhibitor U0126 we abrogated the 4HPR-induced phosphorylation of ERK1/2 and SAPK/JNK, indicating that the neuronal differentiation occurs through a positive cross-talk between the ERK and the SAPK/JNK pathways. Fenretinide 86-90 mitogen-activated protein kinase 9 Homo sapiens 250-253 18519704-0 2008 Involvement of Rac in fenretinide-induced apoptosis. Fenretinide 22-33 AKT serine/threonine kinase 1 Homo sapiens 15-18 18519704-3 2008 We explored the possibility that ROS induction by 4HPR involves the small GTPase Ras-related C3 botulinum toxin substrate (Rac), a regulatory subunit of the NADPH oxidase complex. Fenretinide 50-54 AKT serine/threonine kinase 1 Homo sapiens 123-126 18425327-1 2008 The efficacy and mechanism of action of fenretinide (4-HPR), a vitamin A analogue, was investigated in a panel of six neuroblastoma cell lines and multicellular tumor spheroids. Fenretinide 40-51 haptoglobin-related protein Homo sapiens 55-58 18259116-6 2008 In particular we found that fenretinide treatment resulted in the increase in Beclin 1 expression, the conversion of the soluble form of LC3 to the autophagic vesicle-associated form LC3-II and its shift from diffuse to punctate staining and finally the increase in lysosomes/autophagosomes. Fenretinide 28-39 beclin 1 Homo sapiens 78-86 18259116-6 2008 In particular we found that fenretinide treatment resulted in the increase in Beclin 1 expression, the conversion of the soluble form of LC3 to the autophagic vesicle-associated form LC3-II and its shift from diffuse to punctate staining and finally the increase in lysosomes/autophagosomes. Fenretinide 28-39 microtubule associated protein 1 light chain 3 alpha Homo sapiens 137-140 18259116-6 2008 In particular we found that fenretinide treatment resulted in the increase in Beclin 1 expression, the conversion of the soluble form of LC3 to the autophagic vesicle-associated form LC3-II and its shift from diffuse to punctate staining and finally the increase in lysosomes/autophagosomes. Fenretinide 28-39 microtubule associated protein 1 light chain 3 alpha Homo sapiens 183-186 18259116-7 2008 By contrast, caspase-3 reconstituted MCF-7 cell line showed apoptotic cell death features in response to fenretinide treatment. Fenretinide 105-116 caspase 3 Homo sapiens 13-22 18342855-0 2008 The PERK/eIF2 alpha signaling pathway of Unfolded Protein Response is essential for N-(4-hydroxyphenyl)retinamide (4HPR)-induced cytotoxicity in cancer cells. Fenretinide 84-113 eukaryotic translation initiation factor 2 alpha kinase 3 Homo sapiens 4-8 18342855-0 2008 The PERK/eIF2 alpha signaling pathway of Unfolded Protein Response is essential for N-(4-hydroxyphenyl)retinamide (4HPR)-induced cytotoxicity in cancer cells. Fenretinide 84-113 eukaryotic translation initiation factor 2A Homo sapiens 9-19 18342855-0 2008 The PERK/eIF2 alpha signaling pathway of Unfolded Protein Response is essential for N-(4-hydroxyphenyl)retinamide (4HPR)-induced cytotoxicity in cancer cells. Fenretinide 115-119 eukaryotic translation initiation factor 2 alpha kinase 3 Homo sapiens 4-8 18342855-0 2008 The PERK/eIF2 alpha signaling pathway of Unfolded Protein Response is essential for N-(4-hydroxyphenyl)retinamide (4HPR)-induced cytotoxicity in cancer cells. Fenretinide 115-119 eukaryotic translation initiation factor 2A Homo sapiens 9-19 18342855-1 2008 N-(4-hydroxyphenyl)retinamide (4HPR) is a synthetic retinoid that has been tested in clinical trials as a cancer chemopreventive drug. Fenretinide 0-29 haptoglobin-related protein Homo sapiens 32-35 17988958-0 2008 Fenretinide prevents the development of osteoporosis in Cftr-KO mice. Fenretinide 0-11 cystic fibrosis transmembrane conductance regulator Mus musculus 56-60 17988958-4 2008 In this study, we characterized the protective effect of fenretinide [N-(4-hydroxyphenyl) retinamide], a vitamin A derivative, on the early onset of osteoporosis in cystic fibrosis transmembrane conductance regulator knockout (Cftr-KO) mice. Fenretinide 57-68 cystic fibrosis transmembrane conductance regulator Mus musculus 165-216 17988958-4 2008 In this study, we characterized the protective effect of fenretinide [N-(4-hydroxyphenyl) retinamide], a vitamin A derivative, on the early onset of osteoporosis in cystic fibrosis transmembrane conductance regulator knockout (Cftr-KO) mice. Fenretinide 57-68 cystic fibrosis transmembrane conductance regulator Mus musculus 227-231 17988958-4 2008 In this study, we characterized the protective effect of fenretinide [N-(4-hydroxyphenyl) retinamide], a vitamin A derivative, on the early onset of osteoporosis in cystic fibrosis transmembrane conductance regulator knockout (Cftr-KO) mice. Fenretinide 70-100 cystic fibrosis transmembrane conductance regulator Mus musculus 165-216 17988958-4 2008 In this study, we characterized the protective effect of fenretinide [N-(4-hydroxyphenyl) retinamide], a vitamin A derivative, on the early onset of osteoporosis in cystic fibrosis transmembrane conductance regulator knockout (Cftr-KO) mice. Fenretinide 70-100 cystic fibrosis transmembrane conductance regulator Mus musculus 227-231 18398104-0 2008 Mechanism of synergy of N-(4-hydroxyphenyl)retinamide and ABT-737 in acute lymphoblastic leukemia cell lines: Mcl-1 inactivation. Fenretinide 24-53 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 110-115 18416830-2 2008 Here, we have demonstrated that the observed effects using ATRA and 9-cis RA are shared with the clinically useful RAR ligand, 13-cis retinoic acid (13-cis RA), and the retinoic acid receptor-alpha (RAR-alpha)-selective agonist, AM580 but not with the RAR-beta/gamma ligand, 4-hydroxyphenylretinamide (4-HPR). Fenretinide 275-300 retinoic acid receptor alpha Homo sapiens 115-118 18416830-2 2008 Here, we have demonstrated that the observed effects using ATRA and 9-cis RA are shared with the clinically useful RAR ligand, 13-cis retinoic acid (13-cis RA), and the retinoic acid receptor-alpha (RAR-alpha)-selective agonist, AM580 but not with the RAR-beta/gamma ligand, 4-hydroxyphenylretinamide (4-HPR). Fenretinide 275-300 retinoic acid receptor alpha Homo sapiens 169-197 18416830-2 2008 Here, we have demonstrated that the observed effects using ATRA and 9-cis RA are shared with the clinically useful RAR ligand, 13-cis retinoic acid (13-cis RA), and the retinoic acid receptor-alpha (RAR-alpha)-selective agonist, AM580 but not with the RAR-beta/gamma ligand, 4-hydroxyphenylretinamide (4-HPR). Fenretinide 275-300 retinoic acid receptor alpha Homo sapiens 199-208 18416830-2 2008 Here, we have demonstrated that the observed effects using ATRA and 9-cis RA are shared with the clinically useful RAR ligand, 13-cis retinoic acid (13-cis RA), and the retinoic acid receptor-alpha (RAR-alpha)-selective agonist, AM580 but not with the RAR-beta/gamma ligand, 4-hydroxyphenylretinamide (4-HPR). Fenretinide 302-307 retinoic acid receptor alpha Homo sapiens 115-118 18416830-2 2008 Here, we have demonstrated that the observed effects using ATRA and 9-cis RA are shared with the clinically useful RAR ligand, 13-cis retinoic acid (13-cis RA), and the retinoic acid receptor-alpha (RAR-alpha)-selective agonist, AM580 but not with the RAR-beta/gamma ligand, 4-hydroxyphenylretinamide (4-HPR). Fenretinide 302-307 retinoic acid receptor alpha Homo sapiens 169-197 18416830-2 2008 Here, we have demonstrated that the observed effects using ATRA and 9-cis RA are shared with the clinically useful RAR ligand, 13-cis retinoic acid (13-cis RA), and the retinoic acid receptor-alpha (RAR-alpha)-selective agonist, AM580 but not with the RAR-beta/gamma ligand, 4-hydroxyphenylretinamide (4-HPR). Fenretinide 302-307 retinoic acid receptor alpha Homo sapiens 199-208 18398104-3 2008 The synthetic cytotoxic retinoid N-(4-hydroxyphenyl)retinamide (4-HPR) is known to generate reactive oxygen species (ROS), and ROS have been shown to activate c-Jun kinase (JNK), which in turn phosphorylates and inhibits Mcl-1. Fenretinide 33-62 haptoglobin-related protein Homo sapiens 66-69 18398104-3 2008 The synthetic cytotoxic retinoid N-(4-hydroxyphenyl)retinamide (4-HPR) is known to generate reactive oxygen species (ROS), and ROS have been shown to activate c-Jun kinase (JNK), which in turn phosphorylates and inhibits Mcl-1. Fenretinide 33-62 mitogen-activated protein kinase 8 Homo sapiens 159-171 18398104-3 2008 The synthetic cytotoxic retinoid N-(4-hydroxyphenyl)retinamide (4-HPR) is known to generate reactive oxygen species (ROS), and ROS have been shown to activate c-Jun kinase (JNK), which in turn phosphorylates and inhibits Mcl-1. Fenretinide 33-62 mitogen-activated protein kinase 8 Homo sapiens 173-176 18398104-3 2008 The synthetic cytotoxic retinoid N-(4-hydroxyphenyl)retinamide (4-HPR) is known to generate reactive oxygen species (ROS), and ROS have been shown to activate c-Jun kinase (JNK), which in turn phosphorylates and inhibits Mcl-1. Fenretinide 33-62 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 221-226 17700534-0 2008 The sensitivity of the Ewing"s sarcoma family of tumours to fenretinide-induced cell death is increased by EWS-Fli1-dependent modulation of p38(MAPK) activity. Fenretinide 60-71 EWS RNA binding protein 1 Homo sapiens 107-110 18174255-2 2008 Others and we have previously demonstrated apoptosis-inducing capacity of N-(4-hydroxyphenyl)retinamide (4-HPR) in malignant glioma cells. Fenretinide 74-103 haptoglobin-related protein Homo sapiens 107-110 18164543-0 2008 N-(4-Hydroxyphenyl)retinamide induced differentiation with repression of telomerase and cell cycle to increase interferon-gamma sensitivity for apoptosis in human glioblastoma cells. Fenretinide 0-29 interferon gamma Homo sapiens 111-127 18164543-3 2008 N-(4-Hydroxyphenyl)retinamide (4-HPR) induced astrocytic differentiation and increased sensitivity to interferon-gamma (IFN-gamma) for apoptosis in human glioblastoma A172, LN18, and SNB19 cells. Fenretinide 0-29 haptoglobin-related protein Homo sapiens 33-36 18164543-3 2008 N-(4-Hydroxyphenyl)retinamide (4-HPR) induced astrocytic differentiation and increased sensitivity to interferon-gamma (IFN-gamma) for apoptosis in human glioblastoma A172, LN18, and SNB19 cells. Fenretinide 0-29 interferon gamma Homo sapiens 102-118 18164543-3 2008 N-(4-Hydroxyphenyl)retinamide (4-HPR) induced astrocytic differentiation and increased sensitivity to interferon-gamma (IFN-gamma) for apoptosis in human glioblastoma A172, LN18, and SNB19 cells. Fenretinide 0-29 interferon gamma Homo sapiens 120-129 18248980-0 2008 4-Hydroxyphenylretinamide (4HPR) derivatives regulate aromatase activity and expression in breast cancer cells. Fenretinide 0-25 haptoglobin-related protein Homo sapiens 28-31 18248980-1 2008 Recent studies exhibit that 4-hydroxyphenylretinamide (4HPR) decreases aromatase activity in breast and placental cells. Fenretinide 28-53 haptoglobin-related protein Homo sapiens 56-59 17700534-0 2008 The sensitivity of the Ewing"s sarcoma family of tumours to fenretinide-induced cell death is increased by EWS-Fli1-dependent modulation of p38(MAPK) activity. Fenretinide 60-71 Fli-1 proto-oncogene, ETS transcription factor Homo sapiens 111-115 17700534-0 2008 The sensitivity of the Ewing"s sarcoma family of tumours to fenretinide-induced cell death is increased by EWS-Fli1-dependent modulation of p38(MAPK) activity. Fenretinide 60-71 mitogen-activated protein kinase 14 Homo sapiens 140-143 17700534-3 2008 Here, we demonstrate for the first time that the sensitivity of ESFT cells to fenretinide-induced cell death is decreased following downregulation of the oncogenic fusion protein EWS-Fli1; siRNA targeting EWS-Fli1 attenuated fenretinide-induced cell death in cell lines expressing EWS-Fli1, but not EWS-ERG. Fenretinide 78-89 EWS RNA binding protein 1 Homo sapiens 179-182 17700534-3 2008 Here, we demonstrate for the first time that the sensitivity of ESFT cells to fenretinide-induced cell death is decreased following downregulation of the oncogenic fusion protein EWS-Fli1; siRNA targeting EWS-Fli1 attenuated fenretinide-induced cell death in cell lines expressing EWS-Fli1, but not EWS-ERG. Fenretinide 78-89 Fli-1 proto-oncogene, ETS transcription factor Homo sapiens 183-187 17700534-3 2008 Here, we demonstrate for the first time that the sensitivity of ESFT cells to fenretinide-induced cell death is decreased following downregulation of the oncogenic fusion protein EWS-Fli1; siRNA targeting EWS-Fli1 attenuated fenretinide-induced cell death in cell lines expressing EWS-Fli1, but not EWS-ERG. Fenretinide 78-89 EWS RNA binding protein 1 Homo sapiens 205-208 17700534-3 2008 Here, we demonstrate for the first time that the sensitivity of ESFT cells to fenretinide-induced cell death is decreased following downregulation of the oncogenic fusion protein EWS-Fli1; siRNA targeting EWS-Fli1 attenuated fenretinide-induced cell death in cell lines expressing EWS-Fli1, but not EWS-ERG. Fenretinide 78-89 Fli-1 proto-oncogene, ETS transcription factor Homo sapiens 209-213 17700534-3 2008 Here, we demonstrate for the first time that the sensitivity of ESFT cells to fenretinide-induced cell death is decreased following downregulation of the oncogenic fusion protein EWS-Fli1; siRNA targeting EWS-Fli1 attenuated fenretinide-induced cell death in cell lines expressing EWS-Fli1, but not EWS-ERG. Fenretinide 78-89 EWS RNA binding protein 1 Homo sapiens 205-208 17700534-3 2008 Here, we demonstrate for the first time that the sensitivity of ESFT cells to fenretinide-induced cell death is decreased following downregulation of the oncogenic fusion protein EWS-Fli1; siRNA targeting EWS-Fli1 attenuated fenretinide-induced cell death in cell lines expressing EWS-Fli1, but not EWS-ERG. Fenretinide 78-89 Fli-1 proto-oncogene, ETS transcription factor Homo sapiens 209-213 17700534-3 2008 Here, we demonstrate for the first time that the sensitivity of ESFT cells to fenretinide-induced cell death is decreased following downregulation of the oncogenic fusion protein EWS-Fli1; siRNA targeting EWS-Fli1 attenuated fenretinide-induced cell death in cell lines expressing EWS-Fli1, but not EWS-ERG. Fenretinide 78-89 EWS RNA binding protein 1 Homo sapiens 205-208 17700534-3 2008 Here, we demonstrate for the first time that the sensitivity of ESFT cells to fenretinide-induced cell death is decreased following downregulation of the oncogenic fusion protein EWS-Fli1; siRNA targeting EWS-Fli1 attenuated fenretinide-induced cell death in cell lines expressing EWS-Fli1, but not EWS-ERG. Fenretinide 225-236 EWS RNA binding protein 1 Homo sapiens 179-182 17700534-3 2008 Here, we demonstrate for the first time that the sensitivity of ESFT cells to fenretinide-induced cell death is decreased following downregulation of the oncogenic fusion protein EWS-Fli1; siRNA targeting EWS-Fli1 attenuated fenretinide-induced cell death in cell lines expressing EWS-Fli1, but not EWS-ERG. Fenretinide 225-236 Fli-1 proto-oncogene, ETS transcription factor Homo sapiens 183-187 17700534-3 2008 Here, we demonstrate for the first time that the sensitivity of ESFT cells to fenretinide-induced cell death is decreased following downregulation of the oncogenic fusion protein EWS-Fli1; siRNA targeting EWS-Fli1 attenuated fenretinide-induced cell death in cell lines expressing EWS-Fli1, but not EWS-ERG. Fenretinide 225-236 EWS RNA binding protein 1 Homo sapiens 205-208 17700534-3 2008 Here, we demonstrate for the first time that the sensitivity of ESFT cells to fenretinide-induced cell death is decreased following downregulation of the oncogenic fusion protein EWS-Fli1; siRNA targeting EWS-Fli1 attenuated fenretinide-induced cell death in cell lines expressing EWS-Fli1, but not EWS-ERG. Fenretinide 225-236 Fli-1 proto-oncogene, ETS transcription factor Homo sapiens 209-213 17700534-3 2008 Here, we demonstrate for the first time that the sensitivity of ESFT cells to fenretinide-induced cell death is decreased following downregulation of the oncogenic fusion protein EWS-Fli1; siRNA targeting EWS-Fli1 attenuated fenretinide-induced cell death in cell lines expressing EWS-Fli1, but not EWS-ERG. Fenretinide 225-236 EWS RNA binding protein 1 Homo sapiens 205-208 17700534-3 2008 Here, we demonstrate for the first time that the sensitivity of ESFT cells to fenretinide-induced cell death is decreased following downregulation of the oncogenic fusion protein EWS-Fli1; siRNA targeting EWS-Fli1 attenuated fenretinide-induced cell death in cell lines expressing EWS-Fli1, but not EWS-ERG. Fenretinide 225-236 Fli-1 proto-oncogene, ETS transcription factor Homo sapiens 209-213 17700534-3 2008 Here, we demonstrate for the first time that the sensitivity of ESFT cells to fenretinide-induced cell death is decreased following downregulation of the oncogenic fusion protein EWS-Fli1; siRNA targeting EWS-Fli1 attenuated fenretinide-induced cell death in cell lines expressing EWS-Fli1, but not EWS-ERG. Fenretinide 225-236 EWS RNA binding protein 1 Homo sapiens 205-208 17700534-4 2008 This decrease in cell death was independent of the level of ROS produced following exposure to fenretinide, but was effected through EWS-Fli1-dependent modulation of p38(MAPK) activity. Fenretinide 95-106 EWS RNA binding protein 1 Homo sapiens 133-136 17700534-4 2008 This decrease in cell death was independent of the level of ROS produced following exposure to fenretinide, but was effected through EWS-Fli1-dependent modulation of p38(MAPK) activity. Fenretinide 95-106 Fli-1 proto-oncogene, ETS transcription factor Homo sapiens 137-141 17700534-5 2008 Furthermore, inhibition of p38(MAPK) activity and knockdown of EWS-Fli1 reduced fenretinide-induced mitochondrial permeabilization, cytochrome c release, caspase and PARP cleavage, consistent with the hypothesis that p38(MAPK) is critical for activation of the death cascade by fenretinide in ESFT cells. Fenretinide 80-91 mitogen-activated protein kinase 14 Homo sapiens 27-30 17700534-5 2008 Furthermore, inhibition of p38(MAPK) activity and knockdown of EWS-Fli1 reduced fenretinide-induced mitochondrial permeabilization, cytochrome c release, caspase and PARP cleavage, consistent with the hypothesis that p38(MAPK) is critical for activation of the death cascade by fenretinide in ESFT cells. Fenretinide 80-91 EWS RNA binding protein 1 Homo sapiens 63-66 17700534-5 2008 Furthermore, inhibition of p38(MAPK) activity and knockdown of EWS-Fli1 reduced fenretinide-induced mitochondrial permeabilization, cytochrome c release, caspase and PARP cleavage, consistent with the hypothesis that p38(MAPK) is critical for activation of the death cascade by fenretinide in ESFT cells. Fenretinide 80-91 Fli-1 proto-oncogene, ETS transcription factor Homo sapiens 67-71 17700534-5 2008 Furthermore, inhibition of p38(MAPK) activity and knockdown of EWS-Fli1 reduced fenretinide-induced mitochondrial permeabilization, cytochrome c release, caspase and PARP cleavage, consistent with the hypothesis that p38(MAPK) is critical for activation of the death cascade by fenretinide in ESFT cells. Fenretinide 80-91 cytochrome c, somatic Homo sapiens 132-144 17700534-5 2008 Furthermore, inhibition of p38(MAPK) activity and knockdown of EWS-Fli1 reduced fenretinide-induced mitochondrial permeabilization, cytochrome c release, caspase and PARP cleavage, consistent with the hypothesis that p38(MAPK) is critical for activation of the death cascade by fenretinide in ESFT cells. Fenretinide 80-91 poly(ADP-ribose) polymerase 1 Homo sapiens 166-170 17700534-5 2008 Furthermore, inhibition of p38(MAPK) activity and knockdown of EWS-Fli1 reduced fenretinide-induced mitochondrial permeabilization, cytochrome c release, caspase and PARP cleavage, consistent with the hypothesis that p38(MAPK) is critical for activation of the death cascade by fenretinide in ESFT cells. Fenretinide 80-91 mitogen-activated protein kinase 14 Homo sapiens 217-220 17700534-5 2008 Furthermore, inhibition of p38(MAPK) activity and knockdown of EWS-Fli1 reduced fenretinide-induced mitochondrial permeabilization, cytochrome c release, caspase and PARP cleavage, consistent with the hypothesis that p38(MAPK) is critical for activation of the death cascade by fenretinide in ESFT cells. Fenretinide 278-289 EWS RNA binding protein 1 Homo sapiens 63-66 17700534-6 2008 These data demonstrate that expression of EWS-Fli1 enhances fenretinide-induced cell death in ESFT and that this is effected at least in part through modulation of p38(MAPK) activity. Fenretinide 60-71 EWS RNA binding protein 1 Homo sapiens 42-45 17700534-6 2008 These data demonstrate that expression of EWS-Fli1 enhances fenretinide-induced cell death in ESFT and that this is effected at least in part through modulation of p38(MAPK) activity. Fenretinide 60-71 Fli-1 proto-oncogene, ETS transcription factor Homo sapiens 46-50 17955489-1 2008 Fenretinide, N-(4-hydroxyphenyl)retinamide (4-HPR) is an aminophenol-containing synthetic retinoid derivative of all-trans-retinoic acid, which is a potent chemopreventive and antiproliferative agent against various cancers. Fenretinide 0-11 haptoglobin-related protein Homo sapiens 46-49 17955489-1 2008 Fenretinide, N-(4-hydroxyphenyl)retinamide (4-HPR) is an aminophenol-containing synthetic retinoid derivative of all-trans-retinoic acid, which is a potent chemopreventive and antiproliferative agent against various cancers. Fenretinide 13-42 haptoglobin-related protein Homo sapiens 46-49 17883277-1 2007 N-(4-hydroxyphenyl)retinamide (fenretinide, 4-HPR) has been shown to be active toward many tumors without appreciable side effects. Fenretinide 0-29 haptoglobin-related protein Homo sapiens 46-49 17656682-6 2008 Since it has been previously reported that in vitro treatment with fenretinide induced ceramide in neuroblastoma cell lines, we decided to test this drug in vivo using our Cftr-knockout mice in an attempt to correct this newly identified defect in ceramide levels. Fenretinide 67-78 cystic fibrosis transmembrane conductance regulator Mus musculus 172-176 17656682-8 2008 We further assessed the biological effect of fenretinide on the ability of Cftr-knockout mice to combat lung infection with P. aeruginosa. Fenretinide 45-56 cystic fibrosis transmembrane conductance regulator Mus musculus 75-79 22275964-9 2008 Antithrombin-III showed a 4% borderline significant reduction in the fenretinide arm relative to placebo, irrespective of the HRT administration route (p = 0.055). Fenretinide 69-80 serpin family C member 1 Homo sapiens 0-16 17577665-0 2008 Inhibitors of EGFR signaling retard cytotoxicity of fenretinide in rat gliosarcoma cells. Fenretinide 52-63 epidermal growth factor receptor Rattus norvegicus 14-18 18166136-0 2007 Fenretinide-induced apoptosis of Huh-7 hepatocellular carcinoma is retinoic acid receptor beta dependent. Fenretinide 0-11 MIR7-3 host gene Homo sapiens 33-38 18166136-0 2007 Fenretinide-induced apoptosis of Huh-7 hepatocellular carcinoma is retinoic acid receptor beta dependent. Fenretinide 0-11 retinoic acid receptor beta Homo sapiens 67-94 18166136-5 2007 Transactivation assay and chromatin immunoprecipitation (ChIP) were conducted to evaluate the activation of RXRalpha/RARbeta pathway by fenretinide. Fenretinide 136-147 retinoid X receptor alpha Homo sapiens 108-116 18166136-5 2007 Transactivation assay and chromatin immunoprecipitation (ChIP) were conducted to evaluate the activation of RXRalpha/RARbeta pathway by fenretinide. Fenretinide 136-147 retinoic acid receptor beta Homo sapiens 117-124 18166136-7 2007 RESULTS: Our data revealed that fenretinide effectively induces apoptosis in Huh-7 and Hep3B cells. Fenretinide 32-43 MIR7-3 host gene Homo sapiens 77-82 18166136-8 2007 Gene expression analysis of nuclear receptors revealed that the basal and inducibility of retinoic acid receptor beta (RARbeta) expression positively correlate with the susceptibility of HCC cells to fenretinide treatment. Fenretinide 200-211 retinoic acid receptor beta Homo sapiens 90-117 18166136-8 2007 Gene expression analysis of nuclear receptors revealed that the basal and inducibility of retinoic acid receptor beta (RARbeta) expression positively correlate with the susceptibility of HCC cells to fenretinide treatment. Fenretinide 200-211 retinoic acid receptor beta Homo sapiens 119-126 18166136-9 2007 Furthermore, fenretinide transactivates the RXRalpha/RARbeta-mediated pathway and directly increases the transcriptional activity of RARbeta. Fenretinide 13-24 retinoid X receptor alpha Homo sapiens 44-52 18166136-9 2007 Furthermore, fenretinide transactivates the RXRalpha/RARbeta-mediated pathway and directly increases the transcriptional activity of RARbeta. Fenretinide 13-24 retinoic acid receptor beta Homo sapiens 53-60 18166136-9 2007 Furthermore, fenretinide transactivates the RXRalpha/RARbeta-mediated pathway and directly increases the transcriptional activity of RARbeta. Fenretinide 13-24 retinoic acid receptor beta Homo sapiens 133-140 18166136-10 2007 Knockdown of RARbeta mRNA expression significantly impairs fenretinide-induced apoptosis in Huh-7 cells. Fenretinide 59-70 retinoic acid receptor beta Homo sapiens 13-20 18166136-10 2007 Knockdown of RARbeta mRNA expression significantly impairs fenretinide-induced apoptosis in Huh-7 cells. Fenretinide 59-70 MIR7-3 host gene Homo sapiens 92-97 18166136-11 2007 CONCLUSION: Our findings reveal that endogenous expression of retinoids receptor RARbeta gene determines the susceptibility of HCC cells to fenretinide-induced apoptosis. Fenretinide 140-151 retinoic acid receptor beta Homo sapiens 81-88 18166136-12 2007 Our results also demonstrate fenretinide directly activates RARbeta and induces apoptosis in Huh-7 cells in a RARbeta-dependent manner. Fenretinide 29-40 retinoic acid receptor beta Homo sapiens 60-67 18166136-12 2007 Our results also demonstrate fenretinide directly activates RARbeta and induces apoptosis in Huh-7 cells in a RARbeta-dependent manner. Fenretinide 29-40 MIR7-3 host gene Homo sapiens 93-98 18166136-12 2007 Our results also demonstrate fenretinide directly activates RARbeta and induces apoptosis in Huh-7 cells in a RARbeta-dependent manner. Fenretinide 29-40 retinoic acid receptor beta Homo sapiens 110-117 17883277-1 2007 N-(4-hydroxyphenyl)retinamide (fenretinide, 4-HPR) has been shown to be active toward many tumors without appreciable side effects. Fenretinide 31-42 haptoglobin-related protein Homo sapiens 46-49 17213814-1 2007 Fenretinide (4-HPR) is a synthetic retinoid with antitumor activity, which induces apoptosis in cancer cell lines of different histotypes. Fenretinide 0-11 haptoglobin-related protein Homo sapiens 15-18 17283068-11 2007 Additionally, we found that treatment of SMS-KCNR cells with fenretinide inhibited desaturase activity in a dose-dependent manner. Fenretinide 61-72 Desaturase 1 Drosophila melanogaster 83-93 17471234-7 2007 E2F-1 suppresses extracellular signal-regulated kinase (ERK) phosphorylation through PAC1 and causes cancer cell death by apoptosis following treatment with a chemotherapeutic agent N-4-hydroxyphenylretinamide (4-HPR). Fenretinide 182-209 E2F transcription factor 1 L homeolog Xenopus laevis 0-5 17471234-7 2007 E2F-1 suppresses extracellular signal-regulated kinase (ERK) phosphorylation through PAC1 and causes cancer cell death by apoptosis following treatment with a chemotherapeutic agent N-4-hydroxyphenylretinamide (4-HPR). Fenretinide 182-209 mitogen-activated protein kinase 1 S homeolog Xenopus laevis 56-59 17471234-7 2007 E2F-1 suppresses extracellular signal-regulated kinase (ERK) phosphorylation through PAC1 and causes cancer cell death by apoptosis following treatment with a chemotherapeutic agent N-4-hydroxyphenylretinamide (4-HPR). Fenretinide 211-216 E2F transcription factor 1 L homeolog Xenopus laevis 0-5 17471234-7 2007 E2F-1 suppresses extracellular signal-regulated kinase (ERK) phosphorylation through PAC1 and causes cancer cell death by apoptosis following treatment with a chemotherapeutic agent N-4-hydroxyphenylretinamide (4-HPR). Fenretinide 211-216 mitogen-activated protein kinase 1 S homeolog Xenopus laevis 56-59 17273769-0 2007 Fenretinide up-regulates DR5/TRAIL-R2 expression via the induction of the transcription factor CHOP and combined treatment with fenretinide and TRAIL induces synergistic apoptosis in colon cancer cell lines. Fenretinide 0-11 TNF receptor superfamily member 10b Homo sapiens 25-28 17404501-4 2007 Different experimental approaches have indicated that 4HPR induces ER stress response: electron microscopy, which showed extensive ER dilation; splicing of the X-box binding protein 1 (XBP-1), a marker of unfolded protein response (UPR) activation; and quantitative real-time PCR and immunoblotting, which revealed the upregulation of several ER-stress associated mRNAs and proteins, including the chaperone heat shock protein HSPA1A. Fenretinide 54-58 X-box binding protein 1 Homo sapiens 160-183 17404501-4 2007 Different experimental approaches have indicated that 4HPR induces ER stress response: electron microscopy, which showed extensive ER dilation; splicing of the X-box binding protein 1 (XBP-1), a marker of unfolded protein response (UPR) activation; and quantitative real-time PCR and immunoblotting, which revealed the upregulation of several ER-stress associated mRNAs and proteins, including the chaperone heat shock protein HSPA1A. Fenretinide 54-58 X-box binding protein 1 Homo sapiens 185-190 17404501-4 2007 Different experimental approaches have indicated that 4HPR induces ER stress response: electron microscopy, which showed extensive ER dilation; splicing of the X-box binding protein 1 (XBP-1), a marker of unfolded protein response (UPR) activation; and quantitative real-time PCR and immunoblotting, which revealed the upregulation of several ER-stress associated mRNAs and proteins, including the chaperone heat shock protein HSPA1A. Fenretinide 54-58 heat shock protein family A (Hsp70) member 1A Homo sapiens 427-433 17404501-6 2007 Furthermore, siRNA-mediated silencing and chemical inhibition of HSPA1A, which exerts either pro- or anti-apoptotic effects, decreased 4HPR-induced apoptosis. Fenretinide 135-139 heat shock protein family A (Hsp70) member 1A Homo sapiens 65-71 17404501-7 2007 These results demonstrate that 4HPR induces ER stress and uncovered a pro-apoptotic role for HSPA1A in 4HPR-induced apoptosis. Fenretinide 31-35 heat shock protein family A (Hsp70) member 1A Homo sapiens 93-99 17216584-0 2007 Role of Noxa in p53-independent fenretinide-induced apoptosis of neuroectodermal tumours. Fenretinide 32-43 phorbol-12-myristate-13-acetate-induced protein 1 Homo sapiens 8-12 17216584-1 2007 Fenretinide-induced apoptosis of neuroectodermal tumour cells is mediated through generation of reactive oxygen species (ROS), endoplasmic reticulum (ER) stress, mitochondrial cytochrome c release and caspase activation. Fenretinide 0-11 cytochrome c, somatic Homo sapiens 176-188 17216584-3 2007 Noxa expression was induced by fenretinide in neuroblastoma and melanoma cells, including those with mutated p53, and this induction was abolished by antioxidants. Fenretinide 31-42 phorbol-12-myristate-13-acetate-induced protein 1 Homo sapiens 0-4 17216584-4 2007 Knockdown of p53 by RNA interference (RNAi) demonstrated upregulation of Noxa protein levels in response to fenretinide was p53-independent, although evidence suggested that Noxa may be transcriptionally regulated by p53. Fenretinide 108-119 tumor protein p53 Homo sapiens 13-16 17216584-4 2007 Knockdown of p53 by RNA interference (RNAi) demonstrated upregulation of Noxa protein levels in response to fenretinide was p53-independent, although evidence suggested that Noxa may be transcriptionally regulated by p53. Fenretinide 108-119 phorbol-12-myristate-13-acetate-induced protein 1 Homo sapiens 73-77 17216584-4 2007 Knockdown of p53 by RNA interference (RNAi) demonstrated upregulation of Noxa protein levels in response to fenretinide was p53-independent, although evidence suggested that Noxa may be transcriptionally regulated by p53. Fenretinide 108-119 tumor protein p53 Homo sapiens 124-127 17216584-4 2007 Knockdown of p53 by RNA interference (RNAi) demonstrated upregulation of Noxa protein levels in response to fenretinide was p53-independent, although evidence suggested that Noxa may be transcriptionally regulated by p53. Fenretinide 108-119 tumor protein p53 Homo sapiens 124-127 17216584-8 2007 RNAi-mediated down-regulation of Noxa inhibited apoptosis in response to fenretinide or thapsigargin, whereas apoptosis induced by cisplatin or temozolomide was unaffected. Fenretinide 73-84 phorbol-12-myristate-13-acetate-induced protein 1 Homo sapiens 33-37 17436572-4 2007 In this report we present data to show that fenretinide (a synthetic retinoid) potentiates the apoptotic effects of parthenolide (a drug that inhibits the activation of NF-kappa B) and BAY 11-7085 (an inhibitor of I-kappa B-alpha kinase). Fenretinide 44-55 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 169-179 17436572-4 2007 In this report we present data to show that fenretinide (a synthetic retinoid) potentiates the apoptotic effects of parthenolide (a drug that inhibits the activation of NF-kappa B) and BAY 11-7085 (an inhibitor of I-kappa B-alpha kinase). Fenretinide 44-55 nuclear factor of kappa light polypeptide gene enhancer in B cells inhibitor, alpha Mus musculus 214-229 17436572-5 2007 This potentiation of apoptosis by fenretinide is seen in the p53-deficient, deoxyadenosine-resistant L1210 cells, but not in the parental L1210 cells that express a mutant p53. Fenretinide 34-45 transformation related protein 53, pseudogene Mus musculus 61-64 17436572-5 2007 This potentiation of apoptosis by fenretinide is seen in the p53-deficient, deoxyadenosine-resistant L1210 cells, but not in the parental L1210 cells that express a mutant p53. Fenretinide 34-45 transformation related protein 53, pseudogene Mus musculus 172-175 17436572-7 2007 These data strongly suggest that fenretinide activates or inhibits some step or pathway that interacts with the inhibition of NF-kappa B activation required for the apoptotic response. Fenretinide 33-44 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 126-136 17146731-5 2007 The apparent permeability coefficient (P(app)) of fenretinide across Caco-2 monolayers in the presence of bovine serum albumin (BSA) in the receiver was determined. Fenretinide 50-61 albumin Homo sapiens 113-126 17454142-1 2007 We have previously reported that, in leukemia cells, the cytotoxicity of the anticancer agent N-(4-hydroxyphenyl)retinamide (4-HPR) is mediated by mitochondria-derived reactive oxygen species (ROS) and cardiolipin peroxidation. Fenretinide 94-123 haptoglobin-related protein Homo sapiens 127-130 17353921-4 2007 The retinoid analogue fenretinide [N-(4-hydroxyphenyl) retinamide] is a new cancer preventive and chemotherapeutic drug, that induces apoptosis of some cancer cell types via oxidative stress, accompanied by induction of an ER stress-related transcription factor, GADD153. Fenretinide 22-33 DNA damage inducible transcript 3 Homo sapiens 263-270 17353921-4 2007 The retinoid analogue fenretinide [N-(4-hydroxyphenyl) retinamide] is a new cancer preventive and chemotherapeutic drug, that induces apoptosis of some cancer cell types via oxidative stress, accompanied by induction of an ER stress-related transcription factor, GADD153. Fenretinide 35-65 DNA damage inducible transcript 3 Homo sapiens 263-270 17353921-6 2007 The ER stress genes ERdj5, ERp57, GRP78, calreticulin and calnexin were induced in neuroectodermal tumour cells by fenretinide. Fenretinide 115-126 DnaJ heat shock protein family (Hsp40) member C10 Homo sapiens 20-25 17353921-6 2007 The ER stress genes ERdj5, ERp57, GRP78, calreticulin and calnexin were induced in neuroectodermal tumour cells by fenretinide. Fenretinide 115-126 protein disulfide isomerase family A member 3 Homo sapiens 27-32 17353921-6 2007 The ER stress genes ERdj5, ERp57, GRP78, calreticulin and calnexin were induced in neuroectodermal tumour cells by fenretinide. Fenretinide 115-126 heat shock protein family A (Hsp70) member 5 Homo sapiens 34-39 17353921-6 2007 The ER stress genes ERdj5, ERp57, GRP78, calreticulin and calnexin were induced in neuroectodermal tumour cells by fenretinide. Fenretinide 115-126 calreticulin Homo sapiens 41-53 17353921-6 2007 The ER stress genes ERdj5, ERp57, GRP78, calreticulin and calnexin were induced in neuroectodermal tumour cells by fenretinide. Fenretinide 115-126 calnexin Homo sapiens 58-66 17353921-7 2007 In contrast to the apoptosis-inducing chemotherapeutic drugs vincristine and temozolomide, fenretinide induced the phosphorylation of eIF2alpha, expression of ATF4 and splicing of XBP-1 mRNA, events that define ER stress. Fenretinide 91-102 eukaryotic translation initiation factor 2A Homo sapiens 134-143 17353921-7 2007 In contrast to the apoptosis-inducing chemotherapeutic drugs vincristine and temozolomide, fenretinide induced the phosphorylation of eIF2alpha, expression of ATF4 and splicing of XBP-1 mRNA, events that define ER stress. Fenretinide 91-102 activating transcription factor 4 Homo sapiens 159-163 17353921-7 2007 In contrast to the apoptosis-inducing chemotherapeutic drugs vincristine and temozolomide, fenretinide induced the phosphorylation of eIF2alpha, expression of ATF4 and splicing of XBP-1 mRNA, events that define ER stress. Fenretinide 91-102 X-box binding protein 1 Homo sapiens 180-185 17353921-10 2007 Knockdown of ERp57 or ERdj5 by RNA interference in these cells increased the apoptotic response to fenretinide. Fenretinide 99-110 protein disulfide isomerase family A member 3 Homo sapiens 13-18 17353921-10 2007 Knockdown of ERp57 or ERdj5 by RNA interference in these cells increased the apoptotic response to fenretinide. Fenretinide 99-110 DnaJ heat shock protein family (Hsp40) member C10 Homo sapiens 22-27 17216584-9 2007 These data demonstrate the importance of Noxa induction in determining the apoptotic response to fenretinide and emphasise the role of Noxa in p53-independent apoptosis. Fenretinide 97-108 phorbol-12-myristate-13-acetate-induced protein 1 Homo sapiens 41-45 17273769-4 2007 In this study, we report that fenretinide induces death receptor 5 (DR5)/TRAIL-R2 up-regulation via the induction of the transcription factor CHOP in colon cancer cell lines. Fenretinide 30-41 TNF receptor superfamily member 10b Homo sapiens 73-81 17273769-4 2007 In this study, we report that fenretinide induces death receptor 5 (DR5)/TRAIL-R2 up-regulation via the induction of the transcription factor CHOP in colon cancer cell lines. Fenretinide 30-41 DNA damage inducible transcript 3 Homo sapiens 142-146 17273769-5 2007 Fenretinide induced DR5 expression at protein and mRNA levels. Fenretinide 0-11 TNF receptor superfamily member 10b Homo sapiens 20-23 17273769-6 2007 Furthermore, fenretinide increased DR5 promoter activity and the enhanced activity decreased by mutation of the CHOP binding site. Fenretinide 13-24 TNF receptor superfamily member 10b Homo sapiens 35-38 17273769-6 2007 Furthermore, fenretinide increased DR5 promoter activity and the enhanced activity decreased by mutation of the CHOP binding site. Fenretinide 13-24 DNA damage inducible transcript 3 Homo sapiens 112-116 17273769-7 2007 CHOP was also up-regulated by fenretinide at the promoter level. Fenretinide 30-41 DNA damage inducible transcript 3 Homo sapiens 0-4 17273769-10 2007 Fenretinide and TRAIL cooperatively activated caspase-3, -8, -10 and -9 and cleavage of Bid and PARP, and this activation was also blocked in the presence of DR5/Fc chimeric protein. Fenretinide 0-11 caspase 3 Homo sapiens 46-71 17273769-10 2007 Fenretinide and TRAIL cooperatively activated caspase-3, -8, -10 and -9 and cleavage of Bid and PARP, and this activation was also blocked in the presence of DR5/Fc chimeric protein. Fenretinide 0-11 BH3 interacting domain death agonist Homo sapiens 88-91 17273769-10 2007 Fenretinide and TRAIL cooperatively activated caspase-3, -8, -10 and -9 and cleavage of Bid and PARP, and this activation was also blocked in the presence of DR5/Fc chimeric protein. Fenretinide 0-11 collagen type XI alpha 2 chain Homo sapiens 96-100 17273769-10 2007 Fenretinide and TRAIL cooperatively activated caspase-3, -8, -10 and -9 and cleavage of Bid and PARP, and this activation was also blocked in the presence of DR5/Fc chimeric protein. Fenretinide 0-11 TNF receptor superfamily member 10b Homo sapiens 158-161 17273769-11 2007 These results indicate that fenretinide-induced apoptosis is sensitized by TRAIL. Fenretinide 28-39 TNF superfamily member 10 Homo sapiens 75-80 17273769-0 2007 Fenretinide up-regulates DR5/TRAIL-R2 expression via the induction of the transcription factor CHOP and combined treatment with fenretinide and TRAIL induces synergistic apoptosis in colon cancer cell lines. Fenretinide 0-11 TNF receptor superfamily member 10b Homo sapiens 29-37 17273769-0 2007 Fenretinide up-regulates DR5/TRAIL-R2 expression via the induction of the transcription factor CHOP and combined treatment with fenretinide and TRAIL induces synergistic apoptosis in colon cancer cell lines. Fenretinide 0-11 DNA damage inducible transcript 3 Homo sapiens 95-99 17273769-0 2007 Fenretinide up-regulates DR5/TRAIL-R2 expression via the induction of the transcription factor CHOP and combined treatment with fenretinide and TRAIL induces synergistic apoptosis in colon cancer cell lines. Fenretinide 0-11 TNF superfamily member 10 Homo sapiens 29-34 17273769-3 2007 Oxidative stress induced by fenretinide has been shown to mediate apoptosis through a mitochondrial pathway by the induction of a transcription factor CCAAT/enhancer binding protein homologous protein (CHOP) and Bak. Fenretinide 28-39 DNA damage inducible transcript 3 Homo sapiens 151-200 17273769-3 2007 Oxidative stress induced by fenretinide has been shown to mediate apoptosis through a mitochondrial pathway by the induction of a transcription factor CCAAT/enhancer binding protein homologous protein (CHOP) and Bak. Fenretinide 28-39 DNA damage inducible transcript 3 Homo sapiens 202-206 17273769-4 2007 In this study, we report that fenretinide induces death receptor 5 (DR5)/TRAIL-R2 up-regulation via the induction of the transcription factor CHOP in colon cancer cell lines. Fenretinide 30-41 TNF receptor superfamily member 10b Homo sapiens 50-66 17273769-4 2007 In this study, we report that fenretinide induces death receptor 5 (DR5)/TRAIL-R2 up-regulation via the induction of the transcription factor CHOP in colon cancer cell lines. Fenretinide 30-41 TNF receptor superfamily member 10b Homo sapiens 68-71 17112722-0 2007 Solid phase-assisted synthesis and screening of a small library of N-(4-hydroxyphenyl)retinamide (4-HPR) analogs. Fenretinide 67-96 haptoglobin-related protein Homo sapiens 100-103 17112722-1 2007 Using solid phase-assisted synthesis and purification, a 49 member library of analogs of the mammary tumor chemopreventive retinoid N-(4-hydroxyphenyl)retinamide (4-HPR) has been prepared. Fenretinide 132-161 haptoglobin-related protein Homo sapiens 165-168 17235241-4 2007 Annexin V was induced by nimesulide, 4-hydroxyphenylretinamide, and difluoromethylornithine in ultraviolet-B-treated radial growth-phase-like melanoma cells. Fenretinide 37-62 annexin A5 Homo sapiens 0-9 16950796-5 2007 Among the synthetic agents, oltipraz and N-(4-hydroxyphenyl) retinamide (4-HPR) also increased MGMT expression, albeit to a lesser extent. Fenretinide 41-71 haptoglobin-related protein Homo sapiens 75-78 16950796-5 2007 Among the synthetic agents, oltipraz and N-(4-hydroxyphenyl) retinamide (4-HPR) also increased MGMT expression, albeit to a lesser extent. Fenretinide 41-71 O-6-methylguanine-DNA methyltransferase Homo sapiens 95-99 17159502-6 2007 Western blot analysis further showed down-regulation of procaspase-3, X-linked inhibitor of apoptosis protein and poly(ADP-ribose) polymerase cleavage in Bel-7402 cells treated with 15 mumol/l fenretinide for 48 h. Overexpression of p53 was observed in a time-dependent manner, along with a decrease in the Bcl-2/Bax ratio. Fenretinide 193-204 caspase 3 Homo sapiens 56-68 17159502-6 2007 Western blot analysis further showed down-regulation of procaspase-3, X-linked inhibitor of apoptosis protein and poly(ADP-ribose) polymerase cleavage in Bel-7402 cells treated with 15 mumol/l fenretinide for 48 h. Overexpression of p53 was observed in a time-dependent manner, along with a decrease in the Bcl-2/Bax ratio. Fenretinide 193-204 tumor protein p53 Homo sapiens 233-236 17159502-6 2007 Western blot analysis further showed down-regulation of procaspase-3, X-linked inhibitor of apoptosis protein and poly(ADP-ribose) polymerase cleavage in Bel-7402 cells treated with 15 mumol/l fenretinide for 48 h. Overexpression of p53 was observed in a time-dependent manner, along with a decrease in the Bcl-2/Bax ratio. Fenretinide 193-204 BCL2 apoptosis regulator Homo sapiens 307-312 17159502-6 2007 Western blot analysis further showed down-regulation of procaspase-3, X-linked inhibitor of apoptosis protein and poly(ADP-ribose) polymerase cleavage in Bel-7402 cells treated with 15 mumol/l fenretinide for 48 h. Overexpression of p53 was observed in a time-dependent manner, along with a decrease in the Bcl-2/Bax ratio. Fenretinide 193-204 BCL2 associated X, apoptosis regulator Homo sapiens 313-316 17237288-5 2007 We found that 4HPR causes lysosomal membrane permeabilization and cytosolic relocation of cathepsin D. Fenretinide 14-18 cathepsin D Homo sapiens 90-101 17044020-0 2006 Cell death induced by N-(4-hydroxyphenyl)retinamide in human epidermal keratinocytes is modulated by TGF-beta and diminishes during the progression of squamous cell carcinoma. Fenretinide 22-51 transforming growth factor beta 1 Homo sapiens 101-109 17302189-8 2007 In one study, fenretinide (4-HPR) delayed development of ovarian cancer in women at increased risk of developing breast and ovarian cancer. Fenretinide 14-25 haptoglobin-related protein Homo sapiens 29-32 17044020-1 2006 It has been demonstrated that the chemopreventive agent N-(4-hydroxyphenyl)retinamide (4-HPR) induces apoptotic cell death, but recent data has suggested that late stage/recurrent tumours lose their response to 4-HPR-induced cell death by mechanisms that are unknown. Fenretinide 56-85 haptoglobin-related protein Homo sapiens 89-92 17044020-1 2006 It has been demonstrated that the chemopreventive agent N-(4-hydroxyphenyl)retinamide (4-HPR) induces apoptotic cell death, but recent data has suggested that late stage/recurrent tumours lose their response to 4-HPR-induced cell death by mechanisms that are unknown. Fenretinide 56-85 haptoglobin-related protein Homo sapiens 213-216 16972258-0 2006 N-(4-hydroxyphenyl)retinamide induces apoptosis in human retinal pigment epithelial cells: retinoic acid receptors regulate apoptosis, reactive oxygen species generation, and the expression of heme oxygenase-1 and Gadd153. Fenretinide 0-29 heme oxygenase 1 Homo sapiens 193-209 17001693-11 2006 Fenretinide, a novel synthetic retinoic acid (RA) analog lowers RBP4 in glucose intolerant obese mice. Fenretinide 0-11 retinol binding protein 4, plasma Mus musculus 64-68 16972258-0 2006 N-(4-hydroxyphenyl)retinamide induces apoptosis in human retinal pigment epithelial cells: retinoic acid receptors regulate apoptosis, reactive oxygen species generation, and the expression of heme oxygenase-1 and Gadd153. Fenretinide 0-29 DNA damage inducible transcript 3 Homo sapiens 214-221 16972258-8 2006 Pyrrolidine dithiocarbamate (PDTC), a free-radical scavenger, inhibited 4HPR-induced ROS generation, the expression of its downstream mediator, Gadd153, and apoptosis in the pretreated cells. Fenretinide 72-76 DNA damage inducible transcript 3 Homo sapiens 144-151 16979119-4 2006 As fenretinide is a ceramide up-regulating drug that inhibits the activation of the pro-inflammatory transcriptional factor, nuclear factor (NF)-kappaB, the impact of fenretinide on unstimulated and tumor necrosis factor (TNF)-alpha stimulated production of NF-kappaB-dependent interleukin (IL)-8 was studied in immortalized wild-type (non-CF; 9HTEo-) and mutant DeltaF508 CFTR (CF; CFTE29o-) tracheal epithelial cells. Fenretinide 3-14 CF transmembrane conductance regulator Homo sapiens 340-342 16979119-4 2006 As fenretinide is a ceramide up-regulating drug that inhibits the activation of the pro-inflammatory transcriptional factor, nuclear factor (NF)-kappaB, the impact of fenretinide on unstimulated and tumor necrosis factor (TNF)-alpha stimulated production of NF-kappaB-dependent interleukin (IL)-8 was studied in immortalized wild-type (non-CF; 9HTEo-) and mutant DeltaF508 CFTR (CF; CFTE29o-) tracheal epithelial cells. Fenretinide 3-14 CF transmembrane conductance regulator Homo sapiens 373-377 16979119-0 2006 Inhibition of IL-8 release from CFTR-deficient lung epithelial cells following pre-treatment with fenretinide. Fenretinide 98-109 C-X-C motif chemokine ligand 8 Homo sapiens 14-18 16979119-6 2006 Clinically relevant concentrations of fenretinide (1.25, 2.5 and 5 microM) inhibited TNF-alpha-induced IL-8 production of CF cells by up to 73% but had no effect or increased the IL-8 production in non-CF cells. Fenretinide 38-49 tumor necrosis factor Homo sapiens 85-94 16979119-0 2006 Inhibition of IL-8 release from CFTR-deficient lung epithelial cells following pre-treatment with fenretinide. Fenretinide 98-109 CF transmembrane conductance regulator Homo sapiens 32-36 16979119-4 2006 As fenretinide is a ceramide up-regulating drug that inhibits the activation of the pro-inflammatory transcriptional factor, nuclear factor (NF)-kappaB, the impact of fenretinide on unstimulated and tumor necrosis factor (TNF)-alpha stimulated production of NF-kappaB-dependent interleukin (IL)-8 was studied in immortalized wild-type (non-CF; 9HTEo-) and mutant DeltaF508 CFTR (CF; CFTE29o-) tracheal epithelial cells. Fenretinide 3-14 tumor necrosis factor Homo sapiens 199-232 16979119-6 2006 Clinically relevant concentrations of fenretinide (1.25, 2.5 and 5 microM) inhibited TNF-alpha-induced IL-8 production of CF cells by up to 73% but had no effect or increased the IL-8 production in non-CF cells. Fenretinide 38-49 C-X-C motif chemokine ligand 8 Homo sapiens 103-107 16979119-7 2006 Although fenretinide treatment was associated with a higher intracellular ceramide content in the mutant DeltaF508 CFTR cells, the fenretinide-mediated decrease in IL-8 secretion was not consistently explained by changes in the intracellular content of this sphingolipid. Fenretinide 9-20 CF transmembrane conductance regulator Homo sapiens 115-119 16979119-7 2006 Although fenretinide treatment was associated with a higher intracellular ceramide content in the mutant DeltaF508 CFTR cells, the fenretinide-mediated decrease in IL-8 secretion was not consistently explained by changes in the intracellular content of this sphingolipid. Fenretinide 131-142 C-X-C motif chemokine ligand 8 Homo sapiens 164-168 16979119-9 2006 The fenretinide mediated decrease in IL-8 release in CF cells under TNF-alpha stimulated conditions presents the possibility that the lung inflammation in CF could be attenuated via low dose fenretinide treatment. Fenretinide 4-15 C-X-C motif chemokine ligand 8 Homo sapiens 37-41 16979119-9 2006 The fenretinide mediated decrease in IL-8 release in CF cells under TNF-alpha stimulated conditions presents the possibility that the lung inflammation in CF could be attenuated via low dose fenretinide treatment. Fenretinide 4-15 tumor necrosis factor Homo sapiens 68-77 16979119-9 2006 The fenretinide mediated decrease in IL-8 release in CF cells under TNF-alpha stimulated conditions presents the possibility that the lung inflammation in CF could be attenuated via low dose fenretinide treatment. Fenretinide 191-202 C-X-C motif chemokine ligand 8 Homo sapiens 37-41 16979119-9 2006 The fenretinide mediated decrease in IL-8 release in CF cells under TNF-alpha stimulated conditions presents the possibility that the lung inflammation in CF could be attenuated via low dose fenretinide treatment. Fenretinide 191-202 tumor necrosis factor Homo sapiens 68-77 17018636-0 2006 Involvement of mitochondrial and Akt signaling pathways in augmented apoptosis induced by a combination of low doses of celecoxib and N-(4-hydroxyphenyl) retinamide in premalignant human bronchial epithelial cells. Fenretinide 134-164 AKT serine/threonine kinase 1 Homo sapiens 33-36 17110918-0 2006 Fenretinide-induced neuronal differentiation of ARPE-19 human retinal pigment epithelial cells is associated with the differential expression of Hsp70, 14-3-3, pax-6, tubulin beta-III, NSE, and bag-1 proteins. Fenretinide 0-11 paired box 6 Homo sapiens 160-165 17110918-0 2006 Fenretinide-induced neuronal differentiation of ARPE-19 human retinal pigment epithelial cells is associated with the differential expression of Hsp70, 14-3-3, pax-6, tubulin beta-III, NSE, and bag-1 proteins. Fenretinide 0-11 tubulin beta 3 class III Homo sapiens 167-183 17110918-0 2006 Fenretinide-induced neuronal differentiation of ARPE-19 human retinal pigment epithelial cells is associated with the differential expression of Hsp70, 14-3-3, pax-6, tubulin beta-III, NSE, and bag-1 proteins. Fenretinide 0-11 enolase 2 Homo sapiens 185-188 17110918-0 2006 Fenretinide-induced neuronal differentiation of ARPE-19 human retinal pigment epithelial cells is associated with the differential expression of Hsp70, 14-3-3, pax-6, tubulin beta-III, NSE, and bag-1 proteins. Fenretinide 0-11 BAG cochaperone 1 Homo sapiens 194-199 17110918-6 2006 Fenretinide induced neuronal differentiation of ARPE-19 cells led to a decrease in pax-6 protein and an increase in tubulin beta-III protein expression after 5 days fenretinide treatment. Fenretinide 0-11 paired box 6 Homo sapiens 83-88 17110918-6 2006 Fenretinide induced neuronal differentiation of ARPE-19 cells led to a decrease in pax-6 protein and an increase in tubulin beta-III protein expression after 5 days fenretinide treatment. Fenretinide 0-11 tubulin beta 3 class III Homo sapiens 116-132 17110918-8 2006 We also found a time-dependent increase in Hsp70 protein expression in ARPE-19 cells treated with fenretinide. Fenretinide 98-109 heat shock protein family A (Hsp70) member 4 Homo sapiens 43-48 17110918-10 2006 CONCLUSIONS: The fenretinide-induced neuronal differentiation of ARPE-19 cells is associated with an increase in expression of the neuronal specific protein tubulin beta-III, and a decrease in expression of the progenitor cell marker pax-6. Fenretinide 17-28 tubulin beta 3 class III Homo sapiens 157-173 17110918-10 2006 CONCLUSIONS: The fenretinide-induced neuronal differentiation of ARPE-19 cells is associated with an increase in expression of the neuronal specific protein tubulin beta-III, and a decrease in expression of the progenitor cell marker pax-6. Fenretinide 17-28 paired box 6 Homo sapiens 234-239 16850162-3 2006 Diverse signaling molecules including reactive oxygen species, ceramide, and ganglioside GD3 can mediate apoptosis induction by fenretinide in transformed, premalignant, and malignant cells. Fenretinide 128-139 GRDX Homo sapiens 89-92 17018636-8 2006 Furthermore, combinations of celecoxib and 4HPR suppressed the phosphorylation levels of serine/threonine kinase Akt and its substrate glycogen synthase kinase-3beta more effectively than the single agents did. Fenretinide 43-47 AKT serine/threonine kinase 1 Homo sapiens 113-116 17018636-8 2006 Furthermore, combinations of celecoxib and 4HPR suppressed the phosphorylation levels of serine/threonine kinase Akt and its substrate glycogen synthase kinase-3beta more effectively than the single agents did. Fenretinide 43-47 glycogen synthase kinase 3 beta Homo sapiens 135-165 17018636-9 2006 Accordingly, overexpression of constitutively active Akt protected bronchial epithelial cells from undergoing apoptosis after incubation with both celecoxib and 4HPR. Fenretinide 161-165 AKT serine/threonine kinase 1 Homo sapiens 53-56 17035399-1 2006 PURPOSE: To evaluate study feasibility, toxicity, drug concentrations, and activity of escalating doses of the synthetic retinoid fenretinide [N-(4-hydroxyphenyl)retinamide (4-HPR)] in ovarian cancer by measuring serum CA125 and cytomorphometric biomarkers in cancer cells collected from ascitic fluid before and after treatment. Fenretinide 130-141 mucin 16, cell surface associated Homo sapiens 219-224 16675469-1 2006 N-(4-hydroxyphenyl)retinamide (4-HPR), a synthetic retinoid is under clinical evaluation as a therapeutic agent in a variety of cancers. Fenretinide 0-29 haptoglobin-related protein Homo sapiens 33-36 16520989-0 2006 Hypoxia-mediated fenretinide (4-HPR) resistance in childhood acute lymphoblastic leukemia cells. Fenretinide 17-28 haptoglobin-related protein Homo sapiens 32-35 16520989-1 2006 PURPOSES: N-(4-Hydroxyphenyl)-retinamide (4-HPR, Fenretinide) is a synthetic retinoid with cytotoxicity in acute lymphoblastic leukemia (ALL) cell lines. Fenretinide 10-40 haptoglobin-related protein Homo sapiens 44-47 16520989-1 2006 PURPOSES: N-(4-Hydroxyphenyl)-retinamide (4-HPR, Fenretinide) is a synthetic retinoid with cytotoxicity in acute lymphoblastic leukemia (ALL) cell lines. Fenretinide 49-60 haptoglobin-related protein Homo sapiens 44-47 16837623-8 2006 Direct comparison of other inhibitors such as fenretinide and 13-cis-retinoic acid showed multiple advantages of Ret-NH2 and its amides, including a higher potency, specificity, and lower transcription activation. Fenretinide 46-57 ret proto-oncogene Mus musculus 113-116 16671099-1 2006 N-(4-Hydroxyphenyl)retinamide (4-HPR) is a nonclassical retinoid with cancer preventive effects in vivo and antiproliferative and apoptotic activities in vitro. Fenretinide 0-29 haptoglobin-related protein Homo sapiens 33-36 16766039-7 2006 The miniaturised method was used to study the binding of three different ligands (4-HPR, arotinoid, warfarinyl palmitate) modelled to bind to betaLG. Fenretinide 82-87 beta-lactoglobulin Bos taurus 142-148 16407847-8 2006 PD98059, an MEK1/2 inhibitor, also suppressed ERK1/2 activation and apoptosis induced by 4HPR. Fenretinide 89-93 mitogen-activated protein kinase kinase 1 Homo sapiens 12-18 16407847-8 2006 PD98059, an MEK1/2 inhibitor, also suppressed ERK1/2 activation and apoptosis induced by 4HPR. Fenretinide 89-93 mitogen-activated protein kinase 3 Homo sapiens 46-52 16407847-2 2006 As ROS can activate MAP kinases and protein kinase C (PKC), we examined the role of such enzymes in 4HPR-induced apoptosis in HNSCC UMSCC22B cells. Fenretinide 100-104 proline rich transmembrane protein 2 Homo sapiens 54-57 16407847-10 2006 These data indicate that 4HPR-induced apoptosis is triggered by ROS increase, leading to the activation of the mitogen-activated protein serine/threonine kinases JNK, p38, PKC and ERK, and subsequent apoptosis. Fenretinide 25-29 mitogen-activated protein kinase 8 Homo sapiens 162-165 16407847-5 2006 SP600125, a JNK inhibitor, suppressed 4HPR-induced c-Jun phosphorylation, cytochrome c release from mitochondria and apoptosis. Fenretinide 38-42 mitogen-activated protein kinase 8 Homo sapiens 12-15 16407847-5 2006 SP600125, a JNK inhibitor, suppressed 4HPR-induced c-Jun phosphorylation, cytochrome c release from mitochondria and apoptosis. Fenretinide 38-42 cytochrome c, somatic Homo sapiens 74-86 16407847-10 2006 These data indicate that 4HPR-induced apoptosis is triggered by ROS increase, leading to the activation of the mitogen-activated protein serine/threonine kinases JNK, p38, PKC and ERK, and subsequent apoptosis. Fenretinide 25-29 mitogen-activated protein kinase 1 Homo sapiens 167-170 16407847-10 2006 These data indicate that 4HPR-induced apoptosis is triggered by ROS increase, leading to the activation of the mitogen-activated protein serine/threonine kinases JNK, p38, PKC and ERK, and subsequent apoptosis. Fenretinide 25-29 proline rich transmembrane protein 2 Homo sapiens 172-175 16407847-10 2006 These data indicate that 4HPR-induced apoptosis is triggered by ROS increase, leading to the activation of the mitogen-activated protein serine/threonine kinases JNK, p38, PKC and ERK, and subsequent apoptosis. Fenretinide 25-29 mitogen-activated protein kinase 1 Homo sapiens 180-183 16127422-0 2005 Cyclooxygenase-2 protein reduces tamoxifen and N-(4-hydroxyphenyl)retinamide inhibitory effects in breast cancer cells. Fenretinide 47-76 prostaglandin-endoperoxide synthase 2 Homo sapiens 0-16 16091745-0 2005 BBC3 mediates fenretinide-induced cell death in neuroblastoma. Fenretinide 14-25 BCL2 binding component 3 Homo sapiens 0-4 16091745-1 2005 Fenretinide (4-HPR) is a synthetic retinoid whose apoptosis-inducing effects have been demonstrated in many tumor types. Fenretinide 0-11 haptoglobin-related protein Homo sapiens 15-18 16403262-1 2005 The retinoid N-4-hydroxyphenyl retinamide (4-HPR also known as fenretinide), a synthetic derivative of all trans retinoic acid (ATRA), has shown as an efficient chemopreventive, chemotherapeutic agent and a potent inducer of apoptosis in various cancer cell types in vitro, including leukemic cells. Fenretinide 13-41 haptoglobin-related protein Homo sapiens 45-48 16403262-1 2005 The retinoid N-4-hydroxyphenyl retinamide (4-HPR also known as fenretinide), a synthetic derivative of all trans retinoic acid (ATRA), has shown as an efficient chemopreventive, chemotherapeutic agent and a potent inducer of apoptosis in various cancer cell types in vitro, including leukemic cells. Fenretinide 63-74 haptoglobin-related protein Homo sapiens 45-48 16540676-1 2006 4-oxo-N-(4-hydroxyphenyl)retinamide (4-oxo-4-HPR) is a recently identified metabolite of fenretinide (4-HPR). Fenretinide 89-100 haptoglobin-related protein Homo sapiens 45-48 16540676-1 2006 4-oxo-N-(4-hydroxyphenyl)retinamide (4-oxo-4-HPR) is a recently identified metabolite of fenretinide (4-HPR). Fenretinide 89-100 haptoglobin-related protein Homo sapiens 104-107 16302003-6 2006 Furthermore, we found that pharmacological intervention with low micromolar concentrations of N-(4-hydroxyphenyl)retinamide (4-HPR), which downmodulates cyclin D1, induced G1 arrest and apoptosis in rhabdoid cell lines. Fenretinide 94-123 haptoglobin-related protein Homo sapiens 127-130 16302003-6 2006 Furthermore, we found that pharmacological intervention with low micromolar concentrations of N-(4-hydroxyphenyl)retinamide (4-HPR), which downmodulates cyclin D1, induced G1 arrest and apoptosis in rhabdoid cell lines. Fenretinide 94-123 cyclin D1 Homo sapiens 153-162 16375981-9 2006 This effect of fenretinide appears to be specific for virus as the endosomal accumulation of gp120, transferrin and horse-radish peroxidase was not increased. Fenretinide 15-26 inhibitor of carbonic anhydrase Equus caballus 100-111 16399227-1 2006 N-(4-hydroxyphenyl) retinamide (4-HPR, fenretinide) a synthetic retinoid is in clinical trials for the treatment of several malignancies. Fenretinide 0-30 haptoglobin-related protein Homo sapiens 34-37 16399227-1 2006 N-(4-hydroxyphenyl) retinamide (4-HPR, fenretinide) a synthetic retinoid is in clinical trials for the treatment of several malignancies. Fenretinide 39-50 haptoglobin-related protein Homo sapiens 34-37 16399227-6 2006 Fenretinide-induced decrease in cell viability was in part due to activation of caspase-3 dependent cell death, which was further supported by the cleavage of poly(ADP-ribose) polymerase-1 (PARP-1), a caspase-3 substrate. Fenretinide 0-11 caspase 3 Homo sapiens 80-89 16399227-6 2006 Fenretinide-induced decrease in cell viability was in part due to activation of caspase-3 dependent cell death, which was further supported by the cleavage of poly(ADP-ribose) polymerase-1 (PARP-1), a caspase-3 substrate. Fenretinide 0-11 poly(ADP-ribose) polymerase 1 Homo sapiens 159-188 16399227-6 2006 Fenretinide-induced decrease in cell viability was in part due to activation of caspase-3 dependent cell death, which was further supported by the cleavage of poly(ADP-ribose) polymerase-1 (PARP-1), a caspase-3 substrate. Fenretinide 0-11 poly(ADP-ribose) polymerase 1 Homo sapiens 190-196 16399227-6 2006 Fenretinide-induced decrease in cell viability was in part due to activation of caspase-3 dependent cell death, which was further supported by the cleavage of poly(ADP-ribose) polymerase-1 (PARP-1), a caspase-3 substrate. Fenretinide 0-11 caspase 3 Homo sapiens 201-210 16382122-5 2006 After a median follow-up of 40 months, there was a reduction of 13%, 2%, 20%, and 1% in IGF-I levels for patients on tamoxifen, fenretinide, tamoxifen plus fenretinide, and placebo, respectively. Fenretinide 128-139 insulin like growth factor 1 Homo sapiens 88-93 16382122-5 2006 After a median follow-up of 40 months, there was a reduction of 13%, 2%, 20%, and 1% in IGF-I levels for patients on tamoxifen, fenretinide, tamoxifen plus fenretinide, and placebo, respectively. Fenretinide 156-167 insulin like growth factor 1 Homo sapiens 88-93 17024972-0 2006 Cholecalciferol (vitamin D3) and the retinoid N-(4-hydroxyphenyl)retinamide (4-HPR) are synergistic for chemoprevention of prostate cancer. Fenretinide 46-75 haptoglobin-related protein Homo sapiens 79-82 17024972-6 2006 This study examines the effects of the synthetic retinoid N-(4-hydroxyphenyl)retinamide (4-HPR) in combination with cholecalciferol (vitamin D3) on growth, and on the expression of vimentin, matrix metalloproteinase-2 (MMP-2), and retinoid and vitamin D receptor expression, using the non-tumorigenic, human prostate epithelial cell line RWPE-1. Fenretinide 58-87 haptoglobin-related protein Homo sapiens 91-94 16127422-6 2005 MCF-7/COX-2 cells were also resistant to the retinoid N-(4-hydroxyphenyl)retinamide (4-HPR) but not to its analog, all-trans retinoic acid. Fenretinide 54-83 prostaglandin-endoperoxide synthase 2 Homo sapiens 6-11 16127422-6 2005 MCF-7/COX-2 cells were also resistant to the retinoid N-(4-hydroxyphenyl)retinamide (4-HPR) but not to its analog, all-trans retinoic acid. Fenretinide 54-83 haptoglobin-related protein Homo sapiens 87-90 16230421-0 2005 N-(4-hydroxyphenyl)retinamide inhibits invasion, suppresses osteoclastogenesis, and potentiates apoptosis through down-regulation of I(kappa)B(alpha) kinase and nuclear factor-kappaB-regulated gene products. Fenretinide 0-29 nuclear factor kappa B subunit 1 Homo sapiens 161-182 16274795-1 2005 OBJECTIVE: To explore whether the mechanism of action of 4-hydroxyphenylretinamide (4-HPR, fenretidine), a synthetic retinoid, involves the functional activation of the nuclear hormone receptor class known as PPARs (peroxisome proliferator-activated receptors). Fenretinide 57-82 haptoglobin-related protein Homo sapiens 86-89 16230421-1 2005 N-(4-hydroxyphenyl) retinamide [4-HPR], a synthetic retinoid, has been shown to inhibit tumor cell growth, invasion, and metastasis by a mechanism that is not fully understood. Fenretinide 0-30 haptoglobin-related protein Homo sapiens 34-37 15958647-0 2005 The transforming growth factor-beta family members bone morphogenetic protein-2 and macrophage inhibitory cytokine-1 as mediators of the antiangiogenic activity of N-(4-hydroxyphenyl)retinamide. Fenretinide 164-193 bone morphogenetic protein 2 Homo sapiens 51-79 15907365-2 2005 In SH-SY5Y neuroblastoma cells, evidence suggests that acid sphingomyelinase activity is essential for the induction of ceramide and apoptosis in response to fenretinide. Fenretinide 158-169 sphingomyelin phosphodiesterase 1 Homo sapiens 55-76 15907365-3 2005 Downstream of ceramide, apoptosis in response to fenretinide is mediated by increased glucosylceramide synthase activity resulting in increased levels of gangliosides GD3 and GD2 via GD3 synthase. Fenretinide 49-60 UDP-glucose ceramide glucosyltransferase Homo sapiens 86-111 15907365-3 2005 Downstream of ceramide, apoptosis in response to fenretinide is mediated by increased glucosylceramide synthase activity resulting in increased levels of gangliosides GD3 and GD2 via GD3 synthase. Fenretinide 49-60 GRDX Homo sapiens 167-170 15907365-3 2005 Downstream of ceramide, apoptosis in response to fenretinide is mediated by increased glucosylceramide synthase activity resulting in increased levels of gangliosides GD3 and GD2 via GD3 synthase. Fenretinide 49-60 GRDX Homo sapiens 183-186 16270382-11 2005 The mechanism for 4-HPR-induced apoptosis was not clear, but we observed that 4-HPR could regulate p27(kip1), and overexpression of cerebroside sulfotransferase (CST) diminished the apoptosis induced by 4-HPR in melanoma cells. Fenretinide 18-23 cyclin-dependent kinase inhibitor 1B Mus musculus 103-107 16270382-11 2005 The mechanism for 4-HPR-induced apoptosis was not clear, but we observed that 4-HPR could regulate p27(kip1), and overexpression of cerebroside sulfotransferase (CST) diminished the apoptosis induced by 4-HPR in melanoma cells. Fenretinide 18-23 galactose-3-O-sulfotransferase 1 Mus musculus 132-160 16270382-11 2005 The mechanism for 4-HPR-induced apoptosis was not clear, but we observed that 4-HPR could regulate p27(kip1), and overexpression of cerebroside sulfotransferase (CST) diminished the apoptosis induced by 4-HPR in melanoma cells. Fenretinide 18-23 galactose-3-O-sulfotransferase 1 Mus musculus 162-165 16270382-11 2005 The mechanism for 4-HPR-induced apoptosis was not clear, but we observed that 4-HPR could regulate p27(kip1), and overexpression of cerebroside sulfotransferase (CST) diminished the apoptosis induced by 4-HPR in melanoma cells. Fenretinide 78-83 cyclin-dependent kinase inhibitor 1B Mus musculus 99-102 16270382-11 2005 The mechanism for 4-HPR-induced apoptosis was not clear, but we observed that 4-HPR could regulate p27(kip1), and overexpression of cerebroside sulfotransferase (CST) diminished the apoptosis induced by 4-HPR in melanoma cells. Fenretinide 78-83 cyclin-dependent kinase inhibitor 1B Mus musculus 103-107 15700313-0 2005 Randomized trial of fenretinide (4-HPR) to prevent recurrences, new localizations and carcinomas in patients operated on for oral leukoplakia: long-term results. Fenretinide 20-31 haptoglobin-related protein Homo sapiens 35-38 16077929-1 2005 N-(4-hydroxyphenyl)retinamide (4HPR) is a synthetic retinoid that has shown efficacy in cancer chemoprevention and therapy possibly due to its ability to induce growth inhibition and apoptosis in cancer cells. Fenretinide 0-29 haptoglobin-related protein Homo sapiens 32-35 16077929-5 2005 Suppression of endogenous and 4HPR-induced levels of p67phox using small interfering RNA did not result in a change in ROS generation or apoptosis. Fenretinide 30-34 neutrophil cytosolic factor 2 Homo sapiens 53-60 15894166-3 2005 We investigated the functional role of IB1/JIP-1 in LNCaP cells exposed to the proapoptotic agent N-(4-hydroxyphenyl)retinamide (4-HPR) which induced a reduction of IB1/JIP-1 content and a concomittant increase in JNK activity. Fenretinide 98-127 mitogen-activated protein kinase 8 interacting protein 1 Homo sapiens 39-42 15894166-3 2005 We investigated the functional role of IB1/JIP-1 in LNCaP cells exposed to the proapoptotic agent N-(4-hydroxyphenyl)retinamide (4-HPR) which induced a reduction of IB1/JIP-1 content and a concomittant increase in JNK activity. Fenretinide 98-127 mitogen-activated protein kinase 8 interacting protein 1 Homo sapiens 43-48 15894166-3 2005 We investigated the functional role of IB1/JIP-1 in LNCaP cells exposed to the proapoptotic agent N-(4-hydroxyphenyl)retinamide (4-HPR) which induced a reduction of IB1/JIP-1 content and a concomittant increase in JNK activity. Fenretinide 98-127 mitogen-activated protein kinase 8 interacting protein 1 Homo sapiens 165-168 15894166-3 2005 We investigated the functional role of IB1/JIP-1 in LNCaP cells exposed to the proapoptotic agent N-(4-hydroxyphenyl)retinamide (4-HPR) which induced a reduction of IB1/JIP-1 content and a concomittant increase in JNK activity. Fenretinide 98-127 mitogen-activated protein kinase 8 interacting protein 1 Homo sapiens 169-174 15894166-3 2005 We investigated the functional role of IB1/JIP-1 in LNCaP cells exposed to the proapoptotic agent N-(4-hydroxyphenyl)retinamide (4-HPR) which induced a reduction of IB1/JIP-1 content and a concomittant increase in JNK activity. Fenretinide 98-127 mitogen-activated protein kinase 8 Homo sapiens 214-217 15907982-2 2005 The synthetic retinoid, N-(4-hydroxyphenyl) retinamide (4-HPR), has been used in the chemoprevention of ovarian cancer. Fenretinide 24-54 haptoglobin-related protein Homo sapiens 58-61 16123139-5 2005 In this article, we investigated the ability of retinoids, such as 9-cis retinoic acid, all-trans retinoic acid, and N-4-hydroxyphenylretinamide (4-HPR), to sensitize the ductal epithelial cells of virgin mammary glands to DNA damage responses. Fenretinide 117-144 haptoglobin-related protein Homo sapiens 148-151 16034410-8 2005 Fenretinide, a synthetic retinoid that increases urinary excretion of RBP4, normalizes serum RBP4 levels and improves insulin resistance and glucose intolerance in mice with obesity induced by a high-fat diet. Fenretinide 0-11 retinol binding protein 4, plasma Mus musculus 70-74 16034410-8 2005 Fenretinide, a synthetic retinoid that increases urinary excretion of RBP4, normalizes serum RBP4 levels and improves insulin resistance and glucose intolerance in mice with obesity induced by a high-fat diet. Fenretinide 0-11 retinol binding protein 4, plasma Mus musculus 93-97 15958647-0 2005 The transforming growth factor-beta family members bone morphogenetic protein-2 and macrophage inhibitory cytokine-1 as mediators of the antiangiogenic activity of N-(4-hydroxyphenyl)retinamide. Fenretinide 164-193 growth differentiation factor 15 Homo sapiens 84-116 15958647-7 2005 The inhibition of invasion induced by 4HPR was also associated with decreased activities of the metalloproteases matrix metalloproteinase-2 and CD13/APN. Fenretinide 38-42 matrix metallopeptidase 2 Homo sapiens 113-139 15958647-7 2005 The inhibition of invasion induced by 4HPR was also associated with decreased activities of the metalloproteases matrix metalloproteinase-2 and CD13/APN. Fenretinide 38-42 alanyl aminopeptidase, membrane Homo sapiens 144-148 15958647-7 2005 The inhibition of invasion induced by 4HPR was also associated with decreased activities of the metalloproteases matrix metalloproteinase-2 and CD13/APN. Fenretinide 38-42 alanyl aminopeptidase, membrane Homo sapiens 149-152 15958647-10 2005 Blocking antibodies to bone morphogenetic protein-2 were able to reverse the suppressive effects of 4HPR in vitro and in vivo. Fenretinide 100-104 bone morphogenetic protein 2 Homo sapiens 23-51 15865936-0 2005 Fenretinide: a p53-independent way to kill cancer cells. Fenretinide 0-11 tumor protein p53 Homo sapiens 15-18 15917187-0 2005 ROS mediates 4HPR-induced posttranscriptional expression of the Gadd153 gene. Fenretinide 13-17 DNA damage inducible transcript 3 Homo sapiens 64-71 15917187-1 2005 All-trans-N-(4-hydroxyphenyl)retinamide (4HPR) is a synthetic retinoid that can induce apoptosis in many cancer cell lines. Fenretinide 0-39 haptoglobin-related protein Homo sapiens 42-45 15917187-4 2005 In 4HPR-treated cells, the elevation of Gadd153 protein level was prevented by vitamin C, which had no effect on the activation of the Gadd153 gene promoter. Fenretinide 3-7 DNA damage inducible transcript 3 Homo sapiens 40-47 15917187-8 2005 The inhibition of 4HPR-induced expression of Gadd153 protein by vitamin C was independent of intracellular proteasome activity and vitamin C had no effect on the intracellular decay of Gadd153 protein. Fenretinide 18-22 DNA damage inducible transcript 3 Homo sapiens 45-52 15865936-4 2005 Increased evidence suggests that the apoptotic pathway activated by fenretinide is p53-independent and this may represent a novel way to treat tumors resistant to DNA-damaging chemotherapeutic agents. Fenretinide 68-79 tumor protein p53 Homo sapiens 83-86 15756035-1 2005 PURPOSE: Fenretinide (4-HPR) is a synthetic retinoid that has shown a preventive activity in prostate cancer animal models. Fenretinide 9-20 haptoglobin-related protein Homo sapiens 24-27 16124896-1 2005 OBJECTIVE: To study the mechanism of the apoptosis induced by N-[4-hydroxyphenyl] retinamide (4-HPR) in bladder cancer cell line T24, and the involvement of DNA damage and repair. Fenretinide 62-92 haptoglobin-related protein Homo sapiens 96-99 16158929-1 2005 BACKGROUND: We have previously demonstrated that the synthetic retinoid N-(4-hydroxyphenyl) retinamide (4-HPR) induces the overproduction of reactive oxygen species (ROS) in human leukemia cells, which in turn triggers the intrinsic (mitochondrial) apoptotic pathway. Fenretinide 72-102 haptoglobin-related protein Homo sapiens 106-109 15695235-0 2005 N-(4-Hydroxyphenyl)retinamide is more potent than other phenylretinamides in inhibiting the growth of BRCA1-mutated breast cancer cells. Fenretinide 0-29 BRCA1 DNA repair associated Homo sapiens 102-107 15695235-2 2005 The synthetic retinoid N-(4-hydroxyphenyl)retinamide (4-HPR) has been shown to have a clinical chemopreventive activity in patients with premenopausal breast cancer. Fenretinide 23-52 haptoglobin-related protein Homo sapiens 56-59 15909111-1 2005 Fenretinide induces apoptosis in SH-SY5Y neuroblastoma cells via a signaling pathway involving the production of reactive oxygen species (ROS), 12-lipoxygenase activity and the induction of the GADD153 transcription factor. Fenretinide 0-11 DNA damage inducible transcript 3 Homo sapiens 194-201 15909111-4 2005 The aim of this study was to test the hypothesis that NF-kappa B activity mediates fenretinide-induced apoptosis in SH-SY5Y neuroblastoma cells. Fenretinide 83-94 nuclear factor kappa B subunit 1 Homo sapiens 54-64 15909111-6 2005 In parental SH-SY5Y cells, fenretinide induced NF-kappa B activity and Ikappa Balpha phosphorylation. Fenretinide 27-38 nuclear factor kappa B subunit 1 Homo sapiens 47-57 15909111-7 2005 These results suggest that NF-kappa B activity links fenretinide-induced ROS to the induction of apoptosis in SH-SH5Y cells, and may be a target for the future development of drugs for neuroblastoma therapy. Fenretinide 53-64 nuclear factor kappa B subunit 1 Homo sapiens 27-37 15837770-0 2005 p38MAPK-Dependent sensitivity of Ewing"s sarcoma family of tumors to fenretinide-induced cell death. Fenretinide 69-80 mitogen-activated protein kinase 14 Mus musculus 0-7 15837770-4 2005 The role of the stress-activated kinases p38(MAPK) and c-Jun NH(2)-terminal kinase in fenretinide-induced death was investigated by Western blot and inhibitor experiments. Fenretinide 86-97 mitogen-activated protein kinase 14 Mus musculus 41-44 15837770-9 2005 p38(MAPK) was activated within 15 minutes of fenretinide treatment and was dependent on ROS accumulation. Fenretinide 45-56 mitogen-activated protein kinase 14 Mus musculus 0-3 15837770-10 2005 Inhibition of p38(MAPK) activity partially rescued fenretinide-mediated cell death in ESFT but not in SH-SY5Y neuroblastoma cells. Fenretinide 51-62 mitogen-activated protein kinase 14 Homo sapiens 14-17 15837770-12 2005 After 8 hours, fenretinide induced mitochondrial depolarization (Deltapsi(m)) and release of cytochrome c into the cytoplasm in a ROS- and p38(MAPK)-dependent manner. Fenretinide 15-26 mitogen-activated protein kinase 14 Mus musculus 139-142 15837770-13 2005 CONCLUSIONS: These data show that the high sensitivity of ESFT cells to fenretinide is dependent in part on the rapid and sustained activation of p38(MAPK). Fenretinide 72-83 mitogen-activated protein kinase 14 Mus musculus 146-149 15735044-6 2005 Fenretinide treatment also increased levels of the death receptor DR5 and caused mitochondrial membrane depolarization. Fenretinide 0-11 TNF receptor superfamily member 10b Homo sapiens 66-69 15735044-7 2005 The levels of the retinoid receptors, retinoic acid receptor alpha and retinoid X receptor gamma, were up-regulated in response to fenretinide, suggestive of ligand-induced receptor up-regulation. Fenretinide 131-142 retinoic acid receptor alpha Homo sapiens 38-66 15735044-7 2005 The levels of the retinoid receptors, retinoic acid receptor alpha and retinoid X receptor gamma, were up-regulated in response to fenretinide, suggestive of ligand-induced receptor up-regulation. Fenretinide 131-142 retinoid X receptor gamma Homo sapiens 71-96 15735044-8 2005 IGF-I-induced proliferation in the meningioma cells was abolished by fenretinide. Fenretinide 69-80 insulin like growth factor 1 Homo sapiens 0-5 15735044-9 2005 We conclude that fenretinide induces apoptosis in all three histologic subtypes of meningioma and exerts diverse cellular effects, including DR5 up-regulation, modulation of retinoid receptor levels, and inhibition of IGF-I-induced proliferation. Fenretinide 17-28 TNF receptor superfamily member 10b Homo sapiens 141-144 15735044-9 2005 We conclude that fenretinide induces apoptosis in all three histologic subtypes of meningioma and exerts diverse cellular effects, including DR5 up-regulation, modulation of retinoid receptor levels, and inhibition of IGF-I-induced proliferation. Fenretinide 17-28 insulin like growth factor 1 Homo sapiens 218-223 15498786-0 2005 Chemopreventive N-(4-hydroxyphenyl)retinamide (fenretinide) targets deregulated NF-{kappa}B and Mat1A genes in the early stages of rat liver carcinogenesis. Fenretinide 16-45 methionine adenosyltransferase 1A Rattus norvegicus 96-101 15498786-0 2005 Chemopreventive N-(4-hydroxyphenyl)retinamide (fenretinide) targets deregulated NF-{kappa}B and Mat1A genes in the early stages of rat liver carcinogenesis. Fenretinide 47-58 methionine adenosyltransferase 1A Rattus norvegicus 96-101 15650234-2 2004 Fenretinide-induced cell death of neuroblastoma cells is caspase dependent and results in the release of cytochrome c from mitochondria independently of changes in permeability transition. Fenretinide 0-11 cytochrome c, somatic Homo sapiens 105-117 15862961-0 2005 Aromatase inhibition by 15-deoxy-prostaglandin J(2) (15-dPGJ(2)) and N-(4-hydroxyphenyl)-retinamide (4HPR) is associated with enhanced ceramide production. Fenretinide 69-99 haptoglobin-related protein Homo sapiens 102-105 15816545-0 2005 Action of fenretinide (4-HPR) on ovarian cancer and endothelial cells. Fenretinide 10-21 haptoglobin-related protein Homo sapiens 25-28 15816545-1 2005 BACKGROUND: Fenretinide (4-HPR) is a synthetic retinoid that has been reported to inhibit the growth of cancer cell lines in vitro. Fenretinide 12-23 haptoglobin-related protein Homo sapiens 27-30 15816545-8 2005 In OVCAR-5 cells treated with 1 mM fenretinide for 3 days, actin cytoskeleton stress fibers were disrupted and FAK tyrosine phosphorylation was elevated dose-dependently. Fenretinide 35-46 protein tyrosine kinase 2 Homo sapiens 111-114 15816545-10 2005 CONCLUSION: Fenretinide has anti-tumor activity by acting on the actin cytoskeleton and by regulating FAK tyrosine phosphorylation. Fenretinide 12-23 protein tyrosine kinase 2 Homo sapiens 102-105 15862961-2 2005 Recent studies on aromatase inhibition by the synthetic retinoid 4HPR, also known as fenretinide, and the PPARgamma agonist 15-dPGJ(2) have implicated a direct receptor-independent, redox-sensitive mechanism of action. Fenretinide 85-96 haptoglobin-related protein Homo sapiens 66-69 15650234-4 2004 Upstream events of fenretinide-induced signaling involve increased levels of ceramide as a result of increased sphingomyelinase activity, and the subsequent metabolism of ceramide to gangliosides via glucosylceramide synthase and GD3 synthase. Fenretinide 19-30 UDP-glucose ceramide glucosyltransferase Homo sapiens 200-225 15650242-5 2004 In addition, the apoptotic agent fenretinide (4HPR) and interferon-gamma (IFN-gamma) induce caspase-8 expression without modifying the methylation status of this gene. Fenretinide 33-44 caspase 8 Homo sapiens 92-101 15650242-5 2004 In addition, the apoptotic agent fenretinide (4HPR) and interferon-gamma (IFN-gamma) induce caspase-8 expression without modifying the methylation status of this gene. Fenretinide 46-50 caspase 8 Homo sapiens 92-101 15352033-5 2004 In the present study, fenretinide induced sustained-activation of both JNK and p38 MAPK in neuroblastoma cells. Fenretinide 22-33 mitogen-activated protein kinase 8 Homo sapiens 71-74 15352033-0 2004 Fenretinide induces sustained-activation of JNK/p38 MAPK and apoptosis in a reactive oxygen species-dependent manner in neuroblastoma cells. Fenretinide 0-11 mitogen-activated protein kinase 8 Homo sapiens 44-47 15352033-5 2004 In the present study, fenretinide induced sustained-activation of both JNK and p38 MAPK in neuroblastoma cells. Fenretinide 22-33 mitogen-activated protein kinase 14 Homo sapiens 79-82 15352033-0 2004 Fenretinide induces sustained-activation of JNK/p38 MAPK and apoptosis in a reactive oxygen species-dependent manner in neuroblastoma cells. Fenretinide 0-11 mitogen-activated protein kinase 14 Homo sapiens 48-51 15352033-6 2004 Pretreatment with the antioxidant L-ascorbic acid almost completely inhibited the accumulation of fenretinide-induced intracellular reactive oxygen species (ROS), activation of JNK and p38 MAPK and apoptosis. Fenretinide 98-109 mitogen-activated protein kinase 8 Homo sapiens 177-180 15352033-6 2004 Pretreatment with the antioxidant L-ascorbic acid almost completely inhibited the accumulation of fenretinide-induced intracellular reactive oxygen species (ROS), activation of JNK and p38 MAPK and apoptosis. Fenretinide 98-109 mitogen-activated protein kinase 14 Homo sapiens 185-188 15352033-8 2004 Our study demonstrates that in neuroblastoma cells, fenretinide induces sustained-activation of JNK and p38 MAPK in an ROS-dependent manner and indicates that JNK and p38 MAPK signaling might mediate fenretinide-induced apoptosis. Fenretinide 52-63 mitogen-activated protein kinase 8 Homo sapiens 96-99 15352033-8 2004 Our study demonstrates that in neuroblastoma cells, fenretinide induces sustained-activation of JNK and p38 MAPK in an ROS-dependent manner and indicates that JNK and p38 MAPK signaling might mediate fenretinide-induced apoptosis. Fenretinide 52-63 mitogen-activated protein kinase 14 Homo sapiens 104-107 15352033-8 2004 Our study demonstrates that in neuroblastoma cells, fenretinide induces sustained-activation of JNK and p38 MAPK in an ROS-dependent manner and indicates that JNK and p38 MAPK signaling might mediate fenretinide-induced apoptosis. Fenretinide 52-63 mitogen-activated protein kinase 8 Homo sapiens 159-162 15352033-8 2004 Our study demonstrates that in neuroblastoma cells, fenretinide induces sustained-activation of JNK and p38 MAPK in an ROS-dependent manner and indicates that JNK and p38 MAPK signaling might mediate fenretinide-induced apoptosis. Fenretinide 52-63 mitogen-activated protein kinase 14 Homo sapiens 167-170 15352033-8 2004 Our study demonstrates that in neuroblastoma cells, fenretinide induces sustained-activation of JNK and p38 MAPK in an ROS-dependent manner and indicates that JNK and p38 MAPK signaling might mediate fenretinide-induced apoptosis. Fenretinide 200-211 mitogen-activated protein kinase 8 Homo sapiens 159-162 15352033-8 2004 Our study demonstrates that in neuroblastoma cells, fenretinide induces sustained-activation of JNK and p38 MAPK in an ROS-dependent manner and indicates that JNK and p38 MAPK signaling might mediate fenretinide-induced apoptosis. Fenretinide 200-211 mitogen-activated protein kinase 14 Homo sapiens 167-170 15352033-9 2004 Our results also indicate that suppression of the fenretinide-induced ROS productive system and the downstream JNK and p38 MAPK signaling pathways causes neuroblastoma cells to become resistant to fenretinide. Fenretinide 50-61 mitogen-activated protein kinase 14 Homo sapiens 119-122 15375546-7 2004 All-trans retinoic acid (atRA), 9-cis retinoic acid (9cRA), and 4-(hydroxyphenyl) retinamide (4-HPR) induced RARbeta2 protein expression exclusively in the benign MCF10A cells and the former two retinoids, mRNA expression in MCF10A and MCF10AT cells, but not in malignant, MCF10CA1a cells, suggesting that the loss of inducible RARbeta expression is associated with the progression and malignant transformation of MCF10A cells. Fenretinide 64-92 haptoglobin-related protein Homo sapiens 96-99 15375546-7 2004 All-trans retinoic acid (atRA), 9-cis retinoic acid (9cRA), and 4-(hydroxyphenyl) retinamide (4-HPR) induced RARbeta2 protein expression exclusively in the benign MCF10A cells and the former two retinoids, mRNA expression in MCF10A and MCF10AT cells, but not in malignant, MCF10CA1a cells, suggesting that the loss of inducible RARbeta expression is associated with the progression and malignant transformation of MCF10A cells. Fenretinide 64-92 retinoic acid receptor beta Homo sapiens 109-116 15289350-2 2004 We hypothesize that targeting minimal residual disease with the cytotoxic retinoid N-(4-hydroxyphenyl) retinamide (4-HPR; fenretinide) may decrease relapse. Fenretinide 83-113 haptoglobin-related protein Homo sapiens 117-120 15448016-0 2004 Identification of the fenretinide metabolite 4-oxo-fenretinide present in human plasma and formed in human ovarian carcinoma cells through induction of cytochrome P450 26A1. Fenretinide 22-33 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 152-172 15448016-1 2004 PURPOSE: The synthetic retinoid fenretinide (4-HPR) exhibits preventive and therapeutic activity against ovarian tumors. Fenretinide 32-43 haptoglobin-related protein Homo sapiens 47-50 15339967-7 2004 RESULTS: In neuroblastoma cells, ROS generation and apoptosis were associated with fenretinide-induced increased levels of ceramide, glucosylceramide synthase activity, GD3 synthase activity, and GD3. Fenretinide 83-94 UDP-glucose ceramide glucosyltransferase Homo sapiens 133-158 15339967-7 2004 RESULTS: In neuroblastoma cells, ROS generation and apoptosis were associated with fenretinide-induced increased levels of ceramide, glucosylceramide synthase activity, GD3 synthase activity, and GD3. Fenretinide 83-94 GRDX Homo sapiens 169-172 15339967-7 2004 RESULTS: In neuroblastoma cells, ROS generation and apoptosis were associated with fenretinide-induced increased levels of ceramide, glucosylceramide synthase activity, GD3 synthase activity, and GD3. Fenretinide 83-94 GRDX Homo sapiens 196-199 15339967-8 2004 Fenretinide also induced increased levels of GD2, a ganglioside derived from GD3. Fenretinide 0-11 GRDX Homo sapiens 77-80 15339967-12 2004 CONCLUSIONS: A novel pathway of fenretinide-induced apoptosis is mediated by acidic sphingomyelinase, glucosylceramide synthase, and GD3 synthase, which may represent targets for future drug development. Fenretinide 32-43 UDP-glucose ceramide glucosyltransferase Homo sapiens 102-127 15339967-12 2004 CONCLUSIONS: A novel pathway of fenretinide-induced apoptosis is mediated by acidic sphingomyelinase, glucosylceramide synthase, and GD3 synthase, which may represent targets for future drug development. Fenretinide 32-43 GRDX Homo sapiens 133-136 15289350-2 2004 We hypothesize that targeting minimal residual disease with the cytotoxic retinoid N-(4-hydroxyphenyl) retinamide (4-HPR; fenretinide) may decrease relapse. Fenretinide 122-133 haptoglobin-related protein Homo sapiens 117-120 15240527-8 2004 Fenretinide decreased IGFBP-3 relative to placebo (P = 0.04). Fenretinide 0-11 insulin like growth factor binding protein 3 Homo sapiens 22-29 15297165-1 2004 OBJECTIVE: This trial examined the use of 4-hydroxyphenyl-retinamide (4-HPR), demonstrated to be a potent inhibitor of carcinogenesis in vitro and in animal models, in patients with cervical intraepithelial neoplasia (CIN) grades 2 to 3. Fenretinide 42-68 haptoglobin-related protein Homo sapiens 72-75 14765134-0 2004 N-(4-hydroxyphenyl) retinamide (4HPR) enhances TRAIL-mediated apoptosis through enhancement of a mitochondrial-dependent amplification loop in ovarian cancer cell lines. Fenretinide 0-30 haptoglobin-related protein Homo sapiens 33-36 15326375-0 2004 Inhibition of N-(4-hydroxyphenyl)retinamide-induced apoptosis in breast cancer cells by galectin-3. Fenretinide 14-43 galectin 3 Homo sapiens 88-98 15326375-6 2004 4HPR-induced apoptosis in these cells was associated with stimulation of reactive oxygen species generation, decreased levels of Bcl-2 protein, release of cytochrome c into the cytosol, increased caspase-3 activity, and poly(ADP-ribose) polymerase cleavage. Fenretinide 0-4 BCL2 apoptosis regulator Homo sapiens 129-134 15326375-6 2004 4HPR-induced apoptosis in these cells was associated with stimulation of reactive oxygen species generation, decreased levels of Bcl-2 protein, release of cytochrome c into the cytosol, increased caspase-3 activity, and poly(ADP-ribose) polymerase cleavage. Fenretinide 0-4 cytochrome c, somatic Homo sapiens 155-167 15326375-6 2004 4HPR-induced apoptosis in these cells was associated with stimulation of reactive oxygen species generation, decreased levels of Bcl-2 protein, release of cytochrome c into the cytosol, increased caspase-3 activity, and poly(ADP-ribose) polymerase cleavage. Fenretinide 0-4 caspase 3 Homo sapiens 196-205 14765134-0 2004 N-(4-hydroxyphenyl) retinamide (4HPR) enhances TRAIL-mediated apoptosis through enhancement of a mitochondrial-dependent amplification loop in ovarian cancer cell lines. Fenretinide 0-30 TNF superfamily member 10 Homo sapiens 47-52 15326375-8 2004 The demonstration that galectin-3 suppresses 4HPR-induced apoptosis in human breast carcinoma cells suggests that the increased expression of galectin-3 during cancer progression may be associated with 4HPR resistance. Fenretinide 45-49 galectin 3 Homo sapiens 23-33 15326375-8 2004 The demonstration that galectin-3 suppresses 4HPR-induced apoptosis in human breast carcinoma cells suggests that the increased expression of galectin-3 during cancer progression may be associated with 4HPR resistance. Fenretinide 45-49 galectin 3 Homo sapiens 142-152 15326375-8 2004 The demonstration that galectin-3 suppresses 4HPR-induced apoptosis in human breast carcinoma cells suggests that the increased expression of galectin-3 during cancer progression may be associated with 4HPR resistance. Fenretinide 202-206 galectin 3 Homo sapiens 23-33 15326375-8 2004 The demonstration that galectin-3 suppresses 4HPR-induced apoptosis in human breast carcinoma cells suggests that the increased expression of galectin-3 during cancer progression may be associated with 4HPR resistance. Fenretinide 202-206 galectin 3 Homo sapiens 142-152 15133197-1 2004 Retinoic acid and its amide derivative, N-(4-hydroxyphenyl)retinamide (4-HPR), have been proposed as chemopreventative and chemotherapeutic agents. Fenretinide 40-69 haptoglobin-related protein Homo sapiens 73-76 14765134-2 2004 Subtoxic concentrations of the semisynthetic retinoid N-(4-hydroxyphenyl)retinamide (4HPR) enhanced TRAIL-mediated apoptosis in ovarian cancer cell lines but not in immortalized nontumorigenic ovarian epithelial cells. Fenretinide 54-83 TNF superfamily member 10 Homo sapiens 100-105 14765134-2 2004 Subtoxic concentrations of the semisynthetic retinoid N-(4-hydroxyphenyl)retinamide (4HPR) enhanced TRAIL-mediated apoptosis in ovarian cancer cell lines but not in immortalized nontumorigenic ovarian epithelial cells. Fenretinide 85-89 TNF superfamily member 10 Homo sapiens 100-105 14647238-0 2004 Involvement of c-Fos in fenretinide-induced apoptosis in human ovarian carcinoma cells. Fenretinide 24-35 Fos proto-oncogene, AP-1 transcription factor subunit Homo sapiens 15-20 15291358-1 2004 N-(4-hydroxyphenyl) retinamide (Fenretinide, 4-HPR) inhibits cell growth by inducing apoptosis in numerous tumor cell types including all-trans-retinoic acid (ATRA)-resistant tumor cells. Fenretinide 0-30 haptoglobin-related protein Homo sapiens 47-50 15291358-1 2004 N-(4-hydroxyphenyl) retinamide (Fenretinide, 4-HPR) inhibits cell growth by inducing apoptosis in numerous tumor cell types including all-trans-retinoic acid (ATRA)-resistant tumor cells. Fenretinide 32-43 haptoglobin-related protein Homo sapiens 47-50 15013522-5 2004 Several anticancer agents, including the cytotoxic retinoid, fenretinide (4-HPR), have been shown to act, at least in part, by increasing tumor cell ceramide via de novo synthesis. Fenretinide 61-72 haptoglobin-related protein Homo sapiens 76-79 14647238-1 2004 Fenretinide (HPR), a synthetic retinoid that exhibits lower toxicity than other retinoids, has shown preventive and therapeutic activity against ovarian tumors. Fenretinide 0-11 haptoglobin-related protein Homo sapiens 13-16 14767528-4 2004 Like RA, 4-hydroxyphenyl retinamide (4-HPR) and 4-APRE, 4-BPRE was an active ligand for all three subtypes of RAR, but not for RXR, as determined by transcription assays in COS-1 cells. Fenretinide 9-35 haptoglobin-related protein Homo sapiens 39-42 14735468-1 2004 Fenretinide, 4-(N-hydroxyphenyl) retinamide (4-HPR), has demonstrated anticancer activity associated with a favorable toxicity profile and is now being investigated in several clinical trials. Fenretinide 0-11 haptoglobin-related protein Homo sapiens 47-50 14973114-2 2004 We show that HER2/neu uses Akt to induce cyclooxygenase-2 (COX-2) expression and that inhibition of Akt or COX-2 increases 4-HPR-induced apoptosis and nitric oxide production. Fenretinide 123-128 erb-b2 receptor tyrosine kinase 2 Homo sapiens 13-21 14973114-2 2004 We show that HER2/neu uses Akt to induce cyclooxygenase-2 (COX-2) expression and that inhibition of Akt or COX-2 increases 4-HPR-induced apoptosis and nitric oxide production. Fenretinide 123-128 AKT serine/threonine kinase 1 Homo sapiens 100-103 14973114-0 2004 Cyclooxygenase-2 is essential for HER2/neu to suppress N- (4-hydroxyphenyl)retinamide apoptotic effects in breast cancer cells. Fenretinide 55-85 prostaglandin-endoperoxide synthase 2 Homo sapiens 0-16 14973114-0 2004 Cyclooxygenase-2 is essential for HER2/neu to suppress N- (4-hydroxyphenyl)retinamide apoptotic effects in breast cancer cells. Fenretinide 55-85 erb-b2 receptor tyrosine kinase 2 Homo sapiens 34-38 14973114-2 2004 We show that HER2/neu uses Akt to induce cyclooxygenase-2 (COX-2) expression and that inhibition of Akt or COX-2 increases 4-HPR-induced apoptosis and nitric oxide production. Fenretinide 123-128 prostaglandin-endoperoxide synthase 2 Homo sapiens 107-112 14973114-0 2004 Cyclooxygenase-2 is essential for HER2/neu to suppress N- (4-hydroxyphenyl)retinamide apoptotic effects in breast cancer cells. Fenretinide 55-85 erb-b2 receptor tyrosine kinase 2 Homo sapiens 39-42 14645667-2 2003 The stress-inducible transcription factor known as growth and DNA damage (GADD)-inducible transcription factor 153 is induced in response to fenretinide and in other cell types modulates apoptosis via pro- and antiapoptotic members of the BCL2 family. Fenretinide 141-152 BCL2 apoptosis regulator Homo sapiens 239-243 14973114-1 2004 We reported that HER2/neu reduces the sensitivity of breast cancer cells to N-(4-hydroxyphenyl)retinamide (4-HPR) by suppressing nitric oxide production. Fenretinide 76-105 erb-b2 receptor tyrosine kinase 2 Homo sapiens 17-25 14973114-1 2004 We reported that HER2/neu reduces the sensitivity of breast cancer cells to N-(4-hydroxyphenyl)retinamide (4-HPR) by suppressing nitric oxide production. Fenretinide 107-112 erb-b2 receptor tyrosine kinase 2 Homo sapiens 17-25 14680805-3 2003 Treatment of the antisense BAG-1-transfected cells with the anti-cancer drugs staurosporine, paclitaxel, all-trans retinoic acid (ATRA), and N-(4-hydroxyphenyl) retinamide (4-HPR) resulted in significantly enhanced apoptosis and reduced cell viability relative to vector-transfected cells. Fenretinide 141-171 BAG cochaperone 1 Homo sapiens 27-32 14645674-0 2003 Heterogeneous role of caspase-8 in fenretinide-induced apoptosis in epithelial ovarian carcinoma cell lines. Fenretinide 35-46 caspase 8 Homo sapiens 22-31 14645674-4 2003 Transient transfection of cDNA-encoding cytokine response modifier A (CrmA), a caspase-8 inhibitor, diminished fenretinide-induced death in OV177 cells. Fenretinide 111-122 caspase 8 Homo sapiens 79-88 14645667-3 2003 Because BCL2-family proteins are important in apoptosis induced by chemotherapeutic drugs, GADD153 may be a key mediator of synergy between fenretinide and chemotherapeutic drugs. Fenretinide 140-151 BCL2 apoptosis regulator Homo sapiens 8-12 14645674-6 2003 Further analysis demonstrated that inhibition of Fas ligand, tumor necrosis factor-alpha, or TRAIL signaling with blocking reagents did not affect fenretinide-induced apoptosis, raising the possibility that fenretinide activates caspase-8 in a death receptor-independent manner. Fenretinide 207-218 caspase 8 Homo sapiens 229-238 14645667-3 2003 Because BCL2-family proteins are important in apoptosis induced by chemotherapeutic drugs, GADD153 may be a key mediator of synergy between fenretinide and chemotherapeutic drugs. Fenretinide 140-151 DNA damage inducible transcript 3 Homo sapiens 91-98 14645674-9 2003 Similarly, fenretinide treatment increased ceramide levels equally in cells that do (OV177) and do not (OV202) rely on caspase-8 to initiate apoptosis. Fenretinide 11-22 caspase 8 Homo sapiens 119-128 14645667-5 2003 Increased expression of GADD153 raised the background level of apoptosis and increased apoptosis induced by fenretinide or the chemotherapeutic drugs cisplatin and etoposide. Fenretinide 108-119 DNA damage inducible transcript 3 Homo sapiens 24-31 14645667-7 2003 Conversely, expression of antisense-GADD153 virtually abolished the induction of apoptosis in response to fenretinide but overall had no significant effect on apoptosis induced by chemotherapeutic drugs. Fenretinide 106-117 DNA damage inducible transcript 3 Homo sapiens 36-43 14645667-8 2003 The effect of antisense-GADD153 on synergy between chemotherapeutic drugs and fenretinide varied with the drug used: there was no effect on synergy between fenretinide and cisplatin, but the combination of fenretinide with etoposide became antagonistic. Fenretinide 78-89 DNA damage inducible transcript 3 Homo sapiens 24-31 14645667-9 2003 These results suggest that mechanisms mediating synergy between fenretinide and chemotherapeutic drugs lie upstream of GADD153. Fenretinide 64-75 DNA damage inducible transcript 3 Homo sapiens 119-126 14724932-7 2003 Although the mechanism of 4-HPR effects was not very clear, over expression of CST, which was inhibited by 4-HPR in our previous study, could diminish the apoptosis--inducing effect by 4-HPR. Fenretinide 26-31 cortistatin Mus musculus 79-82 14592467-1 2003 The retinamide, N-(4-hydroxyphenyl)retinamide (4-HPR), has shown promising anti-tumor activity, but it is unclear whether this compound is hydrolyzed to all-trans retinoic acid (atRA) and if so, whether this plays any role in its chemotherapeutic activity. Fenretinide 16-45 haptoglobin-related protein Homo sapiens 49-52 14555987-0 2003 HER2/neu reduces the apoptotic effects of N-(4-hydroxyphenyl)retinamide (4-HPR) in breast cancer cells by decreasing nitric oxide production. Fenretinide 42-71 erb-b2 receptor tyrosine kinase 2 Homo sapiens 0-8 14649406-1 2003 Effects of two natural (retinol and retinoic acid, RA) and one synthetic N-(4-hydroxyphenyl) retinamide (4-HPR) retinoids on proliferation and expression of urokinase-plasminogen activator (u-PA) by bovine mammary epithelial cells were examined. Fenretinide 73-103 plasminogen activator, urokinase Bos taurus 157-188 14649406-1 2003 Effects of two natural (retinol and retinoic acid, RA) and one synthetic N-(4-hydroxyphenyl) retinamide (4-HPR) retinoids on proliferation and expression of urokinase-plasminogen activator (u-PA) by bovine mammary epithelial cells were examined. Fenretinide 73-103 plasminogen activator, urokinase Bos taurus 190-194 14649406-1 2003 Effects of two natural (retinol and retinoic acid, RA) and one synthetic N-(4-hydroxyphenyl) retinamide (4-HPR) retinoids on proliferation and expression of urokinase-plasminogen activator (u-PA) by bovine mammary epithelial cells were examined. Fenretinide 105-110 plasminogen activator, urokinase Bos taurus 157-188 14581342-0 2003 Relationships between plasma insulin-like growth factor-I and insulin-like growth factor binding protein-3 and second breast cancer risk in a prevention trial of fenretinide. Fenretinide 162-173 insulin like growth factor 1 Homo sapiens 29-57 14581342-6 2003 Fenretinide induced reductions of IGF-I, IGFBP-3, and IGF-I:IGFBP-3 of 8% (95% CI, 2-12%; P = 0.004), 3% (95% CI, 1-5%; P = 0.002), and 5% (95% CI, 0-10%; P = 0.050), respectively. Fenretinide 0-11 insulin like growth factor 1 Homo sapiens 34-39 14581342-6 2003 Fenretinide induced reductions of IGF-I, IGFBP-3, and IGF-I:IGFBP-3 of 8% (95% CI, 2-12%; P = 0.004), 3% (95% CI, 1-5%; P = 0.002), and 5% (95% CI, 0-10%; P = 0.050), respectively. Fenretinide 0-11 insulin like growth factor binding protein 3 Homo sapiens 41-48 14581342-6 2003 Fenretinide induced reductions of IGF-I, IGFBP-3, and IGF-I:IGFBP-3 of 8% (95% CI, 2-12%; P = 0.004), 3% (95% CI, 1-5%; P = 0.002), and 5% (95% CI, 0-10%; P = 0.050), respectively. Fenretinide 0-11 insulin like growth factor 1 Homo sapiens 54-59 14581342-6 2003 Fenretinide induced reductions of IGF-I, IGFBP-3, and IGF-I:IGFBP-3 of 8% (95% CI, 2-12%; P = 0.004), 3% (95% CI, 1-5%; P = 0.002), and 5% (95% CI, 0-10%; P = 0.050), respectively. Fenretinide 0-11 insulin like growth factor binding protein 3 Homo sapiens 60-67 14581342-9 2003 CONCLUSIONS: Fenretinide induced a moderate reduction of IGF-I, which marginally explains observed cancer risk reductions in women </=50 years of age. Fenretinide 13-24 insulin like growth factor 1 Homo sapiens 57-62 14519946-1 2003 4-(N-Hydroxyphenyl)retinamide (also known as 4-HPR or fenretinide), a synthetic amide of all-trans retinoic acid (RA), has been implicated as a promising anticancer agent associated with reducing the toxicity related to RA. Fenretinide 54-65 haptoglobin-related protein Homo sapiens 47-50 14555987-0 2003 HER2/neu reduces the apoptotic effects of N-(4-hydroxyphenyl)retinamide (4-HPR) in breast cancer cells by decreasing nitric oxide production. Fenretinide 42-71 haptoglobin-related protein Homo sapiens 75-78 14555987-1 2003 The retinoid N-(4-hydroxyphenyl)retinamide (4-HPR also known as fenretinide) is a potent inducer of apoptosis in breast cancer cells. Fenretinide 13-42 haptoglobin-related protein Homo sapiens 46-49 14555987-1 2003 The retinoid N-(4-hydroxyphenyl)retinamide (4-HPR also known as fenretinide) is a potent inducer of apoptosis in breast cancer cells. Fenretinide 64-75 haptoglobin-related protein Homo sapiens 46-49 13679861-0 2003 The chemopreventive agent N-(4-hydroxyphenyl)retinamide induces apoptosis through a mitochondrial pathway regulated by proteins from the Bcl-2 family. Fenretinide 26-55 BCL2 apoptosis regulator Homo sapiens 137-142 13679861-1 2003 N-(4-hydroxyphenyl)retinamide (4-HPR, fenretinide) is a potent chemopreventive agent whose effect has been suggested to involve apoptosis induction. Fenretinide 38-49 haptoglobin-related protein Homo sapiens 33-36 14604280-0 2003 N-(4-Hydroxyphenyl)retinamide (4-HPR) induces leukemia cell death via generation of reactive oxygen species. Fenretinide 0-29 haptoglobin-related protein Homo sapiens 33-36 14604280-1 2003 The role of reactive oxygen species (ROS) in the cytotoxicity of N-(4-hydroxyphenyl)retinamide (4-HPR) was studied with use of the B-precursor lymphoblastic leukemia cell line YCUB-2. Fenretinide 65-94 haptoglobin-related protein Homo sapiens 98-101 13679861-1 2003 N-(4-hydroxyphenyl)retinamide (4-HPR, fenretinide) is a potent chemopreventive agent whose effect has been suggested to involve apoptosis induction. Fenretinide 0-29 haptoglobin-related protein Homo sapiens 33-36 12939601-3 2003 Previously, we observed that a synthetic retinoid N-(4-hydroxyphenyl)retinamide (4HPR) effectively impaired cell growth and survival (induction of growth arrest and apoptosis) in human hepatoma cells, which was accompanied by over expression of GADD153. Fenretinide 50-79 DNA damage inducible transcript 3 Homo sapiens 245-252 12939601-3 2003 Previously, we observed that a synthetic retinoid N-(4-hydroxyphenyl)retinamide (4HPR) effectively impaired cell growth and survival (induction of growth arrest and apoptosis) in human hepatoma cells, which was accompanied by over expression of GADD153. Fenretinide 81-85 DNA damage inducible transcript 3 Homo sapiens 245-252 12873973-2 2003 GCS inhibition, by both antisense and the specific inhibitor (D-threo)-1-phenyl-2-decanoylamino-3-morpholino-1-propanol (PDMP), results in a drastic decrease of apoptosis induced by the p53-independent chemotherapeutic agent N-(4-hydroxyphenyl)retinamide in neuroepithelioma cells. Fenretinide 225-254 UDP-glucose ceramide glucosyltransferase Homo sapiens 0-3 12880976-0 2003 Induction of GADD153 and Bak: novel molecular targets of fenretinide-induced apoptosis of neuroblastoma. Fenretinide 57-68 DNA damage inducible transcript 3 Homo sapiens 13-20 12880976-0 2003 Induction of GADD153 and Bak: novel molecular targets of fenretinide-induced apoptosis of neuroblastoma. Fenretinide 57-68 BCL2 antagonist/killer 1 Homo sapiens 25-28 12880976-2 2003 The stress-induced transcription factor GADD153 and the Bcl2-related protein Bak are upregulated in response to fenretinide. Fenretinide 112-123 DNA damage inducible transcript 3 Homo sapiens 40-47 12880976-2 2003 The stress-induced transcription factor GADD153 and the Bcl2-related protein Bak are upregulated in response to fenretinide. Fenretinide 112-123 BCL2 antagonist/killer 1 Homo sapiens 77-80 12880976-5 2003 Neither GADD153 or Bak were induced by chemotherapeutic agents but over expression of GADD153 results in increased sensitivity to fenretinide-induced apoptosis. Fenretinide 130-141 DNA damage inducible transcript 3 Homo sapiens 86-93 12880976-6 2003 Therefore, fenretinide induces apoptosis via RAR-dependent and -independent pathways in which the RAR-independent pathway is characterised by the reactive oxygen species-dependent induction of GADD153 and Bak. Fenretinide 11-22 retinoic acid receptor alpha Homo sapiens 45-48 12880976-6 2003 Therefore, fenretinide induces apoptosis via RAR-dependent and -independent pathways in which the RAR-independent pathway is characterised by the reactive oxygen species-dependent induction of GADD153 and Bak. Fenretinide 11-22 retinoic acid receptor alpha Homo sapiens 98-101 12880976-6 2003 Therefore, fenretinide induces apoptosis via RAR-dependent and -independent pathways in which the RAR-independent pathway is characterised by the reactive oxygen species-dependent induction of GADD153 and Bak. Fenretinide 11-22 DNA damage inducible transcript 3 Homo sapiens 193-200 12880976-6 2003 Therefore, fenretinide induces apoptosis via RAR-dependent and -independent pathways in which the RAR-independent pathway is characterised by the reactive oxygen species-dependent induction of GADD153 and Bak. Fenretinide 11-22 BCL2 antagonist/killer 1 Homo sapiens 205-208 12880980-1 2003 Retinoids are derivatives of vitamin A that include all trans-retinoic acid (ATRA), 13-cis-retinoic acid, (13-cis-RA), and fenretinide (4-HPR). Fenretinide 123-134 haptoglobin-related protein Homo sapiens 138-141 12843642-1 2003 Retinoic acid (RA), a potent inducer of cell differentiation, and N-(4-hydroxyphenyl)retinamide (4-HPR, fenretinide), a potent inducer of apoptosis, are well known as anticancer agents that are administered orally to patients for leukemia, breast and prostate cancer, respectively. Fenretinide 66-95 haptoglobin-related protein Homo sapiens 99-102 12843642-1 2003 Retinoic acid (RA), a potent inducer of cell differentiation, and N-(4-hydroxyphenyl)retinamide (4-HPR, fenretinide), a potent inducer of apoptosis, are well known as anticancer agents that are administered orally to patients for leukemia, breast and prostate cancer, respectively. Fenretinide 104-115 haptoglobin-related protein Homo sapiens 99-102 12815470-0 2003 Fenretinide induces cytochrome c release, caspase 9 activation and apoptosis in the absence of mitochondrial membrane depolarisation. Fenretinide 0-11 cytochrome c, somatic Homo sapiens 20-32 12815470-0 2003 Fenretinide induces cytochrome c release, caspase 9 activation and apoptosis in the absence of mitochondrial membrane depolarisation. Fenretinide 0-11 caspase 9 Homo sapiens 42-51 12736045-6 2003 RESULTS: LNCaP cells were five times more sensitive to N-(4-hydroxyphenyl)retinamide (4-HPR), a synthetic retinoid, compared with PC-3 cells. Fenretinide 55-84 haptoglobin-related protein Homo sapiens 88-91 12855611-1 2003 PURPOSE: The synthetic retinoid N-(4-hydroxyphenyl)retinamide [4-HPR (or fenretinide)] has preclinical and clinical preventive activity in breast carcinogenesis. Fenretinide 32-61 haptoglobin-related protein Homo sapiens 65-68 12855611-1 2003 PURPOSE: The synthetic retinoid N-(4-hydroxyphenyl)retinamide [4-HPR (or fenretinide)] has preclinical and clinical preventive activity in breast carcinogenesis. Fenretinide 73-84 haptoglobin-related protein Homo sapiens 65-68 12792755-0 2003 Induction of apoptosis by fenretinide in tumor cell lines correlates with DLX2, DLX3 and DLX4 gene expression. Fenretinide 26-37 distal-less homeobox 2 Homo sapiens 74-78 12792755-0 2003 Induction of apoptosis by fenretinide in tumor cell lines correlates with DLX2, DLX3 and DLX4 gene expression. Fenretinide 26-37 distal-less homeobox 3 Homo sapiens 80-84 12792755-0 2003 Induction of apoptosis by fenretinide in tumor cell lines correlates with DLX2, DLX3 and DLX4 gene expression. Fenretinide 26-37 distal-less homeobox 4 Homo sapiens 89-93 12796365-1 2003 PURPOSE: Fenretinide (4HPR), a synthetic retinoid, induces apoptosis in neuroblastoma cells. Fenretinide 9-20 haptoglobin-related protein Homo sapiens 23-26 12738746-0 2003 Cyclin d1 overexpression sensitizes breast cancer cells to fenretinide. Fenretinide 59-70 cyclin D1 Homo sapiens 0-9 12738746-1 2003 PURPOSE: Fenretinide has shown promise in the chemoprevention of breast cancer, a tumor type in which the oncogene cyclin D1 is overexpressed frequently. Fenretinide 9-20 cyclin D1 Homo sapiens 115-124 12738746-2 2003 We aimed at determining the effect of cyclin D1 level on the response to fenretinide treatment. Fenretinide 73-84 cyclin D1 Homo sapiens 38-47 12738746-4 2003 RESULTS: We show here that cells overexpressing cyclin D1 are significantly more sensitive to fenretinide than genetically matched cells that express low levels of cyclin D1, and that fenretinide prevents tumor formation arising from cyclin D1-overexpressing cells. Fenretinide 94-105 cyclin D1 Homo sapiens 48-57 12738746-4 2003 RESULTS: We show here that cells overexpressing cyclin D1 are significantly more sensitive to fenretinide than genetically matched cells that express low levels of cyclin D1, and that fenretinide prevents tumor formation arising from cyclin D1-overexpressing cells. Fenretinide 184-195 cyclin D1 Homo sapiens 48-57 12738746-4 2003 RESULTS: We show here that cells overexpressing cyclin D1 are significantly more sensitive to fenretinide than genetically matched cells that express low levels of cyclin D1, and that fenretinide prevents tumor formation arising from cyclin D1-overexpressing cells. Fenretinide 184-195 cyclin D1 Homo sapiens 164-173 12738746-4 2003 RESULTS: We show here that cells overexpressing cyclin D1 are significantly more sensitive to fenretinide than genetically matched cells that express low levels of cyclin D1, and that fenretinide prevents tumor formation arising from cyclin D1-overexpressing cells. Fenretinide 184-195 cyclin D1 Homo sapiens 164-173 12738746-5 2003 Furthermore, we show that fenretinide is also able to promote the regression of cyclin D1-positive tumors. Fenretinide 26-37 cyclin D1 Homo sapiens 80-89 12738746-6 2003 We also show that cells expressing a mutant of cyclin D1 that cannot bind to cdk4 are also more sensitive to fenretinide. Fenretinide 109-120 cyclin D1 Homo sapiens 47-56 12738746-7 2003 CONCLUSIONS: These results suggest that fenretinide may be particularly useful in the treatment of cyclin D1-positive breast cancers, and that the interaction between cyclin D1 and fenretinide is independent of cyclin D1 binding to cdk4. Fenretinide 40-51 cyclin D1 Homo sapiens 99-108 12619033-2 2003 We investigated the influence of androgen stimulation with testosterone on N-(4-hydroxyphenyl)retinamide (4-HPR)-induced apoptosis in the androgen-sensitive prostate cancer cell line LNCaP. Fenretinide 75-104 haptoglobin-related protein Homo sapiens 108-111 12619033-0 2003 Requirement of c-jun for testosterone-induced sensitization to N-(4-hydroxyphenyl)retinamide-induced apoptosis. Fenretinide 63-92 Jun proto-oncogene, AP-1 transcription factor subunit Homo sapiens 15-20 12537979-0 2003 N-(4-Hydroxylphenyl)retinamide (fenretinide, 4-HPR), a retinoid compound with antileukemic and proapoptotic activity in acute lymphoblastic leukemia (ALL). Fenretinide 32-43 haptoglobin-related protein Homo sapiens 47-50 12537979-2 2003 N-(4-Hydroxyphenyl)-all-trans-retinamide (fenretinide, 4-HPR) is a synthetic ATRA derivative with chemopreventive and cytotoxic activity against various cancer cell lines including myeloid leukemia. Fenretinide 0-40 haptoglobin-related protein Homo sapiens 57-60 12537979-2 2003 N-(4-Hydroxyphenyl)-all-trans-retinamide (fenretinide, 4-HPR) is a synthetic ATRA derivative with chemopreventive and cytotoxic activity against various cancer cell lines including myeloid leukemia. Fenretinide 42-53 haptoglobin-related protein Homo sapiens 57-60 12540501-1 2003 OBJECTIVES: To assess, in women participating in a breast cancer prevention trialon fenretinide (4-HPR), the relationship of drug and retinol levels with the risk of second breast malignancy, taking into account age and menopausal status. Fenretinide 84-95 haptoglobin-related protein Homo sapiens 99-102 12388538-2 2002 Herein we investigate the molecular mechanism of ceramide signaling in endothelial apoptosis induced by fenretinide (N-(4-hydroxyphenyl)retinamide (4-HPR)). Fenretinide 104-115 haptoglobin-related protein Homo sapiens 150-153 12517593-2 2003 A promising oral retinoid, 4-hydroxyphenylretinamide (4-HPR) induced apoptosis through non-retinoic acid-mediated pathways and is being studied in National Cancer Institute phase II trials in several organ sites. Fenretinide 27-52 haptoglobin-related protein Homo sapiens 56-59 12388538-2 2002 Herein we investigate the molecular mechanism of ceramide signaling in endothelial apoptosis induced by fenretinide (N-(4-hydroxyphenyl)retinamide (4-HPR)). Fenretinide 117-146 haptoglobin-related protein Homo sapiens 150-153 12234979-0 2002 GADD153 and 12-lipoxygenase mediate fenretinide-induced apoptosis of neuroblastoma. Fenretinide 36-47 DNA damage inducible transcript 3 Homo sapiens 0-7 12417027-1 2002 Fenretinide, N-(4-hydroxyphenyl)retinamide (4-HPR), is a synthetic amide of all-trans-retinoic acid (RA), which inhibits cell growth, induces apoptosis, and is an antioxidant, and cancer chemopreventive and antiproliferative agent. Fenretinide 0-11 haptoglobin-related protein Homo sapiens 46-49 12417027-1 2002 Fenretinide, N-(4-hydroxyphenyl)retinamide (4-HPR), is a synthetic amide of all-trans-retinoic acid (RA), which inhibits cell growth, induces apoptosis, and is an antioxidant, and cancer chemopreventive and antiproliferative agent. Fenretinide 13-42 haptoglobin-related protein Homo sapiens 46-49 12410564-0 2002 Contributions of mitogen-activated protein kinase and nuclear factor kappa B to N-(4-hydroxyphenyl)retinamide-induced apoptosis in prostate cancer cells. Fenretinide 80-109 nuclear factor kappa B subunit 1 Homo sapiens 54-76 12138118-0 2002 GADD153-mediated anticancer effects of N-(4-hydroxyphenyl)retinamide on human hepatoma cells. Fenretinide 39-68 DNA damage inducible transcript 3 Homo sapiens 0-7 12138118-9 2002 N-Acetyl-l-cysteine or GADD153 antisense significantly protected the cells from 4HPR-induced apoptosis, accompanying by the inhibition of GADD153 overexpression. Fenretinide 80-84 DNA damage inducible transcript 3 Homo sapiens 23-30 12138118-9 2002 N-Acetyl-l-cysteine or GADD153 antisense significantly protected the cells from 4HPR-induced apoptosis, accompanying by the inhibition of GADD153 overexpression. Fenretinide 80-84 DNA damage inducible transcript 3 Homo sapiens 138-145 12353227-0 2002 Upregulation of GADD153 expression in the apoptotic signaling of N-(4-hydroxyphenyl)retinamide (4HPR). Fenretinide 65-94 DNA damage inducible transcript 3 Homo sapiens 16-23 12353227-0 2002 Upregulation of GADD153 expression in the apoptotic signaling of N-(4-hydroxyphenyl)retinamide (4HPR). Fenretinide 65-94 haptoglobin-related protein Homo sapiens 97-100 12353227-9 2002 In the HeLa cells, both 4HPR and ATRA caused a 2- to 4-fold stimulation of the promoter activity of gadd153, but similar to the CNE3 cells, ATRA was incapable of upregulating the protein level of gadd153. Fenretinide 24-28 DNA damage inducible transcript 3 Homo sapiens 100-107 12481423-1 2002 N-(4-Hydroxyphenyl)retinamide (4-HPR) induces apoptosis in breast cancer cells; however, the molecular basis by which 4-HPR induces apoptosis is not well understood. Fenretinide 0-29 haptoglobin-related protein Homo sapiens 33-36 12481423-1 2002 N-(4-Hydroxyphenyl)retinamide (4-HPR) induces apoptosis in breast cancer cells; however, the molecular basis by which 4-HPR induces apoptosis is not well understood. Fenretinide 0-29 haptoglobin-related protein Homo sapiens 120-123 12234979-10 2002 These results suggest that fenretinide induces apoptosis via RAR-dependent and -independent pathways in which the RAR-independent pathway is characterized by a fenretinide-dependent increase in 12-LOX activity, leading to the induction of GADD153. Fenretinide 160-171 retinoic acid receptor alpha Homo sapiens 114-117 12234979-10 2002 These results suggest that fenretinide induces apoptosis via RAR-dependent and -independent pathways in which the RAR-independent pathway is characterized by a fenretinide-dependent increase in 12-LOX activity, leading to the induction of GADD153. Fenretinide 160-171 arachidonate 15-lipoxygenase Homo sapiens 194-200 12234979-10 2002 These results suggest that fenretinide induces apoptosis via RAR-dependent and -independent pathways in which the RAR-independent pathway is characterized by a fenretinide-dependent increase in 12-LOX activity, leading to the induction of GADD153. Fenretinide 160-171 DNA damage inducible transcript 3 Homo sapiens 239-246 12234979-4 2002 Expression of the stress-induced transcription factor, GADD153, was up-regulated at both the protein and mRNA levels in response to fenretinide. Fenretinide 132-143 DNA damage inducible transcript 3 Homo sapiens 55-62 12234979-5 2002 Overexpression of GADD153 increased apoptosis in the presence and absence of fenretinide, whereas reduced expression of GADD153 by expression of antisense DNA abrogated the response to fenretinide. Fenretinide 77-88 DNA damage inducible transcript 3 Homo sapiens 18-25 12234979-5 2002 Overexpression of GADD153 increased apoptosis in the presence and absence of fenretinide, whereas reduced expression of GADD153 by expression of antisense DNA abrogated the response to fenretinide. Fenretinide 185-196 DNA damage inducible transcript 3 Homo sapiens 120-127 12234979-6 2002 Although fenretinide is a partial retinoic acid receptor (RAR)-beta/gamma agonist, RARbeta/gamma antagonists did not block the induction of GADD153 by fenretinide; conversely, the induction of GADD153 was blocked by antioxidants. Fenretinide 9-20 retinoic acid receptor alpha Homo sapiens 34-56 12234979-6 2002 Although fenretinide is a partial retinoic acid receptor (RAR)-beta/gamma agonist, RARbeta/gamma antagonists did not block the induction of GADD153 by fenretinide; conversely, the induction of GADD153 was blocked by antioxidants. Fenretinide 9-20 retinoic acid receptor beta Homo sapiens 58-67 12234979-10 2002 These results suggest that fenretinide induces apoptosis via RAR-dependent and -independent pathways in which the RAR-independent pathway is characterized by a fenretinide-dependent increase in 12-LOX activity, leading to the induction of GADD153. Fenretinide 27-38 retinoic acid receptor alpha Homo sapiens 61-64 12234979-10 2002 These results suggest that fenretinide induces apoptosis via RAR-dependent and -independent pathways in which the RAR-independent pathway is characterized by a fenretinide-dependent increase in 12-LOX activity, leading to the induction of GADD153. Fenretinide 27-38 retinoic acid receptor alpha Homo sapiens 114-117 12234979-10 2002 These results suggest that fenretinide induces apoptosis via RAR-dependent and -independent pathways in which the RAR-independent pathway is characterized by a fenretinide-dependent increase in 12-LOX activity, leading to the induction of GADD153. Fenretinide 27-38 arachidonate 15-lipoxygenase Homo sapiens 194-200 12234979-10 2002 These results suggest that fenretinide induces apoptosis via RAR-dependent and -independent pathways in which the RAR-independent pathway is characterized by a fenretinide-dependent increase in 12-LOX activity, leading to the induction of GADD153. Fenretinide 27-38 DNA damage inducible transcript 3 Homo sapiens 239-246 11986953-1 2002 The retinoid, N-(4-hydroxyphenyl)retinamide (4-HPR), mediates p53-independent cytotoxicity and can increase reactive oxygen species and ceramide in solid tumor cell lines. Fenretinide 14-43 tumor protein p53 Homo sapiens 62-65 12595744-4 2002 At 1.3 micro M concentration (a clinically pharmacologically achievable dose), 4HPR increased ATRA sensitivity synergistically in HER2/NEU-overexpressing BT-474, MDA-MB-453, and MCF-7/Her2 breast cancer cells. Fenretinide 79-83 erb-b2 receptor tyrosine kinase 2 Homo sapiens 184-188 12124800-1 2002 The synthetic retinoid fenretinide [N-(4-hydroxyphenyl)retinamide, 4-HPR] has demonstrated growth inhibition and induction of apoptosis of various malignant cells, including lung cancer cell lines. Fenretinide 23-34 haptoglobin-related protein Homo sapiens 69-72 12124800-1 2002 The synthetic retinoid fenretinide [N-(4-hydroxyphenyl)retinamide, 4-HPR] has demonstrated growth inhibition and induction of apoptosis of various malignant cells, including lung cancer cell lines. Fenretinide 36-65 haptoglobin-related protein Homo sapiens 69-72 12079295-11 2002 Furthermore, a possible protective effect of fenretinide in BRCA-mutated women was suggested. Fenretinide 45-56 BRCA1 DNA repair associated Homo sapiens 60-64 12072549-0 2002 Re: Effects of N-(4-hydroxy-phenyl)retinamide on hTERT expression in the bronchial epithelium of cigarette smokers. Fenretinide 15-45 telomerase reverse transcriptase Homo sapiens 49-54 11979432-4 2002 Therefore, we analyzed the expression of OVCA1 and OVCA2 in cells in response to treatment with all-trans retinoic acid (RA) and N-(4-hydroxyphenyl)retinamide (4HPR), or under conditions of low serum and confluence, to determine further the roles of OVCA1 and OVCA2 in cell growth, apoptosis and differentiation. Fenretinide 129-158 diphthamide biosynthesis 1 Homo sapiens 41-46 11979432-4 2002 Therefore, we analyzed the expression of OVCA1 and OVCA2 in cells in response to treatment with all-trans retinoic acid (RA) and N-(4-hydroxyphenyl)retinamide (4HPR), or under conditions of low serum and confluence, to determine further the roles of OVCA1 and OVCA2 in cell growth, apoptosis and differentiation. Fenretinide 129-158 OVCA2 serine hydrolase domain containing Homo sapiens 51-56 11979432-6 2002 In addition, we observed that OVCA2 protein is proteolytically degraded in response to RA and 4HPR treatment in a time- and dose-dependent manner in the promyelocytic leukemia cell line HL60. Fenretinide 94-98 OVCA2 serine hydrolase domain containing Homo sapiens 30-35 12161154-1 2002 Retinoic acid analogues such as N-(4-hydroxyphenyl)retinamide (4-HPR) are effective chemopreventatives and chemotherapeutics for numerous types of cancer. Fenretinide 32-61 haptoglobin-related protein Homo sapiens 65-68 12595744-0 2002 N-(4-hydroxyphenyl)-retinamide selectively increases All-TRANS retinoic acid inhibitory effects in HER2/NEU-overexpressing breast cancer cells. Fenretinide 0-30 erb-b2 receptor tyrosine kinase 2 Homo sapiens 99-103 12595744-0 2002 N-(4-hydroxyphenyl)-retinamide selectively increases All-TRANS retinoic acid inhibitory effects in HER2/NEU-overexpressing breast cancer cells. Fenretinide 0-30 erb-b2 receptor tyrosine kinase 2 Homo sapiens 104-107 12595744-2 2002 N-(4-hydroxyphenyl)-retinamide (4HPR), a synthetic analogue of ATRA, has been shown to repress the expression of HER2/neu and its family member, epidermal growth factor receptor (EGFR). Fenretinide 0-30 erb-b2 receptor tyrosine kinase 2 Homo sapiens 113-117 12595744-2 2002 N-(4-hydroxyphenyl)-retinamide (4HPR), a synthetic analogue of ATRA, has been shown to repress the expression of HER2/neu and its family member, epidermal growth factor receptor (EGFR). Fenretinide 0-30 erb-b2 receptor tyrosine kinase 2 Homo sapiens 118-121 12595744-2 2002 N-(4-hydroxyphenyl)-retinamide (4HPR), a synthetic analogue of ATRA, has been shown to repress the expression of HER2/neu and its family member, epidermal growth factor receptor (EGFR). Fenretinide 0-30 epidermal growth factor receptor Homo sapiens 145-177 12595744-2 2002 N-(4-hydroxyphenyl)-retinamide (4HPR), a synthetic analogue of ATRA, has been shown to repress the expression of HER2/neu and its family member, epidermal growth factor receptor (EGFR). Fenretinide 0-30 epidermal growth factor receptor Homo sapiens 179-183 12595744-2 2002 N-(4-hydroxyphenyl)-retinamide (4HPR), a synthetic analogue of ATRA, has been shown to repress the expression of HER2/neu and its family member, epidermal growth factor receptor (EGFR). Fenretinide 32-36 erb-b2 receptor tyrosine kinase 2 Homo sapiens 113-117 12595744-2 2002 N-(4-hydroxyphenyl)-retinamide (4HPR), a synthetic analogue of ATRA, has been shown to repress the expression of HER2/neu and its family member, epidermal growth factor receptor (EGFR). Fenretinide 32-36 erb-b2 receptor tyrosine kinase 2 Homo sapiens 118-121 12595744-2 2002 N-(4-hydroxyphenyl)-retinamide (4HPR), a synthetic analogue of ATRA, has been shown to repress the expression of HER2/neu and its family member, epidermal growth factor receptor (EGFR). Fenretinide 32-36 epidermal growth factor receptor Homo sapiens 145-177 12595744-2 2002 N-(4-hydroxyphenyl)-retinamide (4HPR), a synthetic analogue of ATRA, has been shown to repress the expression of HER2/neu and its family member, epidermal growth factor receptor (EGFR). Fenretinide 32-36 epidermal growth factor receptor Homo sapiens 179-183 12595744-4 2002 At 1.3 micro M concentration (a clinically pharmacologically achievable dose), 4HPR increased ATRA sensitivity synergistically in HER2/NEU-overexpressing BT-474, MDA-MB-453, and MCF-7/Her2 breast cancer cells. Fenretinide 79-83 erb-b2 receptor tyrosine kinase 2 Homo sapiens 130-134 12175543-0 2002 N-(4-hydroxyphenyl)retinamide (4-HPR) modulates GADD45 expression in radiosensitive bladder cancer cell lines. Fenretinide 0-29 haptoglobin-related protein Homo sapiens 33-36 12175543-0 2002 N-(4-hydroxyphenyl)retinamide (4-HPR) modulates GADD45 expression in radiosensitive bladder cancer cell lines. Fenretinide 0-29 growth arrest and DNA damage inducible alpha Homo sapiens 48-54 12175543-1 2002 We previously demonstrated that N-(4-hydroxyphenyl)retinamide (4-HPR) and gamma-irradiation, when used in combination, had a synergistic effect in inducing apoptosis in bladder cancer cells, suggesting that 4-HPR may increase radiosensitivity in bladder cancer cells. Fenretinide 32-61 haptoglobin-related protein Homo sapiens 65-68 12175543-1 2002 We previously demonstrated that N-(4-hydroxyphenyl)retinamide (4-HPR) and gamma-irradiation, when used in combination, had a synergistic effect in inducing apoptosis in bladder cancer cells, suggesting that 4-HPR may increase radiosensitivity in bladder cancer cells. Fenretinide 32-61 haptoglobin-related protein Homo sapiens 209-212 11986953-1 2002 The retinoid, N-(4-hydroxyphenyl)retinamide (4-HPR), mediates p53-independent cytotoxicity and can increase reactive oxygen species and ceramide in solid tumor cell lines. Fenretinide 14-43 haptoglobin-related protein Homo sapiens 47-50 11705848-2 2001 We tested N-(4-hydroxyphenyl)retinamide (4-HPR), a promising oral retinoid that has been shown to induce apoptosis through nonretinoic receptor acid-mediated pathways, for its toxicity and efficacy against CIN 2/3. Fenretinide 10-39 haptoglobin-related protein Homo sapiens 43-46 11698412-7 2002 Intriguingly, bongkrekic acid, a specific ANT inhibitor, enhanced 4HPR-induced deltapsi(m) disruption, which in turn decreased the level of CO III transcripts, which was accompanied by increases in the generation of reactive oxygen species and in apoptosis. Fenretinide 66-70 solute carrier family 25 member 6 Homo sapiens 42-45 11698412-7 2002 Intriguingly, bongkrekic acid, a specific ANT inhibitor, enhanced 4HPR-induced deltapsi(m) disruption, which in turn decreased the level of CO III transcripts, which was accompanied by increases in the generation of reactive oxygen species and in apoptosis. Fenretinide 66-70 mitochondrially encoded cytochrome c oxidase III Homo sapiens 140-146 11698412-8 2002 In contrast, atractyloside, an activator of ANT, inhibited those 4HPR-induced effects. Fenretinide 65-69 solute carrier family 25 member 6 Homo sapiens 44-47 11921197-6 2002 The Italian trial of fenretinide (4-HPR) in women with stage I breast cancer randomized women to fenretinide or no intervention. Fenretinide 21-32 haptoglobin-related protein Homo sapiens 36-39 11921197-9 2002 It has been suggested that this effect may be related to insulin-like growth factor 1 (IGF-1), which has been shown to be modulated by fenretinide in premenopausal but not postmenopausal women. Fenretinide 135-146 insulin like growth factor 1 Homo sapiens 57-85 11921197-9 2002 It has been suggested that this effect may be related to insulin-like growth factor 1 (IGF-1), which has been shown to be modulated by fenretinide in premenopausal but not postmenopausal women. Fenretinide 135-146 insulin like growth factor 1 Homo sapiens 87-92 11732001-0 2001 Activation of caspase-8 during N-(4-hydroxyphenyl)retinamide-induced apoptosis in Fas-defective hepatoma cells. Fenretinide 31-60 caspase 8 Homo sapiens 14-23 12235869-0 2002 Experimental studies on the effects of the combined use of N-(4-hydroxyphenyl)retinamide (4-HPR) and tamoxifen (TAM) for estrogen receptor (ER)-negative breast cancer. Fenretinide 59-88 estrogen receptor 1 Homo sapiens 121-138 12235869-0 2002 Experimental studies on the effects of the combined use of N-(4-hydroxyphenyl)retinamide (4-HPR) and tamoxifen (TAM) for estrogen receptor (ER)-negative breast cancer. Fenretinide 59-88 estrogen receptor 1 Homo sapiens 140-142 12235869-1 2002 We investigated the effects of combination therapy with N-(4-hydroxyphenyl)retinamide (4-HPR) and tamoxifen (TAM) on estrogen receptor (ER) negative breast cancer, for which no effective supplementary therapy has been established, using the human breast cancer cell line MDA-MB-231. Fenretinide 56-85 estrogen receptor 1 Homo sapiens 117-134 12235869-1 2002 We investigated the effects of combination therapy with N-(4-hydroxyphenyl)retinamide (4-HPR) and tamoxifen (TAM) on estrogen receptor (ER) negative breast cancer, for which no effective supplementary therapy has been established, using the human breast cancer cell line MDA-MB-231. Fenretinide 56-85 estrogen receptor 1 Homo sapiens 136-138 11807954-1 2002 To explore the mechanisms underlying the pro-apoptotic effects of the synthetic retinoid N-4-(hydroxyphenyl)retinamide (4-HPR) on LNCaP human prostate cancer cells, we used the differential display-polymerase chain reaction (DD-PCR) technique to identify 4-HPR-responsive genes. Fenretinide 89-118 haptoglobin-related protein Homo sapiens 122-125 11807954-1 2002 To explore the mechanisms underlying the pro-apoptotic effects of the synthetic retinoid N-4-(hydroxyphenyl)retinamide (4-HPR) on LNCaP human prostate cancer cells, we used the differential display-polymerase chain reaction (DD-PCR) technique to identify 4-HPR-responsive genes. Fenretinide 89-118 haptoglobin-related protein Homo sapiens 257-260 11504771-0 2001 Effects of N-(4-hydroxyphenyl)retinamide on hTERT expression in the bronchial epithelium of cigarette smokers. Fenretinide 11-40 telomerase reverse transcriptase Homo sapiens 44-49 11483862-0 2001 Mechanism of fenretinide (4-HPR)-induced cell death. Fenretinide 13-24 haptoglobin-related protein Homo sapiens 28-31 11483862-1 2001 4-HPR (fenretinide) is a synthetic analog of retinoic acid (RA) whose potential as a chemopreventative agent has gained support from in vitro and animal experiments and in limited clinical trials. Fenretinide 7-18 haptoglobin-related protein Homo sapiens 2-5 11483862-5 2001 Apoptosis in response to fenretinide primarily occurs by a receptor-independent mechanism, which is accompanied by increases in signaling molecules, e.g., ceramide, and cysteine-dependent aspartate-directed proteases, termed caspases, including execution caspase-3. Fenretinide 25-36 caspase 3 Homo sapiens 255-264 11483862-6 2001 Both caspase-3 inhibitor DEVD-CHO and ceramide synthase inhibitor fumonisin B(1) (FB(1)) block fenretinide-induced apoptosis. Fenretinide 95-106 caspase 3 Homo sapiens 5-14 11483862-7 2001 Increase in caspase-3 appears to result from fenretinide-elicited stabilization of procaspase-3 zymogen. Fenretinide 45-56 caspase 3 Homo sapiens 12-21 11483862-7 2001 Increase in caspase-3 appears to result from fenretinide-elicited stabilization of procaspase-3 zymogen. Fenretinide 45-56 caspase 3 Homo sapiens 83-95 11551492-0 2001 N-(4-hydroxyphenyl)retinamide (4-HPR) decreases neoplastic properties of human prostate cells: an agent for prevention. Fenretinide 0-29 haptoglobin-related protein Homo sapiens 33-36 11551492-3 2001 This study focuses on the ability of the synthetic retinoid, N-(4-hydroxyphenyl)-retinamide (4-HPR), to reverse changes associated with malignant transformation and tumor progression, towards a normal phenotype. Fenretinide 61-91 haptoglobin-related protein Homo sapiens 95-98 11745408-2 2001 The synthetic retinoid fenretinide (HPR) inhibits carcinogenesis in various animal models. Fenretinide 23-34 haptoglobin-related protein Homo sapiens 36-39 11588130-2 2001 Because the synthetic retinoid fenretinide showed a beneficial effect on second breast cancers in premenopausal women in a Phase III trial, we studied its long-term effects on IGF-I levels. Fenretinide 31-42 insulin like growth factor 1 Homo sapiens 176-181 11588130-11 2001 Fenretinide induces a moderate decline of IGF-I levels in women < or = 50 years of age. Fenretinide 0-11 insulin like growth factor 1 Homo sapiens 42-47 11504771-2 2001 To determine the role of telomerase in early lung carcinogenesis and as a potential biomarker in chemoprevention trials, we analyzed the expression of the human telomerase reverse transcriptase catalytic subunit (hTERT) in bronchial biopsy specimens from cigarette smokers who were enrolled in a randomized, double-blinded, placebo-controlled chemoprevention trial of N-(4-hydroxyphenyl)retinamide (4-HPR). Fenretinide 368-397 telomerase reverse transcriptase Homo sapiens 213-218 11489831-0 2001 Synergistic effects of the fenretinide (4-HPR) and anti-CD20 monoclonal antibodies on apoptosis induction of malignant human B cells. Fenretinide 27-38 haptoglobin-related protein Homo sapiens 42-45 11489831-4 2001 In this report, we evaluate the in vitro effects of N-(4-hydroxyphenyl) retinamide (4-HPR) with and without anti-CD20 antibodies in B-cell lymphoma lines. Fenretinide 52-82 haptoglobin-related protein Homo sapiens 86-89 11384104-1 2001 We investigated whether the efficacy of fenretinide (HPR) against ovarian tumours may be limited by induction of resistance. Fenretinide 40-51 haptoglobin-related protein Homo sapiens 53-56 11431347-1 2001 The retinoid N-(4-hydroxyphenyl)retinamide (4-HPR; fenretinide) is cytotoxic to a variety of cancer cell lines, and we previously showed an association between ceramide generation and 4-HPR cytotoxicity for neuroblastoma cell lines (B. J. Maurer et al., J. Natl. Fenretinide 13-42 haptoglobin-related protein Homo sapiens 46-49 11431347-1 2001 The retinoid N-(4-hydroxyphenyl)retinamide (4-HPR; fenretinide) is cytotoxic to a variety of cancer cell lines, and we previously showed an association between ceramide generation and 4-HPR cytotoxicity for neuroblastoma cell lines (B. J. Maurer et al., J. Natl. Fenretinide 13-42 haptoglobin-related protein Homo sapiens 186-189 11431347-1 2001 The retinoid N-(4-hydroxyphenyl)retinamide (4-HPR; fenretinide) is cytotoxic to a variety of cancer cell lines, and we previously showed an association between ceramide generation and 4-HPR cytotoxicity for neuroblastoma cell lines (B. J. Maurer et al., J. Natl. Fenretinide 51-62 haptoglobin-related protein Homo sapiens 46-49 11431347-1 2001 The retinoid N-(4-hydroxyphenyl)retinamide (4-HPR; fenretinide) is cytotoxic to a variety of cancer cell lines, and we previously showed an association between ceramide generation and 4-HPR cytotoxicity for neuroblastoma cell lines (B. J. Maurer et al., J. Natl. Fenretinide 51-62 haptoglobin-related protein Homo sapiens 186-189 11412986-2 2001 The synthesis of a nonhydrolyzable, carbon-linked analogue (4-HBR) of the retinoid N-(4-hydroxyphenyl)retinamide (4-HPR) using Umpolung methods is described. Fenretinide 83-112 haptoglobin-related protein Homo sapiens 116-119 11497248-2 2001 In the present study, we determined the influence of high dietary concentrations of 4-hydroxyphenylretinamide (4-HPR) and 13-cis-retinoic acid (13-cis-RA) on RAR-beta mRNA expression in female mice. Fenretinide 84-109 retinoic acid receptor, beta Mus musculus 158-166 11497248-2 2001 In the present study, we determined the influence of high dietary concentrations of 4-hydroxyphenylretinamide (4-HPR) and 13-cis-retinoic acid (13-cis-RA) on RAR-beta mRNA expression in female mice. Fenretinide 111-116 retinoic acid receptor, beta Mus musculus 158-166 11295632-2 2001 A phase 2 chemoprevention trial of 4-hydroxyphenyl retinamide (4-HPR) versus placebo was conducted in men with a histologic diagnosis of early prostate cancer and scheduled to have radical prostatectomy. Fenretinide 35-61 haptoglobin-related protein Homo sapiens 65-68 11287445-3 2001 Some novel retinoids, such as fenretinide, i.e., N-(4-hydroxyphenyl)retinamide (4-HPR), induce apoptosis through retinoic acid receptor-independent mechanisms; however, they appear to do so only at concentrations above those achieved in clinical chemoprevention trials. Fenretinide 30-41 haptoglobin-related protein Homo sapiens 82-85 11287445-3 2001 Some novel retinoids, such as fenretinide, i.e., N-(4-hydroxyphenyl)retinamide (4-HPR), induce apoptosis through retinoic acid receptor-independent mechanisms; however, they appear to do so only at concentrations above those achieved in clinical chemoprevention trials. Fenretinide 49-78 haptoglobin-related protein Homo sapiens 82-85 11368409-3 2001 Suppression of pRB protein expression in HMECs by retroviral-mediated expression of the E7 protein of the human papillomavirus strain 16 (HPV-16) was associated with resistance to ATRA-mediated growth arrest but not to the synthetic retinoid N-(4-hydroxyphenyl) retinamide (4-HPR or fenretinide). Fenretinide 242-272 RB transcriptional corepressor 1 Homo sapiens 15-18 11166911-0 2001 The retinoid fenretinide inhibits proliferation and downregulates cyclooxygenase-2 gene expression in human colon adenocarcinoma cell lines. Fenretinide 13-24 prostaglandin-endoperoxide synthase 2 Homo sapiens 66-82 11166911-1 2001 Fenretinide [N-(4-Hydroxyphenyl)retinamide, 4-HPR] (10(-10)-10(-6) M) treatment of HT-29 human colon cancer cells for 24-72 h significantly inhibited their growth. Fenretinide 0-11 haptoglobin-related protein Homo sapiens 46-49 11368409-3 2001 Suppression of pRB protein expression in HMECs by retroviral-mediated expression of the E7 protein of the human papillomavirus strain 16 (HPV-16) was associated with resistance to ATRA-mediated growth arrest but not to the synthetic retinoid N-(4-hydroxyphenyl) retinamide (4-HPR or fenretinide). Fenretinide 283-294 RB transcriptional corepressor 1 Homo sapiens 15-18 11434448-9 2001 Ongoing studies on the validation of the circulating IGF-1 as a surrogate endpoint biomarker of fenretinide activity and on the effectiveness of the combination with low dose tamoxifen may provide further insight into the future clinical application of fenretinide. Fenretinide 96-107 insulin like growth factor 1 Homo sapiens 53-58 11176527-10 2001 All-trans-retinoic acid and 4HPR induced retinoic acid receptor beta expression in 1 bladder cancer cell line. Fenretinide 28-32 retinoic acid receptor beta Homo sapiens 41-68 11434448-9 2001 Ongoing studies on the validation of the circulating IGF-1 as a surrogate endpoint biomarker of fenretinide activity and on the effectiveness of the combination with low dose tamoxifen may provide further insight into the future clinical application of fenretinide. Fenretinide 253-264 insulin like growth factor 1 Homo sapiens 53-58 11107142-1 2000 The RARbeta/gamma-selective retinoids fenretinide and CD437 induce caspase-dependent apoptosis but generate free radicals independently of caspases. Fenretinide 38-49 retinoic acid receptor beta Homo sapiens 4-11 11106681-1 2000 BACKGROUND: We previously reported that N-(4-hydroxyphenyl)retinamide (4-HPR, fenretinide) treatment caused large increases of ceramide levels in neuroblastoma cell lines and induced cell death by a combination of apoptosis and necrosis through p53 (also known as TP53)-independent and caspase-independent pathways. Fenretinide 40-69 haptoglobin-related protein Homo sapiens 73-76 11106681-1 2000 BACKGROUND: We previously reported that N-(4-hydroxyphenyl)retinamide (4-HPR, fenretinide) treatment caused large increases of ceramide levels in neuroblastoma cell lines and induced cell death by a combination of apoptosis and necrosis through p53 (also known as TP53)-independent and caspase-independent pathways. Fenretinide 40-69 tumor protein p53 Homo sapiens 245-248 11106681-1 2000 BACKGROUND: We previously reported that N-(4-hydroxyphenyl)retinamide (4-HPR, fenretinide) treatment caused large increases of ceramide levels in neuroblastoma cell lines and induced cell death by a combination of apoptosis and necrosis through p53 (also known as TP53)-independent and caspase-independent pathways. Fenretinide 40-69 tumor protein p53 Homo sapiens 264-268 11106681-1 2000 BACKGROUND: We previously reported that N-(4-hydroxyphenyl)retinamide (4-HPR, fenretinide) treatment caused large increases of ceramide levels in neuroblastoma cell lines and induced cell death by a combination of apoptosis and necrosis through p53 (also known as TP53)-independent and caspase-independent pathways. Fenretinide 78-89 tumor protein p53 Homo sapiens 245-248 11106681-1 2000 BACKGROUND: We previously reported that N-(4-hydroxyphenyl)retinamide (4-HPR, fenretinide) treatment caused large increases of ceramide levels in neuroblastoma cell lines and induced cell death by a combination of apoptosis and necrosis through p53 (also known as TP53)-independent and caspase-independent pathways. Fenretinide 78-89 tumor protein p53 Homo sapiens 264-268 11107126-7 2000 Both parental and RA-resistant lines showed 2-4 logs of cell kill in response to N-(4-hydroxyphenyl)retinamide (4- HPR, fenretinide). Fenretinide 81-110 haptoglobin-related protein Homo sapiens 115-118