PMID-sentid Pub_year Sent_text compound_name comp_offset prot_official_name organism prot_offset 33907421-0 2021 Downregulation of Chemokine CCL20 Involved in Myeloma Cells Resistant to Elotuzumab and Lenalidomide. Lenalidomide 88-100 C-C motif chemokine ligand 20 Homo sapiens 28-33 33839882-0 2021 Cereblon expression is a prognostic marker in newly diagnosed POEMS syndrome treated with lenalidomide plus dexamethasone. Lenalidomide 90-102 cereblon Homo sapiens 0-8 33839882-3 2021 Cereblon has been identified as the direct target of lenalidomide, and high cereblon expression is associated with better response and outcome to lenalidomide therapy in multiple myeloma patients. Lenalidomide 53-65 cereblon Homo sapiens 0-8 33839882-3 2021 Cereblon has been identified as the direct target of lenalidomide, and high cereblon expression is associated with better response and outcome to lenalidomide therapy in multiple myeloma patients. Lenalidomide 146-158 cereblon Homo sapiens 76-84 34025681-3 2021 Objective: To evaluate skin responses after treatment with low-dose lenalidomide plus dexamethasone and determine their relationship with vascular endothelial growth factor (VEGF) and hematological responses. Lenalidomide 68-80 vascular endothelial growth factor A Homo sapiens 138-172 34025681-3 2021 Objective: To evaluate skin responses after treatment with low-dose lenalidomide plus dexamethasone and determine their relationship with vascular endothelial growth factor (VEGF) and hematological responses. Lenalidomide 68-80 vascular endothelial growth factor A Homo sapiens 174-178 33989279-1 2021 Single-agent lenalidomide has modest activity in diffuse large B-cell lymphoma (DLBCL) and is thought to be more potent in activated B-cell (ABC) lymphomas, which are more treatment-resistant. Lenalidomide 13-25 ATP binding cassette subfamily B member 6 (Langereis blood group) Homo sapiens 141-144 33989279-2 2021 However, the addition of lenalidomide to rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) in randomized clinical trials has shown equivocal benefit, despite phase 2 studies that suggested otherwise. Lenalidomide 25-37 DNA damage inducible transcript 3 Homo sapiens 118-122 33839882-7 2021 In conclusion, our results emphasized the role of cereblon mRNA expression as a unique biomarker for predicting the clinical response and outcome of lenalidomide-based therapy in newly diagnosed POEMS syndrome patients. Lenalidomide 149-161 cereblon Homo sapiens 50-58 34013974-8 2021 Interestingly, lenalidomide enhanced the expression of IRF4 and the co-stimulatory molecule CD86. Lenalidomide 15-27 interferon regulatory factor 4 Homo sapiens 55-59 34013974-8 2021 Interestingly, lenalidomide enhanced the expression of IRF4 and the co-stimulatory molecule CD86. Lenalidomide 15-27 CD86 molecule Homo sapiens 92-96 34013974-10 2021 CD49d-negative CLL cases were more sensitive to lenalidomide treatment. Lenalidomide 48-60 integrin subunit alpha 4 Homo sapiens 0-5 34003300-0 2022 Lenalidomide enhances the efficacy of anti-BCMA CAR-T treatment in relapsed/refractory multiple myeloma: a case report and revies of the literature. Lenalidomide 0-12 TNF receptor superfamily member 17 Homo sapiens 43-47 34003300-0 2022 Lenalidomide enhances the efficacy of anti-BCMA CAR-T treatment in relapsed/refractory multiple myeloma: a case report and revies of the literature. Lenalidomide 0-12 CXADR pseudogene 1 Homo sapiens 48-51 34003300-1 2022 We report successful clinical experience using anti-BCMA CAR-T combined with lenalidomide in a patient who was refractory to a previous CAR-T treatment. Lenalidomide 77-89 CXADR pseudogene 1 Homo sapiens 136-139 33980611-7 2021 Genetic and pharmacological inhibition of EGR1 synergizes with the BRD4 inhibitor JQ1 or the type I interferon inducer lenalidomide in growth inhibition of ABC DLBCL both in cell cultures and xenograft mouse models. Lenalidomide 119-131 early growth response 1 Mus musculus 42-46 33972500-0 2021 Lenalidomide downregulates ACE2 protein abundance to alleviate infection by SARS-CoV-2 spike protein conditioned pseudoviruses. Lenalidomide 0-12 angiotensin converting enzyme 2 Homo sapiens 27-31 33972500-0 2021 Lenalidomide downregulates ACE2 protein abundance to alleviate infection by SARS-CoV-2 spike protein conditioned pseudoviruses. Lenalidomide 0-12 surface glycoprotein Severe acute respiratory syndrome coronavirus 2 87-92 33751979-3 2021 We firstly screened CRBN ligands as the E3 ligase moiety, then obtained a series of potent ALK degraders based on different CRBN ligands, exemplified by SIAIS091 and SIAIS001 with lenalidomide/thalidomide-based linkers. Lenalidomide 180-192 ALK receptor tyrosine kinase Homo sapiens 91-94 33751979-3 2021 We firstly screened CRBN ligands as the E3 ligase moiety, then obtained a series of potent ALK degraders based on different CRBN ligands, exemplified by SIAIS091 and SIAIS001 with lenalidomide/thalidomide-based linkers. Lenalidomide 180-192 cereblon Homo sapiens 124-128 33938033-1 2021 Thalidomide and its derivatives lenalidomide and pomalidomide, known as immunomodulatory drugs, (IMiDs) bind directly to cereblon (CRBN), a substrate receptor of an E3 ubiquitin ligase, resulting in the rapid ubiquitination and degradation of the substrate protein. Lenalidomide 32-44 cereblon Homo sapiens 131-135 33704530-7 2021 A slight increase of T-bet-positive TH1-equivalents was identifiable under lenalidomide. Lenalidomide 75-87 T-box transcription factor 21 Homo sapiens 21-26 33704530-7 2021 A slight increase of T-bet-positive TH1-equivalents was identifiable under lenalidomide. Lenalidomide 75-87 negative elongation factor complex member C/D Homo sapiens 36-39 33623139-7 2021 We modulated IKAROS protein levels both by genetic manipulation and pharmacologically by treating CLL cells with lenalidomide and avadomide (IMIDs). Lenalidomide 113-125 IKAROS family zinc finger 1 Homo sapiens 13-19 33877296-4 2021 Lenalidomide rapidly and transiently induced an activated T-cell phenotype, including HLA-DR, Tim-3, CD137, and programmed cell death protein 1 (PD-1) upregulation. Lenalidomide 0-12 hepatitis A virus cellular receptor 2 Homo sapiens 94-99 33877296-4 2021 Lenalidomide rapidly and transiently induced an activated T-cell phenotype, including HLA-DR, Tim-3, CD137, and programmed cell death protein 1 (PD-1) upregulation. Lenalidomide 0-12 TNF receptor superfamily member 9 Homo sapiens 101-106 33877296-4 2021 Lenalidomide rapidly and transiently induced an activated T-cell phenotype, including HLA-DR, Tim-3, CD137, and programmed cell death protein 1 (PD-1) upregulation. Lenalidomide 0-12 programmed cell death 1 Homo sapiens 112-143 33885752-12 2021 Lenalidomide also synergized with HexaBody-DR5/DR5, but only via its immunomodulatory effects, presumably by enhancing the antibody-dependent cellular cytotoxicity activity of HexaBody-DR5/DR5. Lenalidomide 0-12 TNF receptor superfamily member 10b Homo sapiens 43-46 33885752-12 2021 Lenalidomide also synergized with HexaBody-DR5/DR5, but only via its immunomodulatory effects, presumably by enhancing the antibody-dependent cellular cytotoxicity activity of HexaBody-DR5/DR5. Lenalidomide 0-12 TNF receptor superfamily member 10b Homo sapiens 47-50 33885752-12 2021 Lenalidomide also synergized with HexaBody-DR5/DR5, but only via its immunomodulatory effects, presumably by enhancing the antibody-dependent cellular cytotoxicity activity of HexaBody-DR5/DR5. Lenalidomide 0-12 TNF receptor superfamily member 10b Homo sapiens 47-50 33885752-12 2021 Lenalidomide also synergized with HexaBody-DR5/DR5, but only via its immunomodulatory effects, presumably by enhancing the antibody-dependent cellular cytotoxicity activity of HexaBody-DR5/DR5. Lenalidomide 0-12 TNF receptor superfamily member 10b Homo sapiens 47-50 33925565-9 2021 Moreover, combination of LAG-3 with the immunomodulatory drug (IMiD) lenalidomide significantly increased IL-2 production by T cells and antibody-dependent cytotoxicity (ADCC) mediated by NK cells. Lenalidomide 69-81 lymphocyte activating 3 Homo sapiens 25-30 33925565-9 2021 Moreover, combination of LAG-3 with the immunomodulatory drug (IMiD) lenalidomide significantly increased IL-2 production by T cells and antibody-dependent cytotoxicity (ADCC) mediated by NK cells. Lenalidomide 69-81 interleukin 2 Homo sapiens 106-110 33907421-9 2021 Conclusion: The expression of CCL20 was decreased in lenalidomide and Elotuzumab resistant U266 cells and in RRMM patients. Lenalidomide 53-65 C-C motif chemokine ligand 20 Homo sapiens 30-35 33907421-10 2021 CCL20 could therefore possibly increase the sensitivity of lenalidomide and Elotuzumab. Lenalidomide 59-71 C-C motif chemokine ligand 20 Homo sapiens 0-5 33621109-2 2021 Phase II studies demonstrated that adding the immunomodulatory agent lenalidomide to R-CHOP improved outcomes in ABC-type DLBCL. Lenalidomide 69-81 DNA damage inducible transcript 3 Homo sapiens 87-91 33555941-1 2021 PURPOSE: Lenalidomide combined with rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) (R2CHOP) in untreated diffuse large B-cell lymphoma (DLBCL) has shown promising activity, particularly in the activated B-cell-like (ABC) subtype. Lenalidomide 9-21 DNA damage inducible transcript 3 Homo sapiens 113-117 33621109-3 2021 The goal of the global, phase III ROBUST study was to compare lenalidomide plus R-CHOP (R2-CHOP) with placebo/R-CHOP in previously untreated, ABC-type DLBCL. Lenalidomide 62-74 ribonucleotide reductase regulatory subunit M2 Homo sapiens 88-95 33444079-3 2021 In combination with R-CHOP (R2-CHOP), lenalidomide has an acceptable level of toxicity and may mitigate the negative prognosis of the non-germinal center B-cell-like phenotype. Lenalidomide 38-50 DNA damage inducible transcript 3 Homo sapiens 31-35 33856277-2 2021 Ibrutinib and lenalidomide are synergistic in vitro in ABC DLBCL and may augment salvage chemotherapy. Lenalidomide 14-26 ATP binding cassette subfamily B member 6 (Langereis blood group) Homo sapiens 55-58 33733978-6 2021 The combination of lenalidomide plus dexamethasone with an anti-CD38 MoAb such as daratumumab and a proteasome inhibitor such as bortezomib is currently one of the most potent first line treatment regimens for MM. Lenalidomide 19-31 CD38 molecule Homo sapiens 64-68 34007370-10 2021 Due to MYC-positivity, lenalidomide was added to the therapy regimen. Lenalidomide 23-35 MYC proto-oncogene, bHLH transcription factor Homo sapiens 7-10 34007370-12 2021 Lenalidomide is an oral immunomodulatory drug that downregulates MYC gene and is commonly used in patients with multiple myeloma. Lenalidomide 0-12 MYC proto-oncogene, bHLH transcription factor Homo sapiens 65-68 33439748-2 2021 We investigated whether treatment with lenalidomide (LEN), which augments erythropoietin receptor signaling in vitro, can restore and improve hemoglobin response to epoetin (EPO) alfa in patients with lower-risk, non-del(5q) MDS who have anemia that is refractory to or have low probability of benefit from treatment with recombinant erythropoietin. Lenalidomide 39-51 erythropoietin receptor Homo sapiens 74-97 33439748-2 2021 We investigated whether treatment with lenalidomide (LEN), which augments erythropoietin receptor signaling in vitro, can restore and improve hemoglobin response to epoetin (EPO) alfa in patients with lower-risk, non-del(5q) MDS who have anemia that is refractory to or have low probability of benefit from treatment with recombinant erythropoietin. Lenalidomide 39-51 erythropoietin Homo sapiens 165-172 33439748-2 2021 We investigated whether treatment with lenalidomide (LEN), which augments erythropoietin receptor signaling in vitro, can restore and improve hemoglobin response to epoetin (EPO) alfa in patients with lower-risk, non-del(5q) MDS who have anemia that is refractory to or have low probability of benefit from treatment with recombinant erythropoietin. Lenalidomide 39-51 erythropoietin Homo sapiens 174-177 33439748-2 2021 We investigated whether treatment with lenalidomide (LEN), which augments erythropoietin receptor signaling in vitro, can restore and improve hemoglobin response to epoetin (EPO) alfa in patients with lower-risk, non-del(5q) MDS who have anemia that is refractory to or have low probability of benefit from treatment with recombinant erythropoietin. Lenalidomide 39-51 erythropoietin Homo sapiens 74-88 33439748-2 2021 We investigated whether treatment with lenalidomide (LEN), which augments erythropoietin receptor signaling in vitro, can restore and improve hemoglobin response to epoetin (EPO) alfa in patients with lower-risk, non-del(5q) MDS who have anemia that is refractory to or have low probability of benefit from treatment with recombinant erythropoietin. Lenalidomide 53-56 erythropoietin receptor Homo sapiens 74-97 33439748-2 2021 We investigated whether treatment with lenalidomide (LEN), which augments erythropoietin receptor signaling in vitro, can restore and improve hemoglobin response to epoetin (EPO) alfa in patients with lower-risk, non-del(5q) MDS who have anemia that is refractory to or have low probability of benefit from treatment with recombinant erythropoietin. Lenalidomide 53-56 erythropoietin Homo sapiens 165-172 33439748-2 2021 We investigated whether treatment with lenalidomide (LEN), which augments erythropoietin receptor signaling in vitro, can restore and improve hemoglobin response to epoetin (EPO) alfa in patients with lower-risk, non-del(5q) MDS who have anemia that is refractory to or have low probability of benefit from treatment with recombinant erythropoietin. Lenalidomide 53-56 erythropoietin Homo sapiens 174-177 33439748-2 2021 We investigated whether treatment with lenalidomide (LEN), which augments erythropoietin receptor signaling in vitro, can restore and improve hemoglobin response to epoetin (EPO) alfa in patients with lower-risk, non-del(5q) MDS who have anemia that is refractory to or have low probability of benefit from treatment with recombinant erythropoietin. Lenalidomide 53-56 erythropoietin Homo sapiens 74-88 33139584-6 2021 The anti-PD-L1 effects on multiple myeloma may be related to a decrease in the immunosuppressive myeloidderived suppressor cells (MDSCs), but there were no changes in the splenic MDSCs after combined treatment with lenalidomide and the anti-PD-L1 antibody. Lenalidomide 215-227 CD274 antigen Mus musculus 9-14 33757580-3 2021 Cereblon (CRBN) is a key mediator of the bioactivities of IMiDs such as lenalidomide. Lenalidomide 72-84 cereblon Homo sapiens 10-14 33757580-7 2021 METHODS: We established lenalidomide-resistant cells by knocking down CRBN with RNAi-mediated downregulation or knocking out CRBN using CRISPR-Cas9 in MM cells. Lenalidomide 24-36 cereblon Homo sapiens 70-74 33757580-12 2021 CRBN-deficient cells showed lenalidomide-induced upregulation of phosphorylated glycogen synthase kinase-3 (p-GSK-3) and c-Myc phosphorylation. Lenalidomide 28-40 cereblon Homo sapiens 0-4 33757580-12 2021 CRBN-deficient cells showed lenalidomide-induced upregulation of phosphorylated glycogen synthase kinase-3 (p-GSK-3) and c-Myc phosphorylation. Lenalidomide 28-40 MYC proto-oncogene, bHLH transcription factor Homo sapiens 121-126 33757580-15 2021 The dual HDAC and PI3K inhibitor, CUDC-907, exhibited cytotoxic and immunotherapy-enhancing effects in MM cells, including multi-drug-resistant lines and primary cells from lenalidomide-resistant patients. Lenalidomide 173-185 histone deacetylase 9 Homo sapiens 9-13 33746969-3 2021 In the absence of myeloma cells, lenalidomide and pomalidomide induce CD4+ T cell secretion of IL-2 and indirect activation of Natural Killer (NK) cells. Lenalidomide 33-45 CD4 molecule Homo sapiens 70-73 33746969-3 2021 In the absence of myeloma cells, lenalidomide and pomalidomide induce CD4+ T cell secretion of IL-2 and indirect activation of Natural Killer (NK) cells. Lenalidomide 33-45 interleukin 2 Homo sapiens 95-99 33534410-2 2021 Lenalidomide prevents the proliferation of multiple myeloma cells through binding to cereblon and promoting the ubiquitinational degradation of IKZF1 (Ikaros)/IKZF3 (Aiolos). Lenalidomide 0-12 IKAROS family zinc finger 1 Homo sapiens 144-149 33534410-2 2021 Lenalidomide prevents the proliferation of multiple myeloma cells through binding to cereblon and promoting the ubiquitinational degradation of IKZF1 (Ikaros)/IKZF3 (Aiolos). Lenalidomide 0-12 IKAROS family zinc finger 1 Homo sapiens 151-157 33534410-2 2021 Lenalidomide prevents the proliferation of multiple myeloma cells through binding to cereblon and promoting the ubiquitinational degradation of IKZF1 (Ikaros)/IKZF3 (Aiolos). Lenalidomide 0-12 IKAROS family zinc finger 3 Homo sapiens 159-164 33534410-2 2021 Lenalidomide prevents the proliferation of multiple myeloma cells through binding to cereblon and promoting the ubiquitinational degradation of IKZF1 (Ikaros)/IKZF3 (Aiolos). Lenalidomide 0-12 IKAROS family zinc finger 3 Homo sapiens 166-172 33563611-8 2021 Lenalidomide is known to exert its beneficial effects by modulating the CRBN-CRL4 E3 ligase to ubiquitinate and degrade the transcription factor IKAROS family zinc finger 1 (IKZF1). Lenalidomide 0-12 cereblon Homo sapiens 72-76 33563611-8 2021 Lenalidomide is known to exert its beneficial effects by modulating the CRBN-CRL4 E3 ligase to ubiquitinate and degrade the transcription factor IKAROS family zinc finger 1 (IKZF1). Lenalidomide 0-12 interleukin 17 receptor B Homo sapiens 77-81 33563611-8 2021 Lenalidomide is known to exert its beneficial effects by modulating the CRBN-CRL4 E3 ligase to ubiquitinate and degrade the transcription factor IKAROS family zinc finger 1 (IKZF1). Lenalidomide 0-12 IKAROS family zinc finger 1 Homo sapiens 145-172 33563611-8 2021 Lenalidomide is known to exert its beneficial effects by modulating the CRBN-CRL4 E3 ligase to ubiquitinate and degrade the transcription factor IKAROS family zinc finger 1 (IKZF1). Lenalidomide 0-12 IKAROS family zinc finger 1 Homo sapiens 174-179 33563611-9 2021 In M2-like MAMs, we observed enhanced IKZF1 levels that vanished through treatment with lenalidomide, yielding MAMs with a bioenergetic profile, T-cell stimulatory properties, and loss of tumor-promoting capabilities that resemble M1 cells. Lenalidomide 88-100 IKAROS family zinc finger 1 Homo sapiens 38-43 32779949-5 2020 Here we report the design, synthesis, and evaluation of a proteolysis targeting chimaera (PROTAC) based on the combination of PARP-1 inhibitor olaparib and the CRBN (cereblon) ligand lenalidomide. Lenalidomide 183-195 cereblon Homo sapiens 160-164 33408186-4 2021 We developed ON and OFF switches for CAR T cells using the clinically approved drug lenalidomide, which mediates the proteasomal degradation of several target proteins by inducing interactions between the CRL4CRBN E3 ubiquitin ligase and a C2H2 zinc finger degron motif. Lenalidomide 84-96 nuclear receptor subfamily 1 group I member 3 Homo sapiens 37-40 33408186-7 2021 Subtherapeutic lenalidomide concentrations controlled the effector functions of ON- and OFF-switch CAR T cells. Lenalidomide 15-27 nuclear receptor subfamily 1 group I member 3 Homo sapiens 99-102 33357426-9 2020 PC4 knockdown in human B cell lymphoma and myeloma cells reduces IKAROS protein as an anticancer drug, lenalidomide. Lenalidomide 103-115 SUB1 regulator of transcription Homo sapiens 0-3 33357426-9 2020 PC4 knockdown in human B cell lymphoma and myeloma cells reduces IKAROS protein as an anticancer drug, lenalidomide. Lenalidomide 103-115 IKAROS family zinc finger 1 Homo sapiens 65-71 33284947-4 2020 The 5 analyzed proteins involved in lenalidomide"s mode of action were associated with time to progression (TTP); low levels of cereblon and IRF4 protein and high levels of Ikaros, AGO2, and Aiolos were significantly associated with shorter TTP. Lenalidomide 36-48 interferon regulatory factor 4 Homo sapiens 141-145 33284947-4 2020 The 5 analyzed proteins involved in lenalidomide"s mode of action were associated with time to progression (TTP); low levels of cereblon and IRF4 protein and high levels of Ikaros, AGO2, and Aiolos were significantly associated with shorter TTP. Lenalidomide 36-48 IKAROS family zinc finger 1 Homo sapiens 173-179 33284947-4 2020 The 5 analyzed proteins involved in lenalidomide"s mode of action were associated with time to progression (TTP); low levels of cereblon and IRF4 protein and high levels of Ikaros, AGO2, and Aiolos were significantly associated with shorter TTP. Lenalidomide 36-48 argonaute RISC catalytic component 2 Homo sapiens 181-185 33284947-4 2020 The 5 analyzed proteins involved in lenalidomide"s mode of action were associated with time to progression (TTP); low levels of cereblon and IRF4 protein and high levels of Ikaros, AGO2, and Aiolos were significantly associated with shorter TTP. Lenalidomide 36-48 IKAROS family zinc finger 3 Homo sapiens 191-197 33273169-7 2020 Agents such as the Bruton tyrosine kinase (BTK) inhibitor ibrutinib or immunomodulatory drugs such as lenalidomide and pomalidomide have shown promising response rates in the relapsed setting. Lenalidomide 102-114 Bruton tyrosine kinase Homo sapiens 43-46 33256379-0 2020 Treatment of patients with MYC rearrangement positive large B-cell lymphoma with R-CHOP plus lenalidomide: results of a multicenter HOVON phase II trial. Lenalidomide 93-105 MYC proto-oncogene, bHLH transcription factor Homo sapiens 27-30 33256379-3 2020 Lenalidomide is an oral immunomodulatory drug which downregulates MYC and its target genes thereby providing support using lenalidomide as additional therapeutic option for MYC+ LBCL. Lenalidomide 0-12 MYC proto-oncogene, bHLH transcription factor Homo sapiens 66-69 33256379-3 2020 Lenalidomide is an oral immunomodulatory drug which downregulates MYC and its target genes thereby providing support using lenalidomide as additional therapeutic option for MYC+ LBCL. Lenalidomide 0-12 MYC proto-oncogene, bHLH transcription factor Homo sapiens 173-176 33256379-3 2020 Lenalidomide is an oral immunomodulatory drug which downregulates MYC and its target genes thereby providing support using lenalidomide as additional therapeutic option for MYC+ LBCL. Lenalidomide 123-135 MYC proto-oncogene, bHLH transcription factor Homo sapiens 66-69 33256379-3 2020 Lenalidomide is an oral immunomodulatory drug which downregulates MYC and its target genes thereby providing support using lenalidomide as additional therapeutic option for MYC+ LBCL. Lenalidomide 123-135 MYC proto-oncogene, bHLH transcription factor Homo sapiens 173-176 33256379-4 2020 A phase II trial was conducted evaluating the efficacy of lenalidomide (15 mg day 1-14) in combination with R-CHOP (R2CHOP) in newly diagnosed MYC+ LBCL patients identified through a nationwide MYC-FISH screening program. Lenalidomide 58-70 MYC proto-oncogene, bHLH transcription factor Homo sapiens 143-146 33085797-1 2021 Carfilzomib-lenalidomide-dexamethasone (KRd) has been approved for the treatment of relapsed/refractory multiple myeloma (RRMM). Lenalidomide 12-24 calcium responsive transcription factor Homo sapiens 0-11 33375215-4 2020 Personalized low dose rIL-2 in combination with either lenalidomide or venetoclax mediates natural killer stimulation and is an effective non-toxic immunotherapy administered in the outpatient setting for poor prognosis CLL. Lenalidomide 55-67 interleukin 2 Rattus norvegicus 22-27 33392195-7 2020 Using myeloma as a model system, we further revealed that either inhibition or genetic depletion of CASP-8 enhances the anti-myeloma activity of lenalidomide (Len) by impairing CRBN cleavage, leading to the attenuated IKZF1 and IKZF3 protein levels and the reduced viability of myeloma cell lines and primary myeloma cells from patients. Lenalidomide 145-157 caspase 8 Homo sapiens 100-106 33392195-7 2020 Using myeloma as a model system, we further revealed that either inhibition or genetic depletion of CASP-8 enhances the anti-myeloma activity of lenalidomide (Len) by impairing CRBN cleavage, leading to the attenuated IKZF1 and IKZF3 protein levels and the reduced viability of myeloma cell lines and primary myeloma cells from patients. Lenalidomide 145-157 cereblon Homo sapiens 177-181 33392195-7 2020 Using myeloma as a model system, we further revealed that either inhibition or genetic depletion of CASP-8 enhances the anti-myeloma activity of lenalidomide (Len) by impairing CRBN cleavage, leading to the attenuated IKZF1 and IKZF3 protein levels and the reduced viability of myeloma cell lines and primary myeloma cells from patients. Lenalidomide 145-157 IKAROS family zinc finger 1 Homo sapiens 218-223 33392195-7 2020 Using myeloma as a model system, we further revealed that either inhibition or genetic depletion of CASP-8 enhances the anti-myeloma activity of lenalidomide (Len) by impairing CRBN cleavage, leading to the attenuated IKZF1 and IKZF3 protein levels and the reduced viability of myeloma cell lines and primary myeloma cells from patients. Lenalidomide 145-157 IKAROS family zinc finger 3 Homo sapiens 228-233 33392195-7 2020 Using myeloma as a model system, we further revealed that either inhibition or genetic depletion of CASP-8 enhances the anti-myeloma activity of lenalidomide (Len) by impairing CRBN cleavage, leading to the attenuated IKZF1 and IKZF3 protein levels and the reduced viability of myeloma cell lines and primary myeloma cells from patients. Lenalidomide 159-162 caspase 8 Homo sapiens 100-106 33392195-7 2020 Using myeloma as a model system, we further revealed that either inhibition or genetic depletion of CASP-8 enhances the anti-myeloma activity of lenalidomide (Len) by impairing CRBN cleavage, leading to the attenuated IKZF1 and IKZF3 protein levels and the reduced viability of myeloma cell lines and primary myeloma cells from patients. Lenalidomide 159-162 cereblon Homo sapiens 177-181 33392195-7 2020 Using myeloma as a model system, we further revealed that either inhibition or genetic depletion of CASP-8 enhances the anti-myeloma activity of lenalidomide (Len) by impairing CRBN cleavage, leading to the attenuated IKZF1 and IKZF3 protein levels and the reduced viability of myeloma cell lines and primary myeloma cells from patients. Lenalidomide 159-162 IKAROS family zinc finger 1 Homo sapiens 218-223 33392195-7 2020 Using myeloma as a model system, we further revealed that either inhibition or genetic depletion of CASP-8 enhances the anti-myeloma activity of lenalidomide (Len) by impairing CRBN cleavage, leading to the attenuated IKZF1 and IKZF3 protein levels and the reduced viability of myeloma cell lines and primary myeloma cells from patients. Lenalidomide 159-162 IKAROS family zinc finger 3 Homo sapiens 228-233 33392195-8 2020 The present study discovered that the stability of the substrate receptor of an E3 ligase can be modulated by CASP-8 and suggested that administration of CASP-8 inhibitors enhances the overall effectiveness of Len-based combination therapy in myeloma. Lenalidomide 210-213 caspase 8 Homo sapiens 110-116 33392195-8 2020 The present study discovered that the stability of the substrate receptor of an E3 ligase can be modulated by CASP-8 and suggested that administration of CASP-8 inhibitors enhances the overall effectiveness of Len-based combination therapy in myeloma. Lenalidomide 210-213 caspase 8 Homo sapiens 154-160 32779949-5 2020 Here we report the design, synthesis, and evaluation of a proteolysis targeting chimaera (PROTAC) based on the combination of PARP-1 inhibitor olaparib and the CRBN (cereblon) ligand lenalidomide. Lenalidomide 183-195 cereblon Homo sapiens 166-174 33227124-1 2020 The immunomodulatory drugs lenalidomide and pomalidomide enhance the potency of AMG 701 in multiple myeloma preclinical models. Lenalidomide 27-39 amelogenin X-linked Homo sapiens 80-83 32852308-4 2020 Also the SLAMF7-targeting antibody, elotuzumab, improves the survival of relapsed/refractory multiple myeloma patients, when it is combined with either lenalidomide or pomalidomide. Lenalidomide 152-164 SLAM family member 7 Homo sapiens 9-15 33124184-9 2020 Lenalidomide non-significantly decreased the level of CD8+ T cells but increased CD4+ T cells leading to increased CD4+ /CD8+ T cell ratio. Lenalidomide 0-12 CD8a molecule Homo sapiens 54-57 33124184-9 2020 Lenalidomide non-significantly decreased the level of CD8+ T cells but increased CD4+ T cells leading to increased CD4+ /CD8+ T cell ratio. Lenalidomide 0-12 CD4 molecule Homo sapiens 81-84 33124184-9 2020 Lenalidomide non-significantly decreased the level of CD8+ T cells but increased CD4+ T cells leading to increased CD4+ /CD8+ T cell ratio. Lenalidomide 0-12 CD4 molecule Homo sapiens 115-118 33124184-9 2020 Lenalidomide non-significantly decreased the level of CD8+ T cells but increased CD4+ T cells leading to increased CD4+ /CD8+ T cell ratio. Lenalidomide 0-12 CD8a molecule Homo sapiens 121-124 32932183-9 2020 Our data shows that lenalidomide alleviated rotenone-induced motor impairments and deficits in dopamine-related behaviors and resulted in increased levels of tumor necrosis factor-alpha and calcium-binding protein B in the SNpc. Lenalidomide 20-32 tumor necrosis factor Rattus norvegicus 158-185 32958952-6 2020 Pomalidomide is more effective than lenalidomide in degrading ARID2 and is capable of inhibiting MYC expression and proliferation in lenalidomide-resistant cell lines. Lenalidomide 36-48 AT-rich interaction domain 2 Homo sapiens 62-67 32958952-6 2020 Pomalidomide is more effective than lenalidomide in degrading ARID2 and is capable of inhibiting MYC expression and proliferation in lenalidomide-resistant cell lines. Lenalidomide 133-145 MYC proto-oncogene, bHLH transcription factor Homo sapiens 97-100 32958952-8 2020 These findings suggest that ARID2 is a promising target for overcoming lenalidomide resistance in patients with multiple myeloma. Lenalidomide 71-83 AT-rich interaction domain 2 Homo sapiens 28-33 32932183-10 2020 Moreover, chronic lenalidomide treatment resulted increase in transforming growth factor immunoreactivity and brain derived neurotrophic factor expression in the SNPc. Lenalidomide 18-30 brain-derived neurotrophic factor Rattus norvegicus 110-143 32350909-0 2020 Clinical benefit of ixazomib plus lenalidomide-dexamethasone in myeloma patients with non-canonical NF-kappaB pathway activation. Lenalidomide 34-46 nuclear factor kappa B subunit 1 Homo sapiens 100-109 32865967-1 2020 Cereblon (CRBN) is an E3 ligase adapter protein that can be reprogrammed by imide-class compounds such as thalidomide, lenalidomide, and pomalidomide to induce the degradation of neo-substrate proteins. Lenalidomide 119-131 cereblon Homo sapiens 0-8 32865967-1 2020 Cereblon (CRBN) is an E3 ligase adapter protein that can be reprogrammed by imide-class compounds such as thalidomide, lenalidomide, and pomalidomide to induce the degradation of neo-substrate proteins. Lenalidomide 119-131 cereblon Homo sapiens 10-14 33190445-0 2020 [Lenalidomide for myelodysplastic syndrome with excess blasts with germline DDX41 mutation: a case report and literatures review]. Lenalidomide 1-13 DEAD-box helicase 41 Homo sapiens 76-81 33008427-5 2020 METHODS: MUK eight is a randomised, controlled, open, parallel group, multi-centre phase II trial that will recruit patients with RRMM who have relapsed after treatment with thalidomide, lenalidomide, and a proteasome inhibitor. Lenalidomide 187-199 mitogen-activated protein kinase kinase kinase 12 Homo sapiens 9-12 32704173-1 2020 BACKGROUND: Purpose: The combination of a mammalian target of rapamycin inhibitor and lenalidomide showed enhanced preclinical cytotoxicity. Lenalidomide 86-98 mechanistic target of rapamycin kinase Homo sapiens 42-71 32669063-5 2020 In RRMM, the annual budget savings of lenalidomide entry were estimated at EGP-(-1,103,969, -3,362,793 and -5,949,228) at year 1, year 2 and year 3 respectively. Lenalidomide 38-50 tumor associated calcium signal transducer 2 Homo sapiens 75-82 32843459-4 2020 Given how aggressive and difficult to treat ALK+ LBCL is, further research is warranted to more completely elucidate the mechanism of action of lenalidomide in ALK+ LBCL and its role in treatment. Lenalidomide 144-156 ALK receptor tyrosine kinase Homo sapiens 44-47 32982239-0 2020 Nanomicellar Lenalidomide-Fenretinide Combination Suppresses Tumor Growth in an MYCN Amplified Neuroblastoma Tumor. Lenalidomide 13-25 MYCN proto-oncogene, bHLH transcription factor Homo sapiens 80-84 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. Lenalidomide 87-99 neurotrophic receptor tyrosine kinase 2 Homo sapiens 211-215 32761232-4 2020 Both lenalidomide and pomalidomide dose-dependently inhibited interleukin-12 (IL-12) and TNF production and STAT4 expression, and enhanced IL-10 production in response to LPS. Lenalidomide 5-17 tumor necrosis factor Homo sapiens 89-92 32761232-4 2020 Both lenalidomide and pomalidomide dose-dependently inhibited interleukin-12 (IL-12) and TNF production and STAT4 expression, and enhanced IL-10 production in response to LPS. Lenalidomide 5-17 signal transducer and activator of transcription 4 Homo sapiens 108-113 32761232-4 2020 Both lenalidomide and pomalidomide dose-dependently inhibited interleukin-12 (IL-12) and TNF production and STAT4 expression, and enhanced IL-10 production in response to LPS. Lenalidomide 5-17 interleukin 10 Homo sapiens 139-144 32761232-6 2020 In 46 myeloma patients, serum CCL17 levels at the onset of lenalidomide-associated rash were significantly higher than those without rashes during lenalidomide treatment and those before treatment. Lenalidomide 59-71 C-C motif chemokine ligand 17 Homo sapiens 30-35 32761232-6 2020 In 46 myeloma patients, serum CCL17 levels at the onset of lenalidomide-associated rash were significantly higher than those without rashes during lenalidomide treatment and those before treatment. Lenalidomide 147-159 C-C motif chemokine ligand 17 Homo sapiens 30-35 32761232-7 2020 Furthermore, serum CCL17 levels in patients who achieved a very good partial response (VGPR) were significantly higher compared with a less than VGPR during lenalidomide treatment. Lenalidomide 157-169 C-C motif chemokine ligand 17 Homo sapiens 19-24 32618603-0 2020 A Case of Lenalidomide-Associated Papulopustular Rash: Beyond the Epidermal Growth Factor Receptor Inhibitor Spectrum. Lenalidomide 10-22 epidermal growth factor receptor Homo sapiens 66-98 32898244-0 2020 The immunomodulatory drugs lenalidomide and pomalidomide enhance the potency of AMG 701 in multiple myeloma preclinical models. Lenalidomide 27-39 amelogenin X-linked Homo sapiens 80-83 32632752-4 2020 While lenalidomide and pomalidomide induced greater degradation of Ikaros and Aiolos in myeloma cells, BTX306 more potently reduced levels of GSPT1, eRF1, CK1alpha, MCL-1, and c-MYC. Lenalidomide 6-18 IKAROS family zinc finger 1 Mus musculus 67-73 32632752-4 2020 While lenalidomide and pomalidomide induced greater degradation of Ikaros and Aiolos in myeloma cells, BTX306 more potently reduced levels of GSPT1, eRF1, CK1alpha, MCL-1, and c-MYC. Lenalidomide 6-18 IKAROS family zinc finger 3 Mus musculus 78-84 32591583-0 2020 Development of Label-Free Impedimetric Immunosensors for IKZF1 and IKZF3 Femtomolar Detection for Monitoring Multiple Myeloma Patients Treated with Lenalidomide. Lenalidomide 148-160 IKAROS family zinc finger 3 Homo sapiens 67-72 32591583-2 2020 Lenalidomide destroys MM cells by inducing ubiquitination and the consequent degradation of Ikaros family zinc finger proteins 1 and 3 (IKZF1 and IKZF3). Lenalidomide 0-12 IKAROS family zinc finger 1 Homo sapiens 92-134 32591583-2 2020 Lenalidomide destroys MM cells by inducing ubiquitination and the consequent degradation of Ikaros family zinc finger proteins 1 and 3 (IKZF1 and IKZF3). Lenalidomide 0-12 IKAROS family zinc finger 1 Homo sapiens 136-141 32591583-2 2020 Lenalidomide destroys MM cells by inducing ubiquitination and the consequent degradation of Ikaros family zinc finger proteins 1 and 3 (IKZF1 and IKZF3). Lenalidomide 0-12 IKAROS family zinc finger 3 Homo sapiens 146-151 32591583-3 2020 High expression of IKZF1 and IKZF3 in MM results in less sensitivity to lenalidomide treatment and possible cytotoxic effect. Lenalidomide 72-84 IKAROS family zinc finger 1 Homo sapiens 19-24 32591583-3 2020 High expression of IKZF1 and IKZF3 in MM results in less sensitivity to lenalidomide treatment and possible cytotoxic effect. Lenalidomide 72-84 IKAROS family zinc finger 3 Homo sapiens 29-34 32317208-0 2020 Discovery and biological evaluation of proteolysis targeting chimeras (PROTACs) as an EGFR degraders based on osimertinib and lenalidomide. Lenalidomide 126-138 epidermal growth factor receptor Homo sapiens 86-90 32843459-4 2020 Given how aggressive and difficult to treat ALK+ LBCL is, further research is warranted to more completely elucidate the mechanism of action of lenalidomide in ALK+ LBCL and its role in treatment. Lenalidomide 144-156 ALK receptor tyrosine kinase Homo sapiens 160-163 32419317-4 2020 METHODS: An ALF and AKI mouse model was generated using LPS/D-Gal, and a TNF-alpha inhibitor, CC-5013, was used to treat the mice. Lenalidomide 94-101 tumor necrosis factor Mus musculus 73-82 32419317-7 2020 RESULTS: Treatment with CC-5013 decreased the activation of TNF-alpha/HMGB1 pathway and pyroptosis in the treated mice and cells compared with the control mice and cells. Lenalidomide 24-31 tumor necrosis factor Mus musculus 60-69 32419317-7 2020 RESULTS: Treatment with CC-5013 decreased the activation of TNF-alpha/HMGB1 pathway and pyroptosis in the treated mice and cells compared with the control mice and cells. Lenalidomide 24-31 high mobility group box 1 Mus musculus 70-75 32437909-1 2020 Our previous study revealed that expression of G protein-coupled receptor 68 (GPR68) was upregulated in MDSL cells, a cell line representing Myelodysplastic Syndromes (MDS), in response to lenalidomide (LEN), and mediated a calcium/calpain proapoptotic pathway. Lenalidomide 189-201 G protein-coupled receptor 68 Homo sapiens 47-76 32437909-1 2020 Our previous study revealed that expression of G protein-coupled receptor 68 (GPR68) was upregulated in MDSL cells, a cell line representing Myelodysplastic Syndromes (MDS), in response to lenalidomide (LEN), and mediated a calcium/calpain proapoptotic pathway. Lenalidomide 189-201 G protein-coupled receptor 68 Homo sapiens 78-83 32437909-1 2020 Our previous study revealed that expression of G protein-coupled receptor 68 (GPR68) was upregulated in MDSL cells, a cell line representing Myelodysplastic Syndromes (MDS), in response to lenalidomide (LEN), and mediated a calcium/calpain proapoptotic pathway. Lenalidomide 203-206 G protein-coupled receptor 68 Homo sapiens 47-76 32437909-1 2020 Our previous study revealed that expression of G protein-coupled receptor 68 (GPR68) was upregulated in MDSL cells, a cell line representing Myelodysplastic Syndromes (MDS), in response to lenalidomide (LEN), and mediated a calcium/calpain proapoptotic pathway. Lenalidomide 203-206 G protein-coupled receptor 68 Homo sapiens 78-83 32437909-8 2020 Our study suggests a novel mechanism of action of LEN in mediating cytotoxicity in MDS/AML via upregulating RCAN1 thus inhibiting the CaN prosurvival pathway. Lenalidomide 50-53 regulator of calcineurin 1 Homo sapiens 108-113 32064987-6 2020 This analysis suggests patients with lower EPO levels experience the strongest benefit from lenalidomide. Lenalidomide 92-104 erythropoietin Homo sapiens 43-46 32321757-5 2020 Pharmacological targeting of IKZF3 with the drug lenalidomide showed that IKZF3 is required for anti-CD3/CD28 mAb-mediated induction of IL-10 but is dispensable for ex vivo IL-10 expression. Lenalidomide 49-61 IKAROS family zinc finger 3 Homo sapiens 29-34 32321757-5 2020 Pharmacological targeting of IKZF3 with the drug lenalidomide showed that IKZF3 is required for anti-CD3/CD28 mAb-mediated induction of IL-10 but is dispensable for ex vivo IL-10 expression. Lenalidomide 49-61 IKAROS family zinc finger 3 Homo sapiens 74-79 32321757-5 2020 Pharmacological targeting of IKZF3 with the drug lenalidomide showed that IKZF3 is required for anti-CD3/CD28 mAb-mediated induction of IL-10 but is dispensable for ex vivo IL-10 expression. Lenalidomide 49-61 CD28 molecule Homo sapiens 105-109 32321757-5 2020 Pharmacological targeting of IKZF3 with the drug lenalidomide showed that IKZF3 is required for anti-CD3/CD28 mAb-mediated induction of IL-10 but is dispensable for ex vivo IL-10 expression. Lenalidomide 49-61 interleukin 10 Homo sapiens 136-141 32414180-5 2020 For example, lenalidomide and pomalidomide, well-known thalidomide derivatives, degrade the neosubstrates Ikaros and Aiolos, resulting in anti-proliferative effects in multiple myeloma. Lenalidomide 13-25 IKAROS family zinc finger 1 Homo sapiens 106-112 32414180-5 2020 For example, lenalidomide and pomalidomide, well-known thalidomide derivatives, degrade the neosubstrates Ikaros and Aiolos, resulting in anti-proliferative effects in multiple myeloma. Lenalidomide 13-25 IKAROS family zinc finger 3 Homo sapiens 117-123 32251398-8 2020 Megakaryocytic gene-promoter analyses suggested that LEN-induced degradation of IKZF1 enables a RUNX1-GATA2 complex to drive megakaryocytic differentiation. Lenalidomide 53-56 RUNX family transcription factor 1 Homo sapiens 96-101 32251398-8 2020 Megakaryocytic gene-promoter analyses suggested that LEN-induced degradation of IKZF1 enables a RUNX1-GATA2 complex to drive megakaryocytic differentiation. Lenalidomide 53-56 GATA binding protein 2 Homo sapiens 102-107 32466489-8 2020 The CRBN-targeting immunomodulatory drug lenalidomide and the cullin E3 ligase inhibitor MLN4924 promotes and attenuates, respectively, proteasomal degradation of ClC-2. Lenalidomide 41-53 cereblon Homo sapiens 4-8 32466489-8 2020 The CRBN-targeting immunomodulatory drug lenalidomide and the cullin E3 ligase inhibitor MLN4924 promotes and attenuates, respectively, proteasomal degradation of ClC-2. Lenalidomide 41-53 chloride voltage-gated channel 2 Homo sapiens 163-168 32397113-3 2020 The computer inferred genomic abnormalities associating with lenalidomide treatment response in non-del(5q) MDS to include trisomy 8, del(20q), or RUNX1 loss of function mutations. Lenalidomide 61-73 RUNX family transcription factor 1 Homo sapiens 147-152 32397113-4 2020 Genomic abnormalities associating with lenalidomide resistance in non-del(5q) MDS patients included mutations in SF3B1, TET2, WNT3A amplification, MCL1 amplification, and/or PSEN2 amplification. Lenalidomide 39-51 splicing factor 3b subunit 1 Homo sapiens 113-118 32397113-4 2020 Genomic abnormalities associating with lenalidomide resistance in non-del(5q) MDS patients included mutations in SF3B1, TET2, WNT3A amplification, MCL1 amplification, and/or PSEN2 amplification. Lenalidomide 39-51 tet methylcytosine dioxygenase 2 Homo sapiens 120-124 32397113-4 2020 Genomic abnormalities associating with lenalidomide resistance in non-del(5q) MDS patients included mutations in SF3B1, TET2, WNT3A amplification, MCL1 amplification, and/or PSEN2 amplification. Lenalidomide 39-51 Wnt family member 3A Homo sapiens 126-131 32397113-4 2020 Genomic abnormalities associating with lenalidomide resistance in non-del(5q) MDS patients included mutations in SF3B1, TET2, WNT3A amplification, MCL1 amplification, and/or PSEN2 amplification. Lenalidomide 39-51 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 147-151 32397113-4 2020 Genomic abnormalities associating with lenalidomide resistance in non-del(5q) MDS patients included mutations in SF3B1, TET2, WNT3A amplification, MCL1 amplification, and/or PSEN2 amplification. Lenalidomide 39-51 presenilin 2 Homo sapiens 174-179 32435618-6 2020 Among the novel anti-myeloma IT-based agents, anti-CD38 monoclonal antibodies (mAbs) are now becoming the new backbone of treatment for NTE patients, in association with lenalidomide and dexamethasone. Lenalidomide 170-182 CD38 molecule Homo sapiens 51-55 32248251-11 2020 Thus, VMP induction therapy followed by Rd consolidation and lenalidomide maintenance is considered a well-tolerated and effective regimen in transplant-ineligible NDMM. Lenalidomide 61-73 neurensin 1 Homo sapiens 6-9 32323826-10 2020 Western blot analysis and RT-qPCR revealed that p53 activation and the expression of p21 were increased in glioma cells treated with lenalidomide. Lenalidomide 133-145 tumor protein p53 Homo sapiens 48-51 32323826-10 2020 Western blot analysis and RT-qPCR revealed that p53 activation and the expression of p21 were increased in glioma cells treated with lenalidomide. Lenalidomide 133-145 H3 histone pseudogene 16 Homo sapiens 85-88 32373374-5 2020 Lenalidomide, an immunosuppressant, inhibits TNF. Lenalidomide 0-12 tumor necrosis factor Homo sapiens 45-48 32411498-2 2020 We have previously found that GPR68 induces a proapoptotic pathway in bone marrow (BM) cells from the patients with myelodysplastic syndromes (MDS) after treated with lenalidomide. Lenalidomide 167-179 G protein-coupled receptor 68 Homo sapiens 30-35 31702836-0 2020 Lenalidomide maintenance for diffuse large B-cell lymphoma patients responding to R-CHOP: quality of life, dosing, and safety results from the randomised controlled REMARC study. Lenalidomide 0-12 DNA damage inducible transcript 3 Homo sapiens 84-88 31746254-0 2020 Cereblon (CRBN) gene polymorphisms predict clinical response and progression-free survival in relapsed/refractory multiple myeloma patients treated with lenalidomide: a pharmacogenetic study from the IMMEnSE consortium. Lenalidomide 153-165 cereblon Homo sapiens 0-8 32251398-3 2020 TP53 mutations are detected in ~20% of LEN-resistant patients3. Lenalidomide 39-42 tumor protein p53 Homo sapiens 0-4 32251398-8 2020 Megakaryocytic gene-promoter analyses suggested that LEN-induced degradation of IKZF1 enables a RUNX1-GATA2 complex to drive megakaryocytic differentiation. Lenalidomide 53-56 IKAROS family zinc finger 1 Homo sapiens 80-85 31804723-5 2020 To test whether LEN-induced mitochondrial priming predicted clinical response to LEN-MEC therapy, we performed DBP on patient myeloblasts. Lenalidomide 16-19 C-C motif chemokine ligand 28 Homo sapiens 85-88 31289205-10 2020 Three kinases inhibitors (CHK1i, MELKi and PBKi) overcome resistance to Lenalidomide, while CHK1, PBK and DBF4 inhibitors re-sensitized Melphalan resistant cell line to this conventional therapeutic agent. Lenalidomide 72-84 checkpoint kinase 1 Homo sapiens 26-30 31289205-10 2020 Three kinases inhibitors (CHK1i, MELKi and PBKi) overcome resistance to Lenalidomide, while CHK1, PBK and DBF4 inhibitors re-sensitized Melphalan resistant cell line to this conventional therapeutic agent. Lenalidomide 72-84 PDZ binding kinase Homo sapiens 43-46 31746254-0 2020 Cereblon (CRBN) gene polymorphisms predict clinical response and progression-free survival in relapsed/refractory multiple myeloma patients treated with lenalidomide: a pharmacogenetic study from the IMMEnSE consortium. Lenalidomide 153-165 cereblon Homo sapiens 10-14 31746254-2 2020 The objective of this study was to verify whether germline single nucleotide polymorphisms (SNPs) in the CRBN gene may influence response to lenalidomide in multiple myeloma (MM). Lenalidomide 141-153 cereblon Homo sapiens 105-109 31746254-3 2020 Fourteen tagging SNPs covering the genetic variability in the CRBN gene region were genotyped in 167 Polish patients with refractory/relapsed MM treated with lenalidomide-based regimens. Lenalidomide 158-170 cereblon Homo sapiens 62-66 31746254-6 2020 It is suggested that selected germline CRBN allelic variants (rs1714327G > C and rs1705814T > C) affect lenalidomide efficacy in patients with relapsed/refractory MM. Lenalidomide 104-116 cereblon Homo sapiens 39-43 31963193-13 2020 IMiDs decreased TLR4-induced mediator release; this effect was stronger for pomalidomide than for lenalidomide and especially thalidomide. Lenalidomide 98-110 toll like receptor 4 Homo sapiens 16-20 31400013-0 2020 Successful lenalidomide treatment in high risk myelodysplastic syndrome with germline DDX41 mutation. Lenalidomide 11-23 DEAD-box helicase 41 Homo sapiens 86-91 31672767-10 2020 CONCLUSIONS: This study showed that blockade of the CXCR4-CXCL12 axis by ulocuplumab is safe with acceptable AEs and leads to a high response rate in combination with lenalidomide and dexamethasone in patients with relapsed/refractory myeloma, making CXCR4 inhibitors a promising class of antimyeloma drugs that should be further explored in clinical trials. Lenalidomide 167-179 C-X-C motif chemokine receptor 4 Homo sapiens 52-57 32759572-8 2020 Administration of lenalidomide prevented the LPS-triggered activation of MM cells by targeting CD180. Lenalidomide 18-30 CD180 molecule Homo sapiens 95-100 31672767-10 2020 CONCLUSIONS: This study showed that blockade of the CXCR4-CXCL12 axis by ulocuplumab is safe with acceptable AEs and leads to a high response rate in combination with lenalidomide and dexamethasone in patients with relapsed/refractory myeloma, making CXCR4 inhibitors a promising class of antimyeloma drugs that should be further explored in clinical trials. Lenalidomide 167-179 C-X-C motif chemokine ligand 12 Homo sapiens 58-64 32984863-2 2020 Lenalidomide interacts with cereblon (CRBN), a component of the CRL4CRBN E3 ubiquitin ligase complex, leading to ubiquitination and subsequent degradation of substrates, such as transcription factor Ikaros (Ikaros family zinc finger 1, IKZF1). Lenalidomide 0-12 cereblon Homo sapiens 38-42 32984863-2 2020 Lenalidomide interacts with cereblon (CRBN), a component of the CRL4CRBN E3 ubiquitin ligase complex, leading to ubiquitination and subsequent degradation of substrates, such as transcription factor Ikaros (Ikaros family zinc finger 1, IKZF1). Lenalidomide 0-12 IKAROS family zinc finger 1 Homo sapiens 199-205 32984863-2 2020 Lenalidomide interacts with cereblon (CRBN), a component of the CRL4CRBN E3 ubiquitin ligase complex, leading to ubiquitination and subsequent degradation of substrates, such as transcription factor Ikaros (Ikaros family zinc finger 1, IKZF1). Lenalidomide 0-12 IKAROS family zinc finger 1 Homo sapiens 207-234 32984863-2 2020 Lenalidomide interacts with cereblon (CRBN), a component of the CRL4CRBN E3 ubiquitin ligase complex, leading to ubiquitination and subsequent degradation of substrates, such as transcription factor Ikaros (Ikaros family zinc finger 1, IKZF1). Lenalidomide 0-12 IKAROS family zinc finger 1 Homo sapiens 236-241 32984863-5 2020 Depletion of G protein-coupled receptor 68 (GPR68) or an endogenous calcineurin (CaN) inhibitor, regulator of calcineurin 1 (RCAN1), reversed the inhibitory effect of lenalidomide on MDSL cells, an MDS cell line. Lenalidomide 167-179 G protein-coupled receptor 68 Homo sapiens 13-42 32984863-5 2020 Depletion of G protein-coupled receptor 68 (GPR68) or an endogenous calcineurin (CaN) inhibitor, regulator of calcineurin 1 (RCAN1), reversed the inhibitory effect of lenalidomide on MDSL cells, an MDS cell line. Lenalidomide 167-179 G protein-coupled receptor 68 Homo sapiens 44-49 32984863-5 2020 Depletion of G protein-coupled receptor 68 (GPR68) or an endogenous calcineurin (CaN) inhibitor, regulator of calcineurin 1 (RCAN1), reversed the inhibitory effect of lenalidomide on MDSL cells, an MDS cell line. Lenalidomide 167-179 regulator of calcineurin 1 Homo sapiens 97-123 32984863-5 2020 Depletion of G protein-coupled receptor 68 (GPR68) or an endogenous calcineurin (CaN) inhibitor, regulator of calcineurin 1 (RCAN1), reversed the inhibitory effect of lenalidomide on MDSL cells, an MDS cell line. Lenalidomide 167-179 regulator of calcineurin 1 Homo sapiens 125-130 32984863-6 2020 Intriguingly, both GPR68 and RCAN1 expression levels were upregulated in MDSL cells after treatment with lenalidomide that was dependent on diminishment of IKZF1, indicating that IKZF1 functioned as a transcription repressor for GPR68 and RCAN1. Lenalidomide 105-117 G protein-coupled receptor 68 Homo sapiens 19-24 32984863-6 2020 Intriguingly, both GPR68 and RCAN1 expression levels were upregulated in MDSL cells after treatment with lenalidomide that was dependent on diminishment of IKZF1, indicating that IKZF1 functioned as a transcription repressor for GPR68 and RCAN1. Lenalidomide 105-117 regulator of calcineurin 1 Homo sapiens 29-34 32984863-6 2020 Intriguingly, both GPR68 and RCAN1 expression levels were upregulated in MDSL cells after treatment with lenalidomide that was dependent on diminishment of IKZF1, indicating that IKZF1 functioned as a transcription repressor for GPR68 and RCAN1. Lenalidomide 105-117 IKAROS family zinc finger 1 Homo sapiens 156-161 32984863-6 2020 Intriguingly, both GPR68 and RCAN1 expression levels were upregulated in MDSL cells after treatment with lenalidomide that was dependent on diminishment of IKZF1, indicating that IKZF1 functioned as a transcription repressor for GPR68 and RCAN1. Lenalidomide 105-117 IKAROS family zinc finger 1 Homo sapiens 179-184 32984863-6 2020 Intriguingly, both GPR68 and RCAN1 expression levels were upregulated in MDSL cells after treatment with lenalidomide that was dependent on diminishment of IKZF1, indicating that IKZF1 functioned as a transcription repressor for GPR68 and RCAN1. Lenalidomide 105-117 G protein-coupled receptor 68 Homo sapiens 229-234 32984863-6 2020 Intriguingly, both GPR68 and RCAN1 expression levels were upregulated in MDSL cells after treatment with lenalidomide that was dependent on diminishment of IKZF1, indicating that IKZF1 functioned as a transcription repressor for GPR68 and RCAN1. Lenalidomide 105-117 regulator of calcineurin 1 Homo sapiens 239-244 32984863-10 2020 Our studies suggest that lenalidomide mediates degradation of IKZF1, leading to derepression of GPR68 and RCAN1 that activates the Ca2+/calpain pro- apoptotic pathway and inhibits the CaN pro-survival pathway, respectively. Lenalidomide 25-37 IKAROS family zinc finger 1 Homo sapiens 62-67 32984863-10 2020 Our studies suggest that lenalidomide mediates degradation of IKZF1, leading to derepression of GPR68 and RCAN1 that activates the Ca2+/calpain pro- apoptotic pathway and inhibits the CaN pro-survival pathway, respectively. Lenalidomide 25-37 G protein-coupled receptor 68 Homo sapiens 96-101 32984863-10 2020 Our studies suggest that lenalidomide mediates degradation of IKZF1, leading to derepression of GPR68 and RCAN1 that activates the Ca2+/calpain pro- apoptotic pathway and inhibits the CaN pro-survival pathway, respectively. Lenalidomide 25-37 regulator of calcineurin 1 Homo sapiens 106-111 31423579-0 2020 JAK1/2 pathway inhibition suppresses M2 polarization and overcomes resistance of myeloma to lenalidomide by reducing TRIB1, MUC1, CD44, CXCL12, and CXCR4 expression. Lenalidomide 92-104 Janus kinase 1 Homo sapiens 0-6 31423579-0 2020 JAK1/2 pathway inhibition suppresses M2 polarization and overcomes resistance of myeloma to lenalidomide by reducing TRIB1, MUC1, CD44, CXCL12, and CXCR4 expression. Lenalidomide 92-104 tribbles pseudokinase 1 Homo sapiens 117-122 31423579-0 2020 JAK1/2 pathway inhibition suppresses M2 polarization and overcomes resistance of myeloma to lenalidomide by reducing TRIB1, MUC1, CD44, CXCL12, and CXCR4 expression. Lenalidomide 92-104 mucin 1, cell surface associated Homo sapiens 124-128 31423579-0 2020 JAK1/2 pathway inhibition suppresses M2 polarization and overcomes resistance of myeloma to lenalidomide by reducing TRIB1, MUC1, CD44, CXCL12, and CXCR4 expression. Lenalidomide 92-104 CD44 molecule (Indian blood group) Homo sapiens 130-134 31423579-0 2020 JAK1/2 pathway inhibition suppresses M2 polarization and overcomes resistance of myeloma to lenalidomide by reducing TRIB1, MUC1, CD44, CXCL12, and CXCR4 expression. Lenalidomide 92-104 C-X-C motif chemokine ligand 12 Homo sapiens 136-142 31423579-0 2020 JAK1/2 pathway inhibition suppresses M2 polarization and overcomes resistance of myeloma to lenalidomide by reducing TRIB1, MUC1, CD44, CXCL12, and CXCR4 expression. Lenalidomide 92-104 C-X-C motif chemokine receptor 4 Homo sapiens 148-153 32967454-2 2020 Immunomodulator lenalidomide may improve CAR T-cell function. Lenalidomide 16-28 nuclear receptor subfamily 1 group I member 3 Homo sapiens 41-44 32967454-3 2020 In this study, the effects of lenalidomide on CAR T-cell functions (cytotoxicity, cytokine secretion, and cell proliferation) were investigated. Lenalidomide 30-42 nuclear receptor subfamily 1 group I member 3 Homo sapiens 46-49 32967454-5 2020 We found that lenalidomide promoted the killing of U251 CD133-OE by CD133-CAR T cells, the cytokine secretion, and the proliferation of CD133-CAR T cells. Lenalidomide 14-26 prominin 1 Homo sapiens 56-61 32967454-5 2020 We found that lenalidomide promoted the killing of U251 CD133-OE by CD133-CAR T cells, the cytokine secretion, and the proliferation of CD133-CAR T cells. Lenalidomide 14-26 prominin 1 Homo sapiens 68-73 32967454-5 2020 We found that lenalidomide promoted the killing of U251 CD133-OE by CD133-CAR T cells, the cytokine secretion, and the proliferation of CD133-CAR T cells. Lenalidomide 14-26 nuclear receptor subfamily 1 group I member 3 Homo sapiens 74-77 32967454-5 2020 We found that lenalidomide promoted the killing of U251 CD133-OE by CD133-CAR T cells, the cytokine secretion, and the proliferation of CD133-CAR T cells. Lenalidomide 14-26 prominin 1 Homo sapiens 68-73 32967454-5 2020 We found that lenalidomide promoted the killing of U251 CD133-OE by CD133-CAR T cells, the cytokine secretion, and the proliferation of CD133-CAR T cells. Lenalidomide 14-26 nuclear receptor subfamily 1 group I member 3 Homo sapiens 142-145 32967454-6 2020 Lenalidomide also enhanced the cytotoxicity against MDA-MB-453 and the cytokine secretion of HER2-CAR T cells but did not affect their proliferation significantly. Lenalidomide 0-12 erb-b2 receptor tyrosine kinase 2 Homo sapiens 93-97 32967454-6 2020 Lenalidomide also enhanced the cytotoxicity against MDA-MB-453 and the cytokine secretion of HER2-CAR T cells but did not affect their proliferation significantly. Lenalidomide 0-12 nuclear receptor subfamily 1 group I member 3 Homo sapiens 98-101 32967454-7 2020 Furthermore, lenalidomide may regulate the function of CAR T cells by inducing the degradation of transcription factors Ikaros and Aiolos. Lenalidomide 13-25 nuclear receptor subfamily 1 group I member 3 Homo sapiens 55-58 31358854-7 2020 Pharmacological targeting of Ikaros by lenalidomide and its analog pomalidomide downregulated SLAMF7 expression and ameliorated the response of MM cells to sSLAMF7. Lenalidomide 39-51 IKAROS family zinc finger 1 Homo sapiens 29-35 31358854-7 2020 Pharmacological targeting of Ikaros by lenalidomide and its analog pomalidomide downregulated SLAMF7 expression and ameliorated the response of MM cells to sSLAMF7. Lenalidomide 39-51 SLAM family member 7 Homo sapiens 94-100 31287161-3 2019 Activated B-cell-like DLBCL is characterized by NF-kappaB activation and chronic B-cell receptor signaling and may be targeted with lenalidomide or ibrutinib in the relapsed setting. Lenalidomide 132-144 nuclear factor kappa B subunit 1 Homo sapiens 48-57 31074316-4 2019 The MTD was DL3: lenalidomide 5 mg daily D8-21 of C1, then 10 mg D1-21 of C2-6. Lenalidomide 17-29 metallothionein 1E Homo sapiens 4-7 31395689-2 2019 Lenalidomide, an immunomodulatory drug, potentiates T cell functionality, drives antimyeloma activity, and alters the suppressive microenvironment; these properties may effectively combine with anti-BCMA CAR T cells to enhance function. Lenalidomide 0-12 TNF receptor superfamily member 17 Homo sapiens 199-203 31395689-3 2019 Using an anti-BCMA CAR T, we demonstrated that lenalidomide enhances CAR T cell function in a concentration-dependent manner. Lenalidomide 47-59 TNF receptor superfamily member 17 Homo sapiens 14-18 31395689-4 2019 Lenalidomide increased CAR T effector cytokine production, particularly under low CAR stimulation or in the presence of inhibitory ligand programmed cell death 1 ligand 1. Lenalidomide 0-12 CD274 molecule Homo sapiens 138-170 31395689-8 2019 Finally, study of lenalidomide and anti-BCMA CAR T cells in a murine, disseminated, multiple myeloma model indicated that lenalidomide increased CAR T cell counts in blood and significantly prolonged animal survival. Lenalidomide 122-134 tumor necrosis factor receptor superfamily, member 17 Mus musculus 40-44 31413179-0 2019 Lenalidomide Augments the Antitumor Activities of Eps8 Peptide-Specific Cytotoxic T Lymphocytes against Multiple Myeloma. Lenalidomide 0-12 epidermal growth factor receptor pathway substrate 8 Homo sapiens 50-54 31413179-8 2019 Importantly, we revealed that lenalidomide effectively stimulated the antitumor activity of the Eps8cocktail-CTLs, with increasing expression trends for T-cell markers (CD28, CD40L, 41BB, and OX40). Lenalidomide 30-42 epidermal growth factor receptor pathway substrate 8 Homo sapiens 96-100 31413179-8 2019 Importantly, we revealed that lenalidomide effectively stimulated the antitumor activity of the Eps8cocktail-CTLs, with increasing expression trends for T-cell markers (CD28, CD40L, 41BB, and OX40). Lenalidomide 30-42 CD40 ligand Homo sapiens 175-180 31413179-8 2019 Importantly, we revealed that lenalidomide effectively stimulated the antitumor activity of the Eps8cocktail-CTLs, with increasing expression trends for T-cell markers (CD28, CD40L, 41BB, and OX40). Lenalidomide 30-42 TNF receptor superfamily member 4 Homo sapiens 192-196 31413179-9 2019 Compared with unstimulated CTLs and Eps8cocktail-CTLs, lenalidomide-treated Eps8cocktail-CTLs showed superior anti-multiple myeloma activity in humanized multiple myeloma models, including delaying tumor burden increases due to enhanced immune function. Lenalidomide 55-67 epidermal growth factor receptor pathway substrate 8 Homo sapiens 36-40 31413179-9 2019 Compared with unstimulated CTLs and Eps8cocktail-CTLs, lenalidomide-treated Eps8cocktail-CTLs showed superior anti-multiple myeloma activity in humanized multiple myeloma models, including delaying tumor burden increases due to enhanced immune function. Lenalidomide 55-67 epidermal growth factor receptor pathway substrate 8 Homo sapiens 76-80 31493176-3 2019 Lenalidomide is a weak substrate of P-glycoprotein (P-gp), though it is unclear whether P-gp is solely responsible for lenalidomide transport. Lenalidomide 0-12 phosphoglycolate phosphatase Mus musculus 36-50 31493176-3 2019 Lenalidomide is a weak substrate of P-glycoprotein (P-gp), though it is unclear whether P-gp is solely responsible for lenalidomide transport. Lenalidomide 0-12 phosphoglycolate phosphatase Mus musculus 52-56 31493176-12 2019 CONCLUSION: As P-gp transport at the blood-brain barrier did not explain the observed brain concentrations alone, there may be other transporters involved in lenalidomide disposition. Lenalidomide 158-170 phosphoglycolate phosphatase Mus musculus 15-19 30760870-2 2019 The immunomodulatory imide drug (IMiD) lenalidomide promotes myeloma cell death via Cereblon (CRBN)-dependent ubiquitylation and proteasome-dependent degradation of IKZF1 and IKZF3. Lenalidomide 39-51 cereblon Homo sapiens 94-98 31151137-0 2019 Suppression of Aiolos and Ikaros expression by lenalidomide reduces human ILC3-ILC1/NK cell transdifferentiation. Lenalidomide 47-59 IKAROS family zinc finger 3 Homo sapiens 15-21 31151137-0 2019 Suppression of Aiolos and Ikaros expression by lenalidomide reduces human ILC3-ILC1/NK cell transdifferentiation. Lenalidomide 47-59 IKAROS family zinc finger 1 Homo sapiens 26-32 31151137-6 2019 Selective degradation of Aiolos and Ikaros by lenalidomide suppressed ILC1 and NK cell differentiation and expression of ILC1 and NK cell-related transcripts (LEF1, PRF1, GRZB, CD244, NCR3, and IRF8). Lenalidomide 46-58 IKAROS family zinc finger 3 Homo sapiens 25-31 31151137-6 2019 Selective degradation of Aiolos and Ikaros by lenalidomide suppressed ILC1 and NK cell differentiation and expression of ILC1 and NK cell-related transcripts (LEF1, PRF1, GRZB, CD244, NCR3, and IRF8). Lenalidomide 46-58 IKAROS family zinc finger 1 Homo sapiens 36-42 31151137-6 2019 Selective degradation of Aiolos and Ikaros by lenalidomide suppressed ILC1 and NK cell differentiation and expression of ILC1 and NK cell-related transcripts (LEF1, PRF1, GRZB, CD244, NCR3, and IRF8). Lenalidomide 46-58 lymphoid enhancer binding factor 1 Homo sapiens 159-163 31151137-6 2019 Selective degradation of Aiolos and Ikaros by lenalidomide suppressed ILC1 and NK cell differentiation and expression of ILC1 and NK cell-related transcripts (LEF1, PRF1, GRZB, CD244, NCR3, and IRF8). Lenalidomide 46-58 perforin 1 Homo sapiens 165-169 31151137-6 2019 Selective degradation of Aiolos and Ikaros by lenalidomide suppressed ILC1 and NK cell differentiation and expression of ILC1 and NK cell-related transcripts (LEF1, PRF1, GRZB, CD244, NCR3, and IRF8). Lenalidomide 46-58 CD244 molecule Homo sapiens 177-182 31151137-6 2019 Selective degradation of Aiolos and Ikaros by lenalidomide suppressed ILC1 and NK cell differentiation and expression of ILC1 and NK cell-related transcripts (LEF1, PRF1, GRZB, CD244, NCR3, and IRF8). Lenalidomide 46-58 natural cytotoxicity triggering receptor 3 Homo sapiens 184-188 31151137-6 2019 Selective degradation of Aiolos and Ikaros by lenalidomide suppressed ILC1 and NK cell differentiation and expression of ILC1 and NK cell-related transcripts (LEF1, PRF1, GRZB, CD244, NCR3, and IRF8). Lenalidomide 46-58 interferon regulatory factor 8 Homo sapiens 194-198 31371506-4 2019 Genetic disruption of IRE1alpha or XBP1s, or pharmacologic IRE1alpha kinase inhibition, attenuated subcutaneous or orthometastatic growth of MM tumors in mice and augmented efficacy of two established frontline antimyeloma agents, bortezomib and lenalidomide. Lenalidomide 246-258 endoplasmic reticulum (ER) to nucleus signalling 1 Mus musculus 22-31 30721106-0 2019 Detection of hemophagocytic extremely multinucleated giant plasma cells after rituximab/low-dose lenalidomide treatment in CD20+ multiple myeloma. Lenalidomide 97-109 keratin 20 Homo sapiens 123-127 31406628-3 2019 Furthermore, we investigated if lenalidomide, a potential sensitizer to anti-CD20 treatment, could prevent an inhibitory effect of ibrutinib. Lenalidomide 32-44 keratin 20 Homo sapiens 77-81 31268156-3 2019 It was shown that the combination of the HDAC6-selective inhibitor, A452, with either of the IMiDs tested (lenalidomide or pomalidomide) led to the synergistic inhibition of cell growth, a decrease in the viability of MM cells and in an increase in the levels of apoptosis. Lenalidomide 107-119 histone deacetylase 6 Homo sapiens 41-46 30760870-2 2019 The immunomodulatory imide drug (IMiD) lenalidomide promotes myeloma cell death via Cereblon (CRBN)-dependent ubiquitylation and proteasome-dependent degradation of IKZF1 and IKZF3. Lenalidomide 39-51 IKAROS family zinc finger 1 Homo sapiens 165-170 30760870-2 2019 The immunomodulatory imide drug (IMiD) lenalidomide promotes myeloma cell death via Cereblon (CRBN)-dependent ubiquitylation and proteasome-dependent degradation of IKZF1 and IKZF3. Lenalidomide 39-51 IKAROS family zinc finger 3 Homo sapiens 175-180 30760870-5 2019 Interaction with RUNX1 and RUNX3 inhibits CRBN-dependent binding, ubiquitylation, and degradation of IKZF1 and IKZF3 upon lenalidomide treatment. Lenalidomide 122-134 RUNX family transcription factor 1 Homo sapiens 17-22 30760870-5 2019 Interaction with RUNX1 and RUNX3 inhibits CRBN-dependent binding, ubiquitylation, and degradation of IKZF1 and IKZF3 upon lenalidomide treatment. Lenalidomide 122-134 RUNX family transcription factor 3 Homo sapiens 27-32 30760870-5 2019 Interaction with RUNX1 and RUNX3 inhibits CRBN-dependent binding, ubiquitylation, and degradation of IKZF1 and IKZF3 upon lenalidomide treatment. Lenalidomide 122-134 cereblon Homo sapiens 42-46 30760870-5 2019 Interaction with RUNX1 and RUNX3 inhibits CRBN-dependent binding, ubiquitylation, and degradation of IKZF1 and IKZF3 upon lenalidomide treatment. Lenalidomide 122-134 IKAROS family zinc finger 1 Homo sapiens 101-106 30760870-5 2019 Interaction with RUNX1 and RUNX3 inhibits CRBN-dependent binding, ubiquitylation, and degradation of IKZF1 and IKZF3 upon lenalidomide treatment. Lenalidomide 122-134 IKAROS family zinc finger 3 Homo sapiens 111-116 31292266-8 2019 The expression of the variant ERFE transcript that was restricted to SF3B1-mutated erythroblasts decreased in lenalidomide-responsive anemic patients, identifying variant ERFE as a specific biomarker of clonal erythropoiesis. Lenalidomide 110-122 erythroferrone Homo sapiens 30-34 30650184-1 2019 There is limited data regarding the efficacy and safety of lenalidomide, adriamycin and dexamethasone (RAD) combination on newly diagnosed multiple myeloma (NDMM) patients. Lenalidomide 59-71 RRAD, Ras related glycolysis inhibitor and calcium channel regulator Homo sapiens 103-106 30650184-8 2019 Interestingly, RAD also increased bone formation markers bone-specific alkaline phosphatase (p = 0.036), procollagen type 1 amino-terminal propeptide (p = 0.028) and osteocalcin (p = 0.026), which has not been described before with lenalidomide-containing regimens in the absence of bortezomib coadministration. Lenalidomide 232-244 RRAD, Ras related glycolysis inhibitor and calcium channel regulator Homo sapiens 15-18 31255504-8 2019 Factors such as exposure to lenalidomide-based pretreatment regimen, peripheral blood WBC count and CD34 count are associated with the adequate CD34 yield. Lenalidomide 28-40 CD34 molecule Homo sapiens 144-148 31292266-8 2019 The expression of the variant ERFE transcript that was restricted to SF3B1-mutated erythroblasts decreased in lenalidomide-responsive anemic patients, identifying variant ERFE as a specific biomarker of clonal erythropoiesis. Lenalidomide 110-122 splicing factor 3b subunit 1 Homo sapiens 69-74 31292266-8 2019 The expression of the variant ERFE transcript that was restricted to SF3B1-mutated erythroblasts decreased in lenalidomide-responsive anemic patients, identifying variant ERFE as a specific biomarker of clonal erythropoiesis. Lenalidomide 110-122 erythroferrone Homo sapiens 171-175 31043801-10 2019 Bortezomib (40 nmol/L, 24 hours) and lenalidomide (3,200 nmol/L, 24 hours) effectively removed CD38+CD138+ cells from peripheral mononuclear cells. Lenalidomide 37-49 CD38 molecule Homo sapiens 95-99 30655375-4 2019 Bone marrow B lineage clonality decreased significantly and hypermutated IGH clones normalized upon lenalidomide treatment, well in line with the proliferative effect on healthy antigen-inexperienced B-cell precursors previously described for this drug. Lenalidomide 100-112 immunoglobulin heavy constant delta Homo sapiens 73-76 30989487-4 2019 Here, we report the two cases in which hematopoietic stem cells were mobilized using plerixafor plus granulocyte-colony stimulating factor after exposure to lenalidomide and pomalidomide. Lenalidomide 157-169 colony stimulating factor 3 Homo sapiens 101-138 30707764-0 2019 Phase 1 study of lenalidomide plus dose-adjusted EPOCH-R in patients with aggressive B-cell lymphomas with deregulated MYC and BCL2. Lenalidomide 17-29 BCL2 apoptosis regulator Homo sapiens 127-131 31273172-9 2019 Patients with an estimated glomerular filtration rate of less than 50mL/min/1.73m2 received a lower cumulative dose of ixazomib and lenalidomide than those with other rates. Lenalidomide 132-144 CD59 molecule (CD59 blood group) Homo sapiens 72-77 31157769-1 2019 The immunomodulatory drugs (IMiDs) thalidomide and its analogs, lenalidomide and pomalidomide, all FDA approved drugs for the treatment of multiple myeloma, induce ubiquitination and degradation of the lymphoid transcription factors Ikaros (IKZF1) and Aiolos (IKZF3) via the cereblon (CRBN) E3 ubiquitin ligase for proteasomal degradation. Lenalidomide 64-76 IKAROS family zinc finger 1 Homo sapiens 241-246 31157769-1 2019 The immunomodulatory drugs (IMiDs) thalidomide and its analogs, lenalidomide and pomalidomide, all FDA approved drugs for the treatment of multiple myeloma, induce ubiquitination and degradation of the lymphoid transcription factors Ikaros (IKZF1) and Aiolos (IKZF3) via the cereblon (CRBN) E3 ubiquitin ligase for proteasomal degradation. Lenalidomide 64-76 IKAROS family zinc finger 3 Homo sapiens 252-258 31157769-1 2019 The immunomodulatory drugs (IMiDs) thalidomide and its analogs, lenalidomide and pomalidomide, all FDA approved drugs for the treatment of multiple myeloma, induce ubiquitination and degradation of the lymphoid transcription factors Ikaros (IKZF1) and Aiolos (IKZF3) via the cereblon (CRBN) E3 ubiquitin ligase for proteasomal degradation. Lenalidomide 64-76 IKAROS family zinc finger 3 Homo sapiens 260-265 31157769-1 2019 The immunomodulatory drugs (IMiDs) thalidomide and its analogs, lenalidomide and pomalidomide, all FDA approved drugs for the treatment of multiple myeloma, induce ubiquitination and degradation of the lymphoid transcription factors Ikaros (IKZF1) and Aiolos (IKZF3) via the cereblon (CRBN) E3 ubiquitin ligase for proteasomal degradation. Lenalidomide 64-76 cereblon Homo sapiens 285-289 31157769-1 2019 The immunomodulatory drugs (IMiDs) thalidomide and its analogs, lenalidomide and pomalidomide, all FDA approved drugs for the treatment of multiple myeloma, induce ubiquitination and degradation of the lymphoid transcription factors Ikaros (IKZF1) and Aiolos (IKZF3) via the cereblon (CRBN) E3 ubiquitin ligase for proteasomal degradation. Lenalidomide 64-76 mitochondrial E3 ubiquitin protein ligase 1 Homo sapiens 291-310 31089832-0 2019 Influence of ABCB1 polymorphisms on the pharmacokinetics and toxicity of lenalidomide in patients with multiple myeloma. Lenalidomide 73-85 ATP binding cassette subfamily B member 1 Homo sapiens 13-18 31089832-3 2019 Here, we investigated the association between ABCB1 polymorphisms and pharmacokinetics of lenalidomide in patients with multiple myeloma (MM) treated with lenalidomide and dexamethasone. Lenalidomide 90-102 ATP binding cassette subfamily B member 1 Homo sapiens 46-51 31089832-3 2019 Here, we investigated the association between ABCB1 polymorphisms and pharmacokinetics of lenalidomide in patients with multiple myeloma (MM) treated with lenalidomide and dexamethasone. Lenalidomide 155-167 ATP binding cassette subfamily B member 1 Homo sapiens 46-51 31089832-10 2019 We show that lenalidomide pharmacokinetics is influenced by the ABCB1 3435C>T polymorphism, which could be useful to individualize dosage design and reduce unexpected toxicity. Lenalidomide 13-25 ATP binding cassette subfamily B member 1 Homo sapiens 64-69 31141702-3 2019 Lenalidomide did not affect the viability or expression of costimulatory molecules, but it potently suppressed the production of the key inflammatory cytokines IL-12 and IL-23, and enhanced the production of the anti-inflammatory cytokine IL-10 by CD1c+ DCs. Lenalidomide 0-12 interleukin 23 subunit alpha Homo sapiens 170-175 31141702-3 2019 Lenalidomide did not affect the viability or expression of costimulatory molecules, but it potently suppressed the production of the key inflammatory cytokines IL-12 and IL-23, and enhanced the production of the anti-inflammatory cytokine IL-10 by CD1c+ DCs. Lenalidomide 0-12 interleukin 10 Homo sapiens 239-244 31141702-3 2019 Lenalidomide did not affect the viability or expression of costimulatory molecules, but it potently suppressed the production of the key inflammatory cytokines IL-12 and IL-23, and enhanced the production of the anti-inflammatory cytokine IL-10 by CD1c+ DCs. Lenalidomide 0-12 CD1c molecule Homo sapiens 248-252 31141702-4 2019 Lenalidomide also suppressed the production of IFN-alpha by CD141+ DCs but not that by plasmacytoid DCs. Lenalidomide 0-12 interferon alpha 1 Homo sapiens 47-56 31141702-4 2019 Lenalidomide also suppressed the production of IFN-alpha by CD141+ DCs but not that by plasmacytoid DCs. Lenalidomide 0-12 thrombomodulin Homo sapiens 60-65 31141702-5 2019 Lenalidomide likely targets pathways downstream of the nuclear translocation of the transcription factors nuclear factor kappaB (NF-kappaB) and IFN regulatory 5 (IRF5) in CD1c+ DCs. Lenalidomide 0-12 nuclear factor kappa B subunit 1 Homo sapiens 129-138 31141702-5 2019 Lenalidomide likely targets pathways downstream of the nuclear translocation of the transcription factors nuclear factor kappaB (NF-kappaB) and IFN regulatory 5 (IRF5) in CD1c+ DCs. Lenalidomide 0-12 interferon regulatory factor 5 Homo sapiens 162-166 31141702-5 2019 Lenalidomide likely targets pathways downstream of the nuclear translocation of the transcription factors nuclear factor kappaB (NF-kappaB) and IFN regulatory 5 (IRF5) in CD1c+ DCs. Lenalidomide 0-12 CD1c molecule Homo sapiens 171-175 31141702-6 2019 Consistent with the direct immunomodulatory effects on DCs, lenalidomide decreased the capacity of CD1c+ DCs to induce differentiation of naive CD4+ T cells into effector cells producing immune activating and myeloma-promoting cytokines. Lenalidomide 60-72 CD1c molecule Homo sapiens 99-103 31141702-6 2019 Consistent with the direct immunomodulatory effects on DCs, lenalidomide decreased the capacity of CD1c+ DCs to induce differentiation of naive CD4+ T cells into effector cells producing immune activating and myeloma-promoting cytokines. Lenalidomide 60-72 CD4 molecule Homo sapiens 144-147 30926391-2 2019 Retrospective series and 1 prospective study suggest that clinically significant responses were predominantly limited to patients with activated B cell (ABC) lymphoma, a finding in agreement with lenalidomide"s potent inhibition of nuclear factor kappaB, the key driver of ABC lymphomas. Lenalidomide 196-208 ATP binding cassette subfamily B member 6 (Langereis blood group) Homo sapiens 153-156 30608891-8 2019 However, the potential synergistic anti-neoplastic effects of lenalidomide in combination with other biological agents, i.e. ibrutinib and venetoclax, especially in the management of p53-mutated cases, still remain an open issue. Lenalidomide 62-74 tumor protein p53 Homo sapiens 183-186 30949411-8 2019 Lenalidomide and low dose of bortezomib had synergistic effect with chidamide, and similarly this effect was attenuated by SDHA siRNA. Lenalidomide 0-12 succinate dehydrogenase complex flavoprotein subunit A Homo sapiens 123-127 30612232-7 2019 Long-term follow-up found that all 12 patients received autologous stem cell transplant, melphalan-based therapy or lenalidomide/thalidomide-based therapy obtained clinical improvement, of which eight experienced decreased levels of VEGF by 50% or back to normal. Lenalidomide 116-128 vascular endothelial growth factor A Homo sapiens 233-237 30423172-3 2019 Agents like the Bruton tyrosine kinase (BTK) inhibitor ibrutinib or immunomodulatory drugs (IMiDs) like pomalidomide and lenalidomide have shown promising high response rates in the salvage setting. Lenalidomide 121-133 Bruton tyrosine kinase Homo sapiens 16-38 30741931-0 2019 Identification of lenalidomide resistance pathways in myeloma and targeted resensitization using cereblon replacement, inhibition of STAT3 or targeting of IRF4. Lenalidomide 18-30 signal transducer and activator of transcription 3 Homo sapiens 133-138 30741931-0 2019 Identification of lenalidomide resistance pathways in myeloma and targeted resensitization using cereblon replacement, inhibition of STAT3 or targeting of IRF4. Lenalidomide 18-30 interferon regulatory factor 4 Homo sapiens 155-159 30741931-7 2019 Inhibition of STAT3 with a selective compound (PB-1-102) re-sensitized XG1LenRes to lenalidomide. Lenalidomide 84-96 signal transducer and activator of transcription 3 Homo sapiens 14-19 30741931-9 2019 This strategy also appeared to be more broadly applicable as SGC-CBP30 could re-sensitize two resistant HMCLs with low but detectable CRBN expression to lenalidomide, suggesting that targeting CBP/E300 is a promising approach to restore IMiD sensitivity in MM with detectable CRBN expression. Lenalidomide 153-165 CREB binding protein Homo sapiens 65-68 30550941-6 2019 The immunomodulatory drug lenalidomide had direct anti-proliferative effect on activated B-cell like DLBCL spheroids and reduced several cytokines and other markers (e.g., CCL2, CCL3, CCL4, CD137 and ANG-1 levels) compared with untreated spheroids. Lenalidomide 26-38 C-C motif chemokine ligand 2 Homo sapiens 172-176 30550941-6 2019 The immunomodulatory drug lenalidomide had direct anti-proliferative effect on activated B-cell like DLBCL spheroids and reduced several cytokines and other markers (e.g., CCL2, CCL3, CCL4, CD137 and ANG-1 levels) compared with untreated spheroids. Lenalidomide 26-38 C-C motif chemokine ligand 3 Homo sapiens 178-182 30550941-6 2019 The immunomodulatory drug lenalidomide had direct anti-proliferative effect on activated B-cell like DLBCL spheroids and reduced several cytokines and other markers (e.g., CCL2, CCL3, CCL4, CD137 and ANG-1 levels) compared with untreated spheroids. Lenalidomide 26-38 C-C motif chemokine ligand 4 Homo sapiens 184-188 30550941-6 2019 The immunomodulatory drug lenalidomide had direct anti-proliferative effect on activated B-cell like DLBCL spheroids and reduced several cytokines and other markers (e.g., CCL2, CCL3, CCL4, CD137 and ANG-1 levels) compared with untreated spheroids. Lenalidomide 26-38 TNF receptor superfamily member 9 Homo sapiens 190-195 30550941-6 2019 The immunomodulatory drug lenalidomide had direct anti-proliferative effect on activated B-cell like DLBCL spheroids and reduced several cytokines and other markers (e.g., CCL2, CCL3, CCL4, CD137 and ANG-1 levels) compared with untreated spheroids. Lenalidomide 26-38 angiopoietin 1 Homo sapiens 200-205 30423172-3 2019 Agents like the Bruton tyrosine kinase (BTK) inhibitor ibrutinib or immunomodulatory drugs (IMiDs) like pomalidomide and lenalidomide have shown promising high response rates in the salvage setting. Lenalidomide 121-133 Bruton tyrosine kinase Homo sapiens 40-43 29788898-9 2019 This E3 ubiquitin ligase in the absence of lenalidomide ubiquitinates CRBN itself and the other components of CRL4CRBN complex. Lenalidomide 43-55 cereblon Homo sapiens 70-74 30598450-12 2019 Combining MDM2 inhibitors with lenalidomide targeting CK1alpha or an MDM4 inhibitor caused synergistic activation of p53, leading to an apoptotic response in MCV-positive MCC cells and MCC-derived xenografts in mice. Lenalidomide 31-43 tumor protein p53 Homo sapiens 117-120 29788898-10 2019 Presence of lenalidomide changes specificity of CRL4CRBN which ubiquitinates two transcription factors, IKZF1 (Ikaros) and IKZF3 (Aiolos), and casein kinase 1alpha (CK1alpha) and marks them for degradation in proteasomes. Lenalidomide 12-24 IKAROS family zinc finger 1 Homo sapiens 104-109 29788898-10 2019 Presence of lenalidomide changes specificity of CRL4CRBN which ubiquitinates two transcription factors, IKZF1 (Ikaros) and IKZF3 (Aiolos), and casein kinase 1alpha (CK1alpha) and marks them for degradation in proteasomes. Lenalidomide 12-24 IKAROS family zinc finger 1 Homo sapiens 111-117 29788898-10 2019 Presence of lenalidomide changes specificity of CRL4CRBN which ubiquitinates two transcription factors, IKZF1 (Ikaros) and IKZF3 (Aiolos), and casein kinase 1alpha (CK1alpha) and marks them for degradation in proteasomes. Lenalidomide 12-24 IKAROS family zinc finger 3 Homo sapiens 123-128 29788898-10 2019 Presence of lenalidomide changes specificity of CRL4CRBN which ubiquitinates two transcription factors, IKZF1 (Ikaros) and IKZF3 (Aiolos), and casein kinase 1alpha (CK1alpha) and marks them for degradation in proteasomes. Lenalidomide 12-24 IKAROS family zinc finger 3 Homo sapiens 130-136 29788898-12 2019 Low CRBN level was connected with insensitivity of MM cells to lenalidomide. Lenalidomide 63-75 cereblon Homo sapiens 4-8 29788898-13 2019 Lenalidomide decreases expression of protein argonaute-2, which binds to cereblon. Lenalidomide 0-12 argonaute RISC catalytic component 2 Homo sapiens 37-56 29788898-15 2019 Lenalidomide decreases also basigin and monocarboxylate transporter 1 in MM cells. Lenalidomide 0-12 basigin (Ok blood group) Homo sapiens 28-35 29788898-15 2019 Lenalidomide decreases also basigin and monocarboxylate transporter 1 in MM cells. Lenalidomide 0-12 solute carrier family 16 member 1 Homo sapiens 40-69 29788898-16 2019 MM cells with low expression of Ikaros, Aiolos and basigin are more sensitive to lenalidomide treatment. Lenalidomide 81-93 IKAROS family zinc finger 1 Homo sapiens 32-38 29788898-16 2019 MM cells with low expression of Ikaros, Aiolos and basigin are more sensitive to lenalidomide treatment. Lenalidomide 81-93 IKAROS family zinc finger 3 Homo sapiens 40-46 29788898-16 2019 MM cells with low expression of Ikaros, Aiolos and basigin are more sensitive to lenalidomide treatment. Lenalidomide 81-93 basigin (Ok blood group) Homo sapiens 51-58 29788898-21 2019 High level of full length CRBN mRNA in mononuclear cells of bone marrow and of peripheral blood seems to be necessary for successful therapy of del(5q) MDS with lenalidomide. Lenalidomide 161-173 cereblon Homo sapiens 26-30 30026574-1 2019 Immunomodulatory drugs (IMiDs) including lenalidomide and pomalidomide bind cereblon (CRBN) and activate the CRL4CRBN ubiquitin ligase to trigger proteasomal degradation of the essential transcription factors IKZF1 and IKZF3 and multiple myeloma (MM) cytotoxicity. Lenalidomide 41-53 cereblon Homo sapiens 86-90 30826127-8 2019 Preclinical and clinical studies have demonstrated that CD38-targeting antibodies have synergistic activity with several other anti-cancer drugs, including various agents with immune stimulating activity, such as lenalidomide and pomalidomide, as well as PD1/PD-L1 inhibitors. Lenalidomide 213-225 CD38 molecule Homo sapiens 56-60 29718735-3 2019 Using standard immunohistochemistry in formalin-fixed paraffin embedded (FFPE) bone marrow samples of 23 patients treated with a lenalidomide-containing regimen, we found that the malignant plasma cells of all the patients stained positive for CRBN, IKZF1, and IKZF3, regardless of sensitivity to IMiDs. Lenalidomide 129-141 cereblon Homo sapiens 244-248 30608448-0 2019 Novel EZH2 mutation in a patient with secondary B-cell acute lymphocytic leukemia after deletion 5q myelodysplastic syndrome treated with lenalidomide: A case report. Lenalidomide 138-150 enhancer of zeste 2 polycomb repressive complex 2 subunit Homo sapiens 6-10 30608448-4 2019 He was administered lenalidomide for 27 months, then developed acute B-cell lymphocytic leukemia and acquired a previously unreported mutation in the gene enhancer of zeste homolog 2 (EZH2). Lenalidomide 20-32 enhancer of zeste 2 polycomb repressive complex 2 subunit Homo sapiens 155-182 30608448-4 2019 He was administered lenalidomide for 27 months, then developed acute B-cell lymphocytic leukemia and acquired a previously unreported mutation in the gene enhancer of zeste homolog 2 (EZH2). Lenalidomide 20-32 enhancer of zeste 2 polycomb repressive complex 2 subunit Homo sapiens 184-188 31068561-2 2019 All of them had markedly elevated serum vascular endothelial growth factor (VEGF) levels treated with lenalidomide and dexamethasone for severe peripheral neuropathy, which normalized serum VEGF levels and improved peripheral neuropathy. Lenalidomide 102-114 vascular endothelial growth factor A Homo sapiens 40-74 31068561-2 2019 All of them had markedly elevated serum vascular endothelial growth factor (VEGF) levels treated with lenalidomide and dexamethasone for severe peripheral neuropathy, which normalized serum VEGF levels and improved peripheral neuropathy. Lenalidomide 102-114 vascular endothelial growth factor A Homo sapiens 76-80 31068561-2 2019 All of them had markedly elevated serum vascular endothelial growth factor (VEGF) levels treated with lenalidomide and dexamethasone for severe peripheral neuropathy, which normalized serum VEGF levels and improved peripheral neuropathy. Lenalidomide 102-114 vascular endothelial growth factor A Homo sapiens 190-194 30026574-1 2019 Immunomodulatory drugs (IMiDs) including lenalidomide and pomalidomide bind cereblon (CRBN) and activate the CRL4CRBN ubiquitin ligase to trigger proteasomal degradation of the essential transcription factors IKZF1 and IKZF3 and multiple myeloma (MM) cytotoxicity. Lenalidomide 41-53 interleukin 17 receptor B Homo sapiens 109-113 30026574-1 2019 Immunomodulatory drugs (IMiDs) including lenalidomide and pomalidomide bind cereblon (CRBN) and activate the CRL4CRBN ubiquitin ligase to trigger proteasomal degradation of the essential transcription factors IKZF1 and IKZF3 and multiple myeloma (MM) cytotoxicity. Lenalidomide 41-53 IKAROS family zinc finger 1 Homo sapiens 209-214 30026574-1 2019 Immunomodulatory drugs (IMiDs) including lenalidomide and pomalidomide bind cereblon (CRBN) and activate the CRL4CRBN ubiquitin ligase to trigger proteasomal degradation of the essential transcription factors IKZF1 and IKZF3 and multiple myeloma (MM) cytotoxicity. Lenalidomide 41-53 IKAROS family zinc finger 3 Homo sapiens 219-224 30619281-5 2018 The immunomodulatory drug lenalidomide may regulate the expression of ILT2 and its ligands in CLL since it significantly increased the expression of ILT2 and partially reestablished the expression of its ligands on leukemic cells. Lenalidomide 26-38 leukocyte immunoglobulin like receptor B1 Homo sapiens 70-74 30619281-5 2018 The immunomodulatory drug lenalidomide may regulate the expression of ILT2 and its ligands in CLL since it significantly increased the expression of ILT2 and partially reestablished the expression of its ligands on leukemic cells. Lenalidomide 26-38 leukocyte immunoglobulin like receptor B1 Homo sapiens 149-153 30619281-6 2018 Furthermore, lenalidomide significantly increased the activation and proliferation of NK cells, which was strongly enhanced by ILT2 blockade. Lenalidomide 13-25 leukocyte immunoglobulin like receptor B1 Homo sapiens 127-131 30128502-15 2018 Of note, PVX-410 was immunogenic as monotherapy (10 of 11 patients) and in combination with lenalidomide (9 of 9 patients), as demonstrated by an increase in percentage of tetramer-positive cells and IFN-gamma cells in the CD3+CD8+ cell population. Lenalidomide 92-104 interferon gamma Homo sapiens 200-209 30312729-0 2019 Pan-PIM kinase inhibitors enhance Lenalidomide"s anti-myeloma activity via cereblon-IKZF1/3 cascade. Lenalidomide 34-46 IKAROS family zinc finger 1 Mus musculus 84-91 30312729-4 2019 Combining a pan-PIM kinase inhibitor with lenalidomide led to more effective degradation of IKZF1 and IKZF3 in multiple myeloma cell lines as well as xenografts of myeloma tumors. Lenalidomide 42-54 IKAROS family zinc finger 1 Mus musculus 92-97 30312729-4 2019 Combining a pan-PIM kinase inhibitor with lenalidomide led to more effective degradation of IKZF1 and IKZF3 in multiple myeloma cell lines as well as xenografts of myeloma tumors. Lenalidomide 42-54 IKAROS family zinc finger 3 Mus musculus 102-107 30155589-1 2018 Cancer-related fatigue (CRF) is one of the adverse events in multiple myeloma (MM) patients treated with cytotoxic agents, proteasome inhibitors (PIs), and immunomodulatory drugs (IMiDs) such as bortezomib, lenalidomide, and thalidomide. Lenalidomide 207-219 complement C1q like 1 Homo sapiens 0-22 30064974-6 2018 We use the Crbn I391V model to demonstrate that the in vivo therapeutic activity of lenalidomide in del(5q) myelodysplastic syndrome can be explained by heterozygous expression of Ck1alpha in del(5q) cells. Lenalidomide 84-96 cereblon Mus musculus 11-15 30455698-3 2018 Elotuzumab is a humanized IgG1 monoclonal antibody targeting SLAMF7, which is highly expressed on myeloma cells, and the antibody is approved for the treatment of relapsed and/or refractory (RR) MM in combination with lenalidomide and dexamethasone. Lenalidomide 218-230 SLAM family member 7 Homo sapiens 61-67 30385546-1 2018 The small molecules thalidomide, lenalidomide, and pomalidomide induce the ubiquitination and proteasomal degradation of the transcription factors Ikaros (IKZF1) and Aiolos (IKZF3) by recruiting a Cys2-His2 (C2H2) zinc finger domain to Cereblon (CRBN), the substrate receptor of the CRL4CRBN E3 ubiquitin ligase. Lenalidomide 33-45 IKAROS family zinc finger 1 Homo sapiens 155-160 30385546-1 2018 The small molecules thalidomide, lenalidomide, and pomalidomide induce the ubiquitination and proteasomal degradation of the transcription factors Ikaros (IKZF1) and Aiolos (IKZF3) by recruiting a Cys2-His2 (C2H2) zinc finger domain to Cereblon (CRBN), the substrate receptor of the CRL4CRBN E3 ubiquitin ligase. Lenalidomide 33-45 IKAROS family zinc finger 3 Homo sapiens 174-179 30385546-1 2018 The small molecules thalidomide, lenalidomide, and pomalidomide induce the ubiquitination and proteasomal degradation of the transcription factors Ikaros (IKZF1) and Aiolos (IKZF3) by recruiting a Cys2-His2 (C2H2) zinc finger domain to Cereblon (CRBN), the substrate receptor of the CRL4CRBN E3 ubiquitin ligase. Lenalidomide 33-45 cereblon Homo sapiens 236-244 30385546-1 2018 The small molecules thalidomide, lenalidomide, and pomalidomide induce the ubiquitination and proteasomal degradation of the transcription factors Ikaros (IKZF1) and Aiolos (IKZF3) by recruiting a Cys2-His2 (C2H2) zinc finger domain to Cereblon (CRBN), the substrate receptor of the CRL4CRBN E3 ubiquitin ligase. Lenalidomide 33-45 cereblon Homo sapiens 246-250 29938309-0 2018 Low frequency of CD3+CD4+CD161+ T cells correlates with the occurrence of infections in refractory/relapsed multiple myeloma patients receiving lenalidomide plus low-dose dexamethasone treatment. Lenalidomide 144-156 CD4 molecule Homo sapiens 21-24 30349293-8 2018 For this purpose, several drugs may have a role: specific COX-2 inhibitors such as celecoxib or other anti-inflammatory drugs such as lenalidomide may further inhibit lipopolysaccharide-mediated induction of COX-2. Lenalidomide 134-146 mitochondrially encoded cytochrome c oxidase II Homo sapiens 58-63 30349293-8 2018 For this purpose, several drugs may have a role: specific COX-2 inhibitors such as celecoxib or other anti-inflammatory drugs such as lenalidomide may further inhibit lipopolysaccharide-mediated induction of COX-2. Lenalidomide 134-146 mitochondrially encoded cytochrome c oxidase II Homo sapiens 208-213 30285865-7 2018 We also demonstrate a synergistic effect of EPZ-6438 and lenalidomide, a conventional drug used for MM treatment, activating B cell transcription factors and tumor suppressor gene expression in concert with MYC repression. Lenalidomide 57-69 MYC proto-oncogene, bHLH transcription factor Homo sapiens 207-210 30118587-1 2018 The immunomodulatory drugs (IMiDs) thalidomide, lenalidomide, and pomalidomide, all approved for the treatment of multiple myeloma, induce targeted ubiquitination and degradation of Ikaros (IKZF1) and Aiolos (IKZF3) via the cereblon (CRBN) E3 ubiquitin ligase. Lenalidomide 48-60 IKAROS family zinc finger 1 Homo sapiens 182-188 30118587-1 2018 The immunomodulatory drugs (IMiDs) thalidomide, lenalidomide, and pomalidomide, all approved for the treatment of multiple myeloma, induce targeted ubiquitination and degradation of Ikaros (IKZF1) and Aiolos (IKZF3) via the cereblon (CRBN) E3 ubiquitin ligase. Lenalidomide 48-60 cereblon Homo sapiens 234-238 30338042-0 2018 Lenalidomide combined with low-dose cyclophosphamide and prednisone modulates Ikaros and Aiolos in lymphocytes, resulting in immunostimulatory effects in lenalidomide-refractory multiple myeloma patients. Lenalidomide 0-12 IKAROS family zinc finger 3 Homo sapiens 89-95 29870139-5 2018 Binding studies with the IMiD target protein cereblon (CRBN) reveals a narrow structure-activity relationship with only a few compounds showing sub-micromolar binding affinity in the range of pomalidomide and lenalidomide. Lenalidomide 209-221 cereblon Homo sapiens 55-59 30154887-7 2018 Results: Mobilization increased blood CD34+ frequency in lenalidomide-treated subjects and controls similarly (0.25% (95% confidence interval (CI): 0.03-1.39% vs. 0.32%, 0.04-1.47%), p = 0.472). Lenalidomide 57-69 CD34 molecule Homo sapiens 38-42 30154887-10 2018 In contrast, mean CXCR4 intensity on bone marrow cells in lenalidomide-treated subjects increased from 55 +-43 to 89 +-40 (p = 0.017, comparing the deviation between two groups). Lenalidomide 58-70 C-X-C motif chemokine receptor 4 Homo sapiens 18-23 30154887-11 2018 Median numbers of CD34+ cells collected in lenalidomide-treated subjects and controls were 2.3 x 106/kg (0.6-6.8 x 106/kg) and 2.8106/kg (1.0-8.9 x 106/kg; p = 0.521). Lenalidomide 43-55 CD34 molecule Homo sapiens 18-22 30154887-12 2018 Conclusions: Brief lenalidomide treatment for POEMS did not reduce numbers of CD34+ blood cells collected but increased CXCR4 expression on bone marrow CD34+ cells. Lenalidomide 19-31 C-X-C motif chemokine receptor 4 Homo sapiens 120-125 30154887-12 2018 Conclusions: Brief lenalidomide treatment for POEMS did not reduce numbers of CD34+ blood cells collected but increased CXCR4 expression on bone marrow CD34+ cells. Lenalidomide 19-31 CD34 molecule Homo sapiens 152-156 30097406-2 2018 Lenalidomide resistance, including primary resistance, occurs by clonal evolution, which is frequently attributable to the presence of somatic mutations in the DNA-binding domain of the TP53 gene. Lenalidomide 0-12 tumor protein p53 Homo sapiens 186-190 30104242-0 2018 Activation of Th1 Immunity within the Tumor Microenvironment Is Associated with Clinical Response to Lenalidomide in Chronic Lymphocytic Leukemia. Lenalidomide 101-113 negative elongation factor complex member C/D Homo sapiens 14-17 30104242-6 2018 Within days of starting lenalidomide, T cells increased in the tumor microenvironment and showed Th1-type polarization. Lenalidomide 24-36 negative elongation factor complex member C/D Homo sapiens 97-100 30104242-7 2018 Gene expression profiling of pretreatment and on-treatment lymph node biopsy specimens revealed upregulation of IFN-gamma and many of its target genes in response to lenalidomide. Lenalidomide 166-178 interferon gamma Homo sapiens 112-121 30104242-11 2018 Taken together, our data demonstrate that lenalidomide induced Th1 immunity in the lymph node that is associated with clinical response. Lenalidomide 42-54 negative elongation factor complex member C/D Homo sapiens 63-66 30146162-1 2018 CKIalpha ablation induces p53 activation, and CKIalpha degradation underlies the therapeutic effect of lenalidomide in a pre-leukemia syndrome. Lenalidomide 103-115 casein kinase 1 alpha 1 Homo sapiens 0-8 30146162-1 2018 CKIalpha ablation induces p53 activation, and CKIalpha degradation underlies the therapeutic effect of lenalidomide in a pre-leukemia syndrome. Lenalidomide 103-115 casein kinase 1 alpha 1 Homo sapiens 46-54 30234487-1 2018 The cereblon modulating agents (CMs) including lenalidomide, pomalidomide and CC-220 repurpose the Cul4-RBX1-DDB1-CRBN (CRL4CRBN) E3 ubiquitin ligase complex to induce the degradation of specific neomorphic substrates via polyubiquitination in conjunction with E2 ubiquitin-conjugating enzymes, which have until now remained elusive. Lenalidomide 47-59 ring-box 1 Homo sapiens 104-108 30234487-1 2018 The cereblon modulating agents (CMs) including lenalidomide, pomalidomide and CC-220 repurpose the Cul4-RBX1-DDB1-CRBN (CRL4CRBN) E3 ubiquitin ligase complex to induce the degradation of specific neomorphic substrates via polyubiquitination in conjunction with E2 ubiquitin-conjugating enzymes, which have until now remained elusive. Lenalidomide 47-59 damage specific DNA binding protein 1 Homo sapiens 109-113 30234487-1 2018 The cereblon modulating agents (CMs) including lenalidomide, pomalidomide and CC-220 repurpose the Cul4-RBX1-DDB1-CRBN (CRL4CRBN) E3 ubiquitin ligase complex to induce the degradation of specific neomorphic substrates via polyubiquitination in conjunction with E2 ubiquitin-conjugating enzymes, which have until now remained elusive. Lenalidomide 47-59 cereblon Homo sapiens 114-118 30234487-4 2018 For example, UBE2G1 inactivation significantly attenuated the degradation of myeloma survival factors IKZF1 and IKZF3 induced by lenalidomide and pomalidomide, hence conferring drug resistance. Lenalidomide 129-141 ubiquitin conjugating enzyme E2 G1 Homo sapiens 13-19 30234487-4 2018 For example, UBE2G1 inactivation significantly attenuated the degradation of myeloma survival factors IKZF1 and IKZF3 induced by lenalidomide and pomalidomide, hence conferring drug resistance. Lenalidomide 129-141 IKAROS family zinc finger 1 Homo sapiens 102-107 30234487-4 2018 For example, UBE2G1 inactivation significantly attenuated the degradation of myeloma survival factors IKZF1 and IKZF3 induced by lenalidomide and pomalidomide, hence conferring drug resistance. Lenalidomide 129-141 IKAROS family zinc finger 3 Homo sapiens 112-117 30190454-9 2018 Lenalidomide can be given at full dose 25 mg daily 21/28 in patients with a CrCl > 30, and can be given daily to those with CrCl < 30, even when on dialysis, at doses of at least 15 mg daily. Lenalidomide 0-12 CRCL Homo sapiens 76-80 30190454-9 2018 Lenalidomide can be given at full dose 25 mg daily 21/28 in patients with a CrCl > 30, and can be given daily to those with CrCl < 30, even when on dialysis, at doses of at least 15 mg daily. Lenalidomide 0-12 CRCL Homo sapiens 127-131 29545338-0 2018 Cereblon loss and up-regulation of c-Myc are associated with lenalidomide resistance in multiple myeloma patients. Lenalidomide 61-73 MYC proto-oncogene, bHLH transcription factor Homo sapiens 35-40 30118587-1 2018 The immunomodulatory drugs (IMiDs) thalidomide, lenalidomide, and pomalidomide, all approved for the treatment of multiple myeloma, induce targeted ubiquitination and degradation of Ikaros (IKZF1) and Aiolos (IKZF3) via the cereblon (CRBN) E3 ubiquitin ligase. Lenalidomide 48-60 IKAROS family zinc finger 1 Homo sapiens 190-195 30118587-1 2018 The immunomodulatory drugs (IMiDs) thalidomide, lenalidomide, and pomalidomide, all approved for the treatment of multiple myeloma, induce targeted ubiquitination and degradation of Ikaros (IKZF1) and Aiolos (IKZF3) via the cereblon (CRBN) E3 ubiquitin ligase. Lenalidomide 48-60 IKAROS family zinc finger 3 Homo sapiens 201-207 30118587-1 2018 The immunomodulatory drugs (IMiDs) thalidomide, lenalidomide, and pomalidomide, all approved for the treatment of multiple myeloma, induce targeted ubiquitination and degradation of Ikaros (IKZF1) and Aiolos (IKZF3) via the cereblon (CRBN) E3 ubiquitin ligase. Lenalidomide 48-60 IKAROS family zinc finger 3 Homo sapiens 209-214 29611764-0 2018 Lenalidomide combined with R-GDP in a patient with refractory CD5-positive diffuse large B-cell lymphoma: A promising response and review. Lenalidomide 0-12 CD5 molecule Homo sapiens 62-65 30288348-5 2018 Results: We found that high IKZF3, but not IKZF1, expression in T-cells correlates with superior overall survival in MMIII patients treated with immunomodulatory drugs (thalidomide, lenalidomide and pomalidomide). Lenalidomide 182-194 IKAROS family zinc finger 3 Homo sapiens 28-33 30111438-3 2018 The combination of lenalidomide and CRBN recruited a new substrate that binds to the CRBN-CRL4 complex, leading to increased ubiquitination and proteasome-dependent degradation, thus resulting in anti-myeloma activity. Lenalidomide 19-31 cereblon Homo sapiens 85-89 30111438-3 2018 The combination of lenalidomide and CRBN recruited a new substrate that binds to the CRBN-CRL4 complex, leading to increased ubiquitination and proteasome-dependent degradation, thus resulting in anti-myeloma activity. Lenalidomide 19-31 interleukin 17 receptor B Homo sapiens 90-94 30111438-5 2018 The CRBN-dependent degradation of IKZF1 and IKZF3 after lenalidomide treatment is also the result of H2O2-mediated oxidative stress. Lenalidomide 56-68 cereblon Homo sapiens 4-8 30111438-5 2018 The CRBN-dependent degradation of IKZF1 and IKZF3 after lenalidomide treatment is also the result of H2O2-mediated oxidative stress. Lenalidomide 56-68 IKAROS family zinc finger 1 Homo sapiens 34-39 30111438-5 2018 The CRBN-dependent degradation of IKZF1 and IKZF3 after lenalidomide treatment is also the result of H2O2-mediated oxidative stress. Lenalidomide 56-68 IKAROS family zinc finger 3 Homo sapiens 44-49 30111438-6 2018 In addition to ubiquitination, lenalidomide also mediates ubiquitin-independent pathways that prevent CRBN from binding to CD147-MCT1 in a competitive manner to regulate its antitumor activity. Lenalidomide 31-43 cereblon Homo sapiens 102-106 30111438-6 2018 In addition to ubiquitination, lenalidomide also mediates ubiquitin-independent pathways that prevent CRBN from binding to CD147-MCT1 in a competitive manner to regulate its antitumor activity. Lenalidomide 31-43 basigin (Ok blood group) Homo sapiens 123-128 30111438-6 2018 In addition to ubiquitination, lenalidomide also mediates ubiquitin-independent pathways that prevent CRBN from binding to CD147-MCT1 in a competitive manner to regulate its antitumor activity. Lenalidomide 31-43 solute carrier family 16 member 1 Homo sapiens 129-133 30111438-7 2018 Lenalidomide can also play a role in multiple myeloma(MM) cells by modulating miRNA levels and CRBN binding to downstream protein AGO2 expression. Lenalidomide 0-12 cereblon Homo sapiens 95-99 30111438-7 2018 Lenalidomide can also play a role in multiple myeloma(MM) cells by modulating miRNA levels and CRBN binding to downstream protein AGO2 expression. Lenalidomide 0-12 argonaute RISC catalytic component 2 Homo sapiens 130-134 30111438-9 2018 This review summarizes the molecular mechanisms of CRBN in lenalidomide against myeloma activity in terms of ubiquitin-dependent and ubiquitin-independent pathways. Lenalidomide 59-71 cereblon Homo sapiens 51-55 30228947-8 2018 When administered as a single agent, lenalidomide specifically mobilizes non-del5q/5q- AML cells, but not healthy CD34+ cells, to peripheral blood (PB) through specific downregulation of CXCR4 in AML blasts. Lenalidomide 37-49 C-X-C motif chemokine receptor 4 Homo sapiens 187-192 30079070-1 2018 Monoclonal antibodies against the cell surface antigen CD38, e.g., isatuximab, daratumumab, or Mor202, have entered the therapeutic armamentarium in multiple myeloma due to single agent overall response rates of 29 vs. 36 vs. 31%, effectivity in combination regimen, e.g., with lenalidomide or bortezomib plus dexamethasone, and tolerable side effects. Lenalidomide 278-290 CD38 molecule Homo sapiens 55-59 29986852-10 2018 Change in CSF interleukin-10 at 1 month correlated with clinical response and response duration to lenalidomide. Lenalidomide 99-111 interleukin 10 Homo sapiens 14-28 29611764-11 2018 The clinical features, pathogenesis, and possible mechanism of action of lenalidomide in CD5+ DLBCL have been described in the literature. Lenalidomide 73-85 CD5 molecule Homo sapiens 89-92 29611764-13 2018 Lenalidomide is expected to induce favorable responses in patients with CD5+ DLBCL. Lenalidomide 0-12 CD5 molecule Homo sapiens 72-75 29707981-3 2018 METHODS: In MM cell lines and primary cells derived by patients affected by MGUS and MM, we investigated sensitivity to bortezomib and lenalidomide in presence of Arg-1 and PMN-MDSC. Lenalidomide 135-147 arginase 1 Homo sapiens 163-168 29707981-9 2018 PMN-MDSC and Arg-1 are reduced in vivo after lenalidomide but not bortezomib treatment. Lenalidomide 45-57 arginase 1 Homo sapiens 13-18 29702148-8 2018 Preclinical and clinical studies have demonstrated that CD38-targeting antibodies have synergistic activity with several other anti-cancer drugs, including various agents with immune stimulating activity, such as lenalidomide and pomalidomide, as well as PD1/PD-L1 inhibitors. Lenalidomide 213-225 CD38 molecule Homo sapiens 56-60 29958267-10 2018 RESULTS: Lenalidomide in combination with dexamethasone treatment provided 1.41 incremental life years and 0.83 incremental quality-adjusted life years in comparison with bortezomib in combination with dexamethasone, with an incremental cost of 11 864 597.86 CLP (19 589.86 US$). Lenalidomide 9-21 calmodulin like 3 Homo sapiens 259-262 29946110-2 2018 We hypothesized that adding lenalidomide to R-CHOP (R2CHOP) may decrease the risk of CNS relapse. Lenalidomide 28-40 DNA damage inducible transcript 3 Homo sapiens 46-50 29746861-0 2018 Lenalidomide regulates osteocytes fate and related osteoclastogenesis via IL-1beta/NF-kappaB/RANKL signaling. Lenalidomide 0-12 interleukin 1 beta Mus musculus 74-82 29746861-0 2018 Lenalidomide regulates osteocytes fate and related osteoclastogenesis via IL-1beta/NF-kappaB/RANKL signaling. Lenalidomide 0-12 tumor necrosis factor (ligand) superfamily, member 11 Mus musculus 93-98 29746861-3 2018 Our data revealed that lenalidomide treatment notably rescued IL-1beta induced loss of osteocyte viability by inhibiting osteocyte apoptosis with decreased osteoclast-related factors, RANKL and Sclerostin, as demonstrated by the restricted osteoclast formation and reduced bone resorption. Lenalidomide 23-35 interleukin 1 beta Mus musculus 62-70 29746861-3 2018 Our data revealed that lenalidomide treatment notably rescued IL-1beta induced loss of osteocyte viability by inhibiting osteocyte apoptosis with decreased osteoclast-related factors, RANKL and Sclerostin, as demonstrated by the restricted osteoclast formation and reduced bone resorption. Lenalidomide 23-35 tumor necrosis factor (ligand) superfamily, member 11 Mus musculus 184-189 29746861-4 2018 Additionally, iTRAQ assay revealed that IL-1beta induced activation of NF-kappaB inhibitor alpha/beta were remarkably downregulated by lenalidomide, showing that lenalidomide impaired NF-kappaB signaling in osteocytes for inhibiting the expression of osteoclast specific genes in osteoclasts, which was further confirmed by KEGG pathway analysis and Western blot. Lenalidomide 135-147 interleukin 1 beta Mus musculus 40-48 29746861-4 2018 Additionally, iTRAQ assay revealed that IL-1beta induced activation of NF-kappaB inhibitor alpha/beta were remarkably downregulated by lenalidomide, showing that lenalidomide impaired NF-kappaB signaling in osteocytes for inhibiting the expression of osteoclast specific genes in osteoclasts, which was further confirmed by KEGG pathway analysis and Western blot. Lenalidomide 135-147 nuclear factor of kappa light polypeptide gene enhancer in B cells inhibitor, alpha Mus musculus 71-96 29746861-4 2018 Additionally, iTRAQ assay revealed that IL-1beta induced activation of NF-kappaB inhibitor alpha/beta were remarkably downregulated by lenalidomide, showing that lenalidomide impaired NF-kappaB signaling in osteocytes for inhibiting the expression of osteoclast specific genes in osteoclasts, which was further confirmed by KEGG pathway analysis and Western blot. Lenalidomide 162-174 interleukin 1 beta Mus musculus 40-48 29746861-4 2018 Additionally, iTRAQ assay revealed that IL-1beta induced activation of NF-kappaB inhibitor alpha/beta were remarkably downregulated by lenalidomide, showing that lenalidomide impaired NF-kappaB signaling in osteocytes for inhibiting the expression of osteoclast specific genes in osteoclasts, which was further confirmed by KEGG pathway analysis and Western blot. Lenalidomide 162-174 nuclear factor of kappa light polypeptide gene enhancer in B cells inhibitor, alpha Mus musculus 71-96 29746861-5 2018 More interestingly, the in vivo analysis of osteocyte apoptosis and osteoclastogenesis in osteoarthritis mice model indicated a role of lenalidomide in the regulation of osteocyte fate and the consequent inhibition of RANKL-induced osteoclastogenesis. Lenalidomide 136-148 tumor necrosis factor (ligand) superfamily, member 11 Mus musculus 218-223 29746861-6 2018 Together, these results suggest that lenalidomide regulates osteocyte fate by attenuating IL-1beta/NF-kappaB signaling, thereby inhibiting RANKL expression for the attenuated osteoclastogenesis both in vitro and vivo, indicating a more efficient remedy among future anti-osteoclastogenesis approaches. Lenalidomide 37-49 interleukin 1 beta Mus musculus 90-98 29746861-6 2018 Together, these results suggest that lenalidomide regulates osteocyte fate by attenuating IL-1beta/NF-kappaB signaling, thereby inhibiting RANKL expression for the attenuated osteoclastogenesis both in vitro and vivo, indicating a more efficient remedy among future anti-osteoclastogenesis approaches. Lenalidomide 37-49 tumor necrosis factor (ligand) superfamily, member 11 Mus musculus 139-144 30701929-4 2018 Lenalidomide have shown activity in the ABC-DLBCL in combination with R-CHOP. Lenalidomide 0-12 ATP binding cassette subfamily B member 6 (Langereis blood group) Homo sapiens 40-43 29967612-8 2018 These findings suggest that the combination of DCs plus lenalidomide and PD-1 blockade synergistically establishes a robust anti-myeloma immunity through a two-way mechanism, which inhibits immunosuppressive cells while activating effector cells with superior polarization of the Th1/Th2 balance in favor of the tumor immune response. Lenalidomide 56-68 negative elongation factor complex member C/D, Th1l Mus musculus 280-283 29951065-12 2018 In summary, DCs derived from monocytes in the presence of lenalidomide present a semi-mature phenotype, increased phagocytic capacity, reduced production of proinflammatory cytokines, and the ability to polarize T-cells toward predominant Th1-type responses; these are qualities that might be useful in the development of new immunotherapeutic treatments. Lenalidomide 58-70 negative elongation factor complex member C/D Homo sapiens 239-242 29967612-8 2018 These findings suggest that the combination of DCs plus lenalidomide and PD-1 blockade synergistically establishes a robust anti-myeloma immunity through a two-way mechanism, which inhibits immunosuppressive cells while activating effector cells with superior polarization of the Th1/Th2 balance in favor of the tumor immune response. Lenalidomide 56-68 heart and neural crest derivatives expressed 2 Mus musculus 284-287 29380550-7 2018 On the other hand, patients with del(13q14) and del(9p21) were less likely to benefit from lenalidomide. Lenalidomide 91-103 DEL13Q14 Homo sapiens 33-42 29755672-9 2018 PP242 exhibited a synergistic effect with lenalidomide and bortezomib, suggesting that mTOR inhibition can enhance the anti-angiogenic effect of these drugs. Lenalidomide 42-54 mechanistic target of rapamycin kinase Homo sapiens 87-91 29363546-8 2018 Pharmacological targeting of Ikaros by the immunomodulatory drug lenalidomide ameliorated the response of MM cells to LPS in a CD180-dependent manner in vitro and in vivo Thus, the CD180/MD-1 pathway may represent a novel mechanism of growth regulation of MM cells in a BM milieu and may be a therapeutic target of preventing the regrowth of dormant MM cells.Significance: This study describes a novel mechanism by which myeloma cells are regulated in the bone marrow, where drug resistance and dormancy can evolve after treatment, with potential therapeutic implications for treating this often untreatable blood cancer. Lenalidomide 65-77 IKAROS family zinc finger 1 Homo sapiens 29-35 29363546-8 2018 Pharmacological targeting of Ikaros by the immunomodulatory drug lenalidomide ameliorated the response of MM cells to LPS in a CD180-dependent manner in vitro and in vivo Thus, the CD180/MD-1 pathway may represent a novel mechanism of growth regulation of MM cells in a BM milieu and may be a therapeutic target of preventing the regrowth of dormant MM cells.Significance: This study describes a novel mechanism by which myeloma cells are regulated in the bone marrow, where drug resistance and dormancy can evolve after treatment, with potential therapeutic implications for treating this often untreatable blood cancer. Lenalidomide 65-77 CD180 molecule Homo sapiens 127-132 29363546-8 2018 Pharmacological targeting of Ikaros by the immunomodulatory drug lenalidomide ameliorated the response of MM cells to LPS in a CD180-dependent manner in vitro and in vivo Thus, the CD180/MD-1 pathway may represent a novel mechanism of growth regulation of MM cells in a BM milieu and may be a therapeutic target of preventing the regrowth of dormant MM cells.Significance: This study describes a novel mechanism by which myeloma cells are regulated in the bone marrow, where drug resistance and dormancy can evolve after treatment, with potential therapeutic implications for treating this often untreatable blood cancer. Lenalidomide 65-77 CD180 molecule Homo sapiens 181-186 29269520-0 2018 Emergence and evolution of TP53 mutations are key features of disease progression in myelodysplastic patients with lower-risk del(5q) treated with lenalidomide. Lenalidomide 147-159 tumor protein p53 Homo sapiens 27-31 29363149-7 2018 Finally, lenalidomide maintenance after ASCT2 was associated with longer PFS (41.0 vs 21.6 mo; P = .0034) and better OS (not yet reached vs 129.6 mo; P = .0434) compared with patients without maintenance. Lenalidomide 9-21 solute carrier family 1 member 5 Homo sapiens 40-45 29363149-8 2018 Our data suggest that a second ASCT and lenalidomide maintenance given at first relapse in MM after prior ASCT are associated with better survival rates. Lenalidomide 40-52 solute carrier family 1 member 5 Homo sapiens 106-110 29588478-4 2018 Lenalidomide, a drug that induces proteosomal degradation of IKZF1, also decreases pDC numbers in vivo, and reduces the ratio of pDC/cDC1 differentiated from progenitor cells in vitro in a dose-dependent manner. Lenalidomide 0-12 IKAROS family zinc finger 1 Homo sapiens 61-66 29588478-6 2018 DC and monocytes from patients with IKZF1 deficiency or lenalidomide-treated cultures secrete less IFN-alpha, TNF and IL-12. Lenalidomide 56-68 interferon alpha 1 Homo sapiens 99-108 29693175-13 2018 The expression levels of vascular endothelial growth factor (VEGF), interleukin 6 (IL-6) and basic fibroblast growth factor (bFGF) were reduced in the MVs from the RPMI-8226 cells exposed to bortezomib and lenalidomide. Lenalidomide 206-218 vascular endothelial growth factor A Homo sapiens 25-59 29693175-13 2018 The expression levels of vascular endothelial growth factor (VEGF), interleukin 6 (IL-6) and basic fibroblast growth factor (bFGF) were reduced in the MVs from the RPMI-8226 cells exposed to bortezomib and lenalidomide. Lenalidomide 206-218 fibroblast growth factor 2 Homo sapiens 125-129 29616116-7 2018 Lenalidomide treatment alone significantly reduced the p-ERK level compared with the control (P<0.05) and cisplatin treatment alone significantly increased it (P<0.01), however treatment with them in combination significantly reduced the p-ERK level in MDA-MB-231 cells compared with cisplatin treatment alone (P<0.05). Lenalidomide 0-12 mitogen-activated protein kinase 1 Homo sapiens 57-60 29616116-7 2018 Lenalidomide treatment alone significantly reduced the p-ERK level compared with the control (P<0.05) and cisplatin treatment alone significantly increased it (P<0.01), however treatment with them in combination significantly reduced the p-ERK level in MDA-MB-231 cells compared with cisplatin treatment alone (P<0.05). Lenalidomide 0-12 mitogen-activated protein kinase 1 Homo sapiens 246-249 29593101-17 2018 CONCLUSIONS: Thalidomide and lenalidomide improve mural cell coverage of bAVM vessels and reduce bAVM hemorrhage, which is likely through upregulation of Pdgfb expression. Lenalidomide 29-41 platelet derived growth factor, B polypeptide Mus musculus 154-159 29380550-9 2018 Predictive values of del(17p13) and t(14;16) to bortezomib and autoSCT are seemingly universal, but predictive marker del(13q14) and del(9p21) for lenalidomide response appears ethnicity-specific. Lenalidomide 147-159 DEL13Q14 Homo sapiens 118-127 29246938-10 2018 The groups had similar immune responses, including a two- to threefold increase in inducible ICOS+CD4+FoxP3- T-cell number.Conclusions: Our early-phase data suggested that ipilimumab plus lenalidomide is well tolerated after HSCT. Lenalidomide 188-200 inducible T cell costimulator Homo sapiens 93-97 29246938-10 2018 The groups had similar immune responses, including a two- to threefold increase in inducible ICOS+CD4+FoxP3- T-cell number.Conclusions: Our early-phase data suggested that ipilimumab plus lenalidomide is well tolerated after HSCT. Lenalidomide 188-200 forkhead box P3 Homo sapiens 102-107 29237802-0 2018 Phase I Dose-Escalation Study of Anti-CTLA-4 Antibody Ipilimumab and Lenalidomide in Patients with Advanced Cancers. Lenalidomide 69-81 cytotoxic T-lymphocyte associated protein 4 Homo sapiens 38-44 29317217-8 2018 Adding Z-DEVD-FMK, a caspase-3 inhibitor, abolished the HO-1/IRF4 reduction by panobinostat alone or in combination with lenalidomide, suggesting that caspase-3-mediated HO-1/IRF4/MYC degradation occurred. Lenalidomide 121-133 interferon regulatory factor 4 Homo sapiens 61-65 29588478-6 2018 DC and monocytes from patients with IKZF1 deficiency or lenalidomide-treated cultures secrete less IFN-alpha, TNF and IL-12. Lenalidomide 56-68 tumor necrosis factor Homo sapiens 110-113 29588478-7 2018 These results indicate that human DC development and function are regulated by IKZF1, providing further insights into the consequences of IKZF1 mutation on immune function and the mechanism of immunomodulation by lenalidomide. Lenalidomide 213-225 IKAROS family zinc finger 1 Homo sapiens 79-84 29317217-0 2018 Crucial role of HO-1/IRF4-dependent apoptosis induced by panobinostat and lenalidomide in multiple myeloma. Lenalidomide 74-86 heme oxygenase 1 Homo sapiens 16-20 29317217-0 2018 Crucial role of HO-1/IRF4-dependent apoptosis induced by panobinostat and lenalidomide in multiple myeloma. Lenalidomide 74-86 interferon regulatory factor 4 Homo sapiens 21-25 29317217-8 2018 Adding Z-DEVD-FMK, a caspase-3 inhibitor, abolished the HO-1/IRF4 reduction by panobinostat alone or in combination with lenalidomide, suggesting that caspase-3-mediated HO-1/IRF4/MYC degradation occurred. Lenalidomide 121-133 caspase 3 Homo sapiens 21-30 29317217-8 2018 Adding Z-DEVD-FMK, a caspase-3 inhibitor, abolished the HO-1/IRF4 reduction by panobinostat alone or in combination with lenalidomide, suggesting that caspase-3-mediated HO-1/IRF4/MYC degradation occurred. Lenalidomide 121-133 heme oxygenase 1 Homo sapiens 56-60 29445144-3 2018 Here, we identify that lenalidomide alone could promote macrophages M2 polarization to prevent the progression of EAE, which is associated with subsequent inhibition of proinflammatory Th1 and Th17 cells both in peripheral lymph system and CNS. Lenalidomide 23-35 negative elongation factor complex member C/D, Th1l Mus musculus 185-188 29445144-5 2018 The macrophages-derived IL10 was upregulated both in vivo and in vitro after lenalidomide treatment. Lenalidomide 77-89 interleukin 10 Mus musculus 24-28 29445144-6 2018 Moreover, lenalidomide-treated IL10-dificient EAE mice had higher clinical scores and more severe CNS damage, and intravenous injection of lenalidomide-treated IL10-/- BMDMs into mice with EAE at disease onset did not reverse disease severity, implying IL10 may be essential in lenalidomide-ameliorated EAE. Lenalidomide 10-22 interleukin 10 Mus musculus 31-35 29445144-6 2018 Moreover, lenalidomide-treated IL10-dificient EAE mice had higher clinical scores and more severe CNS damage, and intravenous injection of lenalidomide-treated IL10-/- BMDMs into mice with EAE at disease onset did not reverse disease severity, implying IL10 may be essential in lenalidomide-ameliorated EAE. Lenalidomide 139-151 interleukin 10 Mus musculus 160-164 29445144-6 2018 Moreover, lenalidomide-treated IL10-dificient EAE mice had higher clinical scores and more severe CNS damage, and intravenous injection of lenalidomide-treated IL10-/- BMDMs into mice with EAE at disease onset did not reverse disease severity, implying IL10 may be essential in lenalidomide-ameliorated EAE. Lenalidomide 139-151 interleukin 10 Mus musculus 160-164 29445144-6 2018 Moreover, lenalidomide-treated IL10-dificient EAE mice had higher clinical scores and more severe CNS damage, and intravenous injection of lenalidomide-treated IL10-/- BMDMs into mice with EAE at disease onset did not reverse disease severity, implying IL10 may be essential in lenalidomide-ameliorated EAE. Lenalidomide 139-151 interleukin 10 Mus musculus 160-164 29445144-6 2018 Moreover, lenalidomide-treated IL10-dificient EAE mice had higher clinical scores and more severe CNS damage, and intravenous injection of lenalidomide-treated IL10-/- BMDMs into mice with EAE at disease onset did not reverse disease severity, implying IL10 may be essential in lenalidomide-ameliorated EAE. Lenalidomide 139-151 interleukin 10 Mus musculus 160-164 29445144-7 2018 Mechanistically, lenalidomide significantly increased expression and autocrine secretion of IL10, subsequently activated STAT3-mediated expression of Ym1. Lenalidomide 17-29 interleukin 10 Mus musculus 92-96 29445144-7 2018 Mechanistically, lenalidomide significantly increased expression and autocrine secretion of IL10, subsequently activated STAT3-mediated expression of Ym1. Lenalidomide 17-29 signal transducer and activator of transcription 3 Mus musculus 121-126 29445144-7 2018 Mechanistically, lenalidomide significantly increased expression and autocrine secretion of IL10, subsequently activated STAT3-mediated expression of Ym1. Lenalidomide 17-29 chitinase-like 3 Mus musculus 150-153 29440639-8 2018 Furthermore, we found that even though lenalidomide positively regulated RhoU expression leading to higher cell migration rates, it actually led to cell cycle arrest probably through a p21 dependent mechanism. Lenalidomide 39-51 ras homolog family member U Homo sapiens 73-77 28833354-0 2018 Immunohistochemical expression of cereblon and MUM1 as potential predictive markers of response to lenalidomide in extranodal marginal zone B-cell lymphoma of the mucosa-associated lymphoid tissue (MALT lymphoma). Lenalidomide 99-111 cereblon Homo sapiens 34-42 28833354-2 2018 Recently, high expression levels of cereblon (CRBN) and MUM1 have been associated with better response rates in multiple myeloma treated with lenalidomide. Lenalidomide 142-154 cereblon Homo sapiens 36-44 28833354-2 2018 Recently, high expression levels of cereblon (CRBN) and MUM1 have been associated with better response rates in multiple myeloma treated with lenalidomide. Lenalidomide 142-154 cereblon Homo sapiens 46-50 28833354-2 2018 Recently, high expression levels of cereblon (CRBN) and MUM1 have been associated with better response rates in multiple myeloma treated with lenalidomide. Lenalidomide 142-154 PWWP domain containing 3A, DNA repair factor Homo sapiens 56-60 28833354-4 2018 In the current study, we have systematically investigated a potential correlation of CRBN/MUM1 immunohistochemical expression and response to lenalidomide-based therapy in a series of 46 patients with MALT lymphoma treated at the Medical University Vienna 2009 to 2014. Lenalidomide 142-154 cereblon Homo sapiens 85-89 28833354-4 2018 In the current study, we have systematically investigated a potential correlation of CRBN/MUM1 immunohistochemical expression and response to lenalidomide-based therapy in a series of 46 patients with MALT lymphoma treated at the Medical University Vienna 2009 to 2014. Lenalidomide 142-154 PWWP domain containing 3A, DNA repair factor Homo sapiens 90-94 28639485-0 2018 Increased SHISA3 expression characterizes chronic lymphocytic leukemia patients sensitive to lenalidomide. Lenalidomide 93-105 shisa family member 3 Homo sapiens 10-16 28639485-9 2018 SHISA3highCLL are characterized by a restrained activation of Wnt signaling and sensibility to lenalidomide-induced apoptosis. Lenalidomide 95-107 shisa family member 3 Homo sapiens 0-6 28639485-10 2018 In conclusion, SHISA3 is a candidate gene for the identification of CLL patients who will benefit of lenalidomide treatment as single agent. Lenalidomide 101-113 shisa family member 3 Homo sapiens 15-21 28425720-1 2018 The drugs lenalidomide and pomalidomide bind to the protein cereblon, directing the CRL4-CRBN E3 ligase toward the transcription factors Ikaros and Aiolos to cause their ubiquitination and degradation. Lenalidomide 10-22 interleukin 17 receptor B Homo sapiens 84-88 28425720-1 2018 The drugs lenalidomide and pomalidomide bind to the protein cereblon, directing the CRL4-CRBN E3 ligase toward the transcription factors Ikaros and Aiolos to cause their ubiquitination and degradation. Lenalidomide 10-22 cereblon Homo sapiens 89-93 28425720-1 2018 The drugs lenalidomide and pomalidomide bind to the protein cereblon, directing the CRL4-CRBN E3 ligase toward the transcription factors Ikaros and Aiolos to cause their ubiquitination and degradation. Lenalidomide 10-22 IKAROS family zinc finger 3 Homo sapiens 148-154 29180353-5 2018 Inhibition of the Clec16a pathway by the chemotherapeutic lenalidomide, a selective ubiquitin ligase inhibitor associated with new-onset diabetes, impairs beta-cell mitophagy, oxygen consumption, and insulin secretion. Lenalidomide 58-70 C-type lectin domain containing 16A Homo sapiens 18-25 29180353-7 2018 Moreover, the beta-cell Clec16a-Nrdp1-USP8 mitophagy complex is destabilized and dysfunctional after lenalidomide treatment as well as after glucolipotoxic stress. Lenalidomide 101-113 C-type lectin domain containing 16A Homo sapiens 24-31 29180353-7 2018 Moreover, the beta-cell Clec16a-Nrdp1-USP8 mitophagy complex is destabilized and dysfunctional after lenalidomide treatment as well as after glucolipotoxic stress. Lenalidomide 101-113 ring finger protein 41 Homo sapiens 32-37 29180353-7 2018 Moreover, the beta-cell Clec16a-Nrdp1-USP8 mitophagy complex is destabilized and dysfunctional after lenalidomide treatment as well as after glucolipotoxic stress. Lenalidomide 101-113 ubiquitin specific peptidase 8 Homo sapiens 38-42 29295936-7 2018 In addition, STAT3 negatively regulates the lethal type I IFN signaling pathway by inhibiting expression of IRF7, IRF9, STAT1, and STAT2 Inhibition of STAT3 activity by ruxolitinib synergizes with the type I IFN inducer lenalidomide in growth inhibition of ABC DLBCL cells in vitro and in a xenograft mouse model. Lenalidomide 220-232 signal transducer and activator of transcription 3 Mus musculus 13-18 29295936-7 2018 In addition, STAT3 negatively regulates the lethal type I IFN signaling pathway by inhibiting expression of IRF7, IRF9, STAT1, and STAT2 Inhibition of STAT3 activity by ruxolitinib synergizes with the type I IFN inducer lenalidomide in growth inhibition of ABC DLBCL cells in vitro and in a xenograft mouse model. Lenalidomide 220-232 interferon regulatory factor 7 Mus musculus 108-112 29295936-7 2018 In addition, STAT3 negatively regulates the lethal type I IFN signaling pathway by inhibiting expression of IRF7, IRF9, STAT1, and STAT2 Inhibition of STAT3 activity by ruxolitinib synergizes with the type I IFN inducer lenalidomide in growth inhibition of ABC DLBCL cells in vitro and in a xenograft mouse model. Lenalidomide 220-232 interferon regulatory factor 9 Mus musculus 114-118 29295936-7 2018 In addition, STAT3 negatively regulates the lethal type I IFN signaling pathway by inhibiting expression of IRF7, IRF9, STAT1, and STAT2 Inhibition of STAT3 activity by ruxolitinib synergizes with the type I IFN inducer lenalidomide in growth inhibition of ABC DLBCL cells in vitro and in a xenograft mouse model. Lenalidomide 220-232 signal transducer and activator of transcription 3 Mus musculus 151-156 29317217-8 2018 Adding Z-DEVD-FMK, a caspase-3 inhibitor, abolished the HO-1/IRF4 reduction by panobinostat alone or in combination with lenalidomide, suggesting that caspase-3-mediated HO-1/IRF4/MYC degradation occurred. Lenalidomide 121-133 caspase 3 Homo sapiens 151-160 29317217-8 2018 Adding Z-DEVD-FMK, a caspase-3 inhibitor, abolished the HO-1/IRF4 reduction by panobinostat alone or in combination with lenalidomide, suggesting that caspase-3-mediated HO-1/IRF4/MYC degradation occurred. Lenalidomide 121-133 heme oxygenase 1 Homo sapiens 170-174 29317217-9 2018 Given that lenalidomide stabilized cereblon and facilitated IRF4 degradation in MM cells, we combined it with LBH589, an HDAC inhibitor. Lenalidomide 11-23 interferon regulatory factor 4 Homo sapiens 60-64 29317217-10 2018 LBH589 and lenalidomide exerted synergistic effects, and LBH589 reversed the efficacy of lenalidomide on the resistance of CD138+ primary MM cells, in part due to simultaneous suppression of HO-1, IRF4 and MYC. Lenalidomide 89-101 heme oxygenase 1 Homo sapiens 191-195 29379494-0 2017 The CXCR4-STAT3-IL-10 Pathway Controls the Immunoregulatory Function of Chronic Lymphocytic Leukemia and Is Modulated by Lenalidomide. Lenalidomide 121-133 C-X-C motif chemokine receptor 4 Homo sapiens 4-9 29317217-10 2018 LBH589 and lenalidomide exerted synergistic effects, and LBH589 reversed the efficacy of lenalidomide on the resistance of CD138+ primary MM cells, in part due to simultaneous suppression of HO-1, IRF4 and MYC. Lenalidomide 89-101 interferon regulatory factor 4 Homo sapiens 197-201 29379494-0 2017 The CXCR4-STAT3-IL-10 Pathway Controls the Immunoregulatory Function of Chronic Lymphocytic Leukemia and Is Modulated by Lenalidomide. Lenalidomide 121-133 signal transducer and activator of transcription 3 Homo sapiens 10-15 29317217-10 2018 LBH589 and lenalidomide exerted synergistic effects, and LBH589 reversed the efficacy of lenalidomide on the resistance of CD138+ primary MM cells, in part due to simultaneous suppression of HO-1, IRF4 and MYC. Lenalidomide 89-101 MYC proto-oncogene, bHLH transcription factor Homo sapiens 206-209 29379494-0 2017 The CXCR4-STAT3-IL-10 Pathway Controls the Immunoregulatory Function of Chronic Lymphocytic Leukemia and Is Modulated by Lenalidomide. Lenalidomide 121-133 interleukin 10 Homo sapiens 16-21 29177954-0 2018 Pharmacogenetic study of the impact of ABCB1 single-nucleotide polymorphisms on lenalidomide treatment outcomes in patients with multiple myeloma: results from a phase IV observational study and subsequent phase II clinical trial. Lenalidomide 80-92 ATP binding cassette subfamily B member 1 Homo sapiens 39-44 29379494-5 2017 Furthermore, experiments to assess the role of lenalidomide, an immunomodulatory agent with direct antitumor effect as well as pleiotropic activity on the immune system, showed that this agent prevents a CXCL12-induced increase in p-S727-STAT3 and the IL-10 response by CLL cells. Lenalidomide 47-59 C-X-C motif chemokine ligand 12 Homo sapiens 204-210 29379494-5 2017 Furthermore, experiments to assess the role of lenalidomide, an immunomodulatory agent with direct antitumor effect as well as pleiotropic activity on the immune system, showed that this agent prevents a CXCL12-induced increase in p-S727-STAT3 and the IL-10 response by CLL cells. Lenalidomide 47-59 signal transducer and activator of transcription 3 Homo sapiens 238-243 29379494-5 2017 Furthermore, experiments to assess the role of lenalidomide, an immunomodulatory agent with direct antitumor effect as well as pleiotropic activity on the immune system, showed that this agent prevents a CXCL12-induced increase in p-S727-STAT3 and the IL-10 response by CLL cells. Lenalidomide 47-59 interleukin 10 Homo sapiens 252-257 29379494-6 2017 Lenalidomide also suppressed IL-10-induced Y705-STAT3 phosphorylation in healthy T cells, thus reversing CLL-induced T-cell dysfunction. Lenalidomide 0-12 interleukin 10 Homo sapiens 29-34 29379494-6 2017 Lenalidomide also suppressed IL-10-induced Y705-STAT3 phosphorylation in healthy T cells, thus reversing CLL-induced T-cell dysfunction. Lenalidomide 0-12 signal transducer and activator of transcription 3 Homo sapiens 48-53 29379494-8 2017 Lenalidomide appears to be able to reverse CLL-induced immunosuppression through including abrogation of the CXCL12-CXCR4-S727-STAT3-mediated IL-10 response by CLL cells and prevention of IL-10-induced phosphorylation of Y705-STAT3 in T cells. Lenalidomide 0-12 C-X-C motif chemokine ligand 12 Homo sapiens 109-115 29379494-8 2017 Lenalidomide appears to be able to reverse CLL-induced immunosuppression through including abrogation of the CXCL12-CXCR4-S727-STAT3-mediated IL-10 response by CLL cells and prevention of IL-10-induced phosphorylation of Y705-STAT3 in T cells. Lenalidomide 0-12 C-X-C motif chemokine receptor 4 Homo sapiens 116-121 29379494-8 2017 Lenalidomide appears to be able to reverse CLL-induced immunosuppression through including abrogation of the CXCL12-CXCR4-S727-STAT3-mediated IL-10 response by CLL cells and prevention of IL-10-induced phosphorylation of Y705-STAT3 in T cells. Lenalidomide 0-12 signal transducer and activator of transcription 3 Homo sapiens 127-132 29379494-8 2017 Lenalidomide appears to be able to reverse CLL-induced immunosuppression through including abrogation of the CXCL12-CXCR4-S727-STAT3-mediated IL-10 response by CLL cells and prevention of IL-10-induced phosphorylation of Y705-STAT3 in T cells. Lenalidomide 0-12 interleukin 10 Homo sapiens 142-147 29379494-8 2017 Lenalidomide appears to be able to reverse CLL-induced immunosuppression through including abrogation of the CXCL12-CXCR4-S727-STAT3-mediated IL-10 response by CLL cells and prevention of IL-10-induced phosphorylation of Y705-STAT3 in T cells. Lenalidomide 0-12 interleukin 10 Homo sapiens 188-193 29379494-8 2017 Lenalidomide appears to be able to reverse CLL-induced immunosuppression through including abrogation of the CXCL12-CXCR4-S727-STAT3-mediated IL-10 response by CLL cells and prevention of IL-10-induced phosphorylation of Y705-STAT3 in T cells. Lenalidomide 0-12 signal transducer and activator of transcription 3 Homo sapiens 226-231 29445483-0 2018 Lenalidomide decreased the PSA level for castration-resistant prostate cancer: a case report. Lenalidomide 0-12 aminopeptidase puromycin sensitive Homo sapiens 27-30 29177954-3 2018 Lenalidomide (Len) is excreted mainly via the kidneys, and, given the expression of P-gp in the renal tubuli, single-nucleotide polymorphisms (SNPs) in the ABCB1 gene may influence Len plasma concentrations and, subsequently, the outcome of treatment. Lenalidomide 0-12 phosphoglycolate phosphatase Homo sapiens 84-88 29177954-3 2018 Lenalidomide (Len) is excreted mainly via the kidneys, and, given the expression of P-gp in the renal tubuli, single-nucleotide polymorphisms (SNPs) in the ABCB1 gene may influence Len plasma concentrations and, subsequently, the outcome of treatment. Lenalidomide 0-12 ATP binding cassette subfamily B member 1 Homo sapiens 156-161 29177954-3 2018 Lenalidomide (Len) is excreted mainly via the kidneys, and, given the expression of P-gp in the renal tubuli, single-nucleotide polymorphisms (SNPs) in the ABCB1 gene may influence Len plasma concentrations and, subsequently, the outcome of treatment. Lenalidomide 0-3 ATP binding cassette subfamily B member 1 Homo sapiens 156-161 29177954-8 2018 CONCLUSIONS: Our findings show a limited influence of ABCB1 genotype on lenalidomide treatment efficacy and safety. Lenalidomide 72-84 ATP binding cassette subfamily B member 1 Homo sapiens 54-59 29348877-9 2017 IRF4 was identified as the potential mechanism of resistance to lenalidomide and pomalidomide in WM. Lenalidomide 64-76 interferon regulatory factor 4 Homo sapiens 0-4 29061640-0 2018 Lenalidomide Enhances the Function of CS1 Chimeric Antigen Receptor-Redirected T Cells Against Multiple Myeloma. Lenalidomide 0-12 ITPR interacting domain containing 2 Mus musculus 38-41 29061640-3 2018 CS1 CAR T cells were transduced and expanded in the presence of lenalidomide in vitro The phenotype and effector function of CS1 CAR T cells treated with and without lenalidomide were compared. Lenalidomide 64-76 ITPR interacting domain containing 2 Mus musculus 0-3 29061640-4 2018 Finally, CS1 CAR T cells and lenalidomide were administered to treat multiple myeloma-bearing mice as combinatorial therapy.Results: CS1 CAR T cells exhibited efficient antitumor activity when adoptively transferred into mice. Lenalidomide 29-41 ITPR interacting domain containing 2 Mus musculus 133-136 29061640-5 2018 Mechanistic studies indicated that the addition of lenalidomide during CS1 CAR T-cell expansion in vitro enhanced the immune functions of CS1 CAR T cells, including cytotoxicity, memory maintenance, Th1 cytokine production, and immune synapse formation. Lenalidomide 51-63 ITPR interacting domain containing 2 Mus musculus 71-74 29061640-5 2018 Mechanistic studies indicated that the addition of lenalidomide during CS1 CAR T-cell expansion in vitro enhanced the immune functions of CS1 CAR T cells, including cytotoxicity, memory maintenance, Th1 cytokine production, and immune synapse formation. Lenalidomide 51-63 recoverin Mus musculus 75-78 29061640-5 2018 Mechanistic studies indicated that the addition of lenalidomide during CS1 CAR T-cell expansion in vitro enhanced the immune functions of CS1 CAR T cells, including cytotoxicity, memory maintenance, Th1 cytokine production, and immune synapse formation. Lenalidomide 51-63 ITPR interacting domain containing 2 Mus musculus 138-141 29061640-5 2018 Mechanistic studies indicated that the addition of lenalidomide during CS1 CAR T-cell expansion in vitro enhanced the immune functions of CS1 CAR T cells, including cytotoxicity, memory maintenance, Th1 cytokine production, and immune synapse formation. Lenalidomide 51-63 recoverin Mus musculus 142-145 29061640-5 2018 Mechanistic studies indicated that the addition of lenalidomide during CS1 CAR T-cell expansion in vitro enhanced the immune functions of CS1 CAR T cells, including cytotoxicity, memory maintenance, Th1 cytokine production, and immune synapse formation. Lenalidomide 51-63 negative elongation factor complex member C/D, Th1l Mus musculus 199-202 29061640-6 2018 Furthermore, lenalidomide enhanced the antitumor activity and persistence of adoptively transferred CS1 CAR T cells in vivoConclusions: The study demonstrates that lenalidomide improves the anti-multiple myeloma properties of CS1-directed CAR T cells and provides a basis for a planned clinical trial using the combination of lenalidomide with engineered T cells against CS1 in relapsed myeloma. Lenalidomide 13-25 ITPR interacting domain containing 2 Mus musculus 100-103 29061640-6 2018 Furthermore, lenalidomide enhanced the antitumor activity and persistence of adoptively transferred CS1 CAR T cells in vivoConclusions: The study demonstrates that lenalidomide improves the anti-multiple myeloma properties of CS1-directed CAR T cells and provides a basis for a planned clinical trial using the combination of lenalidomide with engineered T cells against CS1 in relapsed myeloma. Lenalidomide 13-25 recoverin Mus musculus 104-107 29061640-6 2018 Furthermore, lenalidomide enhanced the antitumor activity and persistence of adoptively transferred CS1 CAR T cells in vivoConclusions: The study demonstrates that lenalidomide improves the anti-multiple myeloma properties of CS1-directed CAR T cells and provides a basis for a planned clinical trial using the combination of lenalidomide with engineered T cells against CS1 in relapsed myeloma. Lenalidomide 13-25 ITPR interacting domain containing 2 Mus musculus 226-229 29061640-6 2018 Furthermore, lenalidomide enhanced the antitumor activity and persistence of adoptively transferred CS1 CAR T cells in vivoConclusions: The study demonstrates that lenalidomide improves the anti-multiple myeloma properties of CS1-directed CAR T cells and provides a basis for a planned clinical trial using the combination of lenalidomide with engineered T cells against CS1 in relapsed myeloma. Lenalidomide 13-25 recoverin Mus musculus 239-242 29061640-6 2018 Furthermore, lenalidomide enhanced the antitumor activity and persistence of adoptively transferred CS1 CAR T cells in vivoConclusions: The study demonstrates that lenalidomide improves the anti-multiple myeloma properties of CS1-directed CAR T cells and provides a basis for a planned clinical trial using the combination of lenalidomide with engineered T cells against CS1 in relapsed myeloma. Lenalidomide 13-25 ITPR interacting domain containing 2 Mus musculus 226-229 29061640-6 2018 Furthermore, lenalidomide enhanced the antitumor activity and persistence of adoptively transferred CS1 CAR T cells in vivoConclusions: The study demonstrates that lenalidomide improves the anti-multiple myeloma properties of CS1-directed CAR T cells and provides a basis for a planned clinical trial using the combination of lenalidomide with engineered T cells against CS1 in relapsed myeloma. Lenalidomide 164-176 ITPR interacting domain containing 2 Mus musculus 100-103 29061640-6 2018 Furthermore, lenalidomide enhanced the antitumor activity and persistence of adoptively transferred CS1 CAR T cells in vivoConclusions: The study demonstrates that lenalidomide improves the anti-multiple myeloma properties of CS1-directed CAR T cells and provides a basis for a planned clinical trial using the combination of lenalidomide with engineered T cells against CS1 in relapsed myeloma. Lenalidomide 164-176 recoverin Mus musculus 104-107 29061640-6 2018 Furthermore, lenalidomide enhanced the antitumor activity and persistence of adoptively transferred CS1 CAR T cells in vivoConclusions: The study demonstrates that lenalidomide improves the anti-multiple myeloma properties of CS1-directed CAR T cells and provides a basis for a planned clinical trial using the combination of lenalidomide with engineered T cells against CS1 in relapsed myeloma. Lenalidomide 164-176 ITPR interacting domain containing 2 Mus musculus 226-229 29061640-6 2018 Furthermore, lenalidomide enhanced the antitumor activity and persistence of adoptively transferred CS1 CAR T cells in vivoConclusions: The study demonstrates that lenalidomide improves the anti-multiple myeloma properties of CS1-directed CAR T cells and provides a basis for a planned clinical trial using the combination of lenalidomide with engineered T cells against CS1 in relapsed myeloma. Lenalidomide 164-176 recoverin Mus musculus 239-242 29061640-6 2018 Furthermore, lenalidomide enhanced the antitumor activity and persistence of adoptively transferred CS1 CAR T cells in vivoConclusions: The study demonstrates that lenalidomide improves the anti-multiple myeloma properties of CS1-directed CAR T cells and provides a basis for a planned clinical trial using the combination of lenalidomide with engineered T cells against CS1 in relapsed myeloma. Lenalidomide 164-176 ITPR interacting domain containing 2 Mus musculus 226-229 29061640-6 2018 Furthermore, lenalidomide enhanced the antitumor activity and persistence of adoptively transferred CS1 CAR T cells in vivoConclusions: The study demonstrates that lenalidomide improves the anti-multiple myeloma properties of CS1-directed CAR T cells and provides a basis for a planned clinical trial using the combination of lenalidomide with engineered T cells against CS1 in relapsed myeloma. Lenalidomide 164-176 ITPR interacting domain containing 2 Mus musculus 100-103 29061640-6 2018 Furthermore, lenalidomide enhanced the antitumor activity and persistence of adoptively transferred CS1 CAR T cells in vivoConclusions: The study demonstrates that lenalidomide improves the anti-multiple myeloma properties of CS1-directed CAR T cells and provides a basis for a planned clinical trial using the combination of lenalidomide with engineered T cells against CS1 in relapsed myeloma. Lenalidomide 164-176 recoverin Mus musculus 104-107 29061640-6 2018 Furthermore, lenalidomide enhanced the antitumor activity and persistence of adoptively transferred CS1 CAR T cells in vivoConclusions: The study demonstrates that lenalidomide improves the anti-multiple myeloma properties of CS1-directed CAR T cells and provides a basis for a planned clinical trial using the combination of lenalidomide with engineered T cells against CS1 in relapsed myeloma. Lenalidomide 164-176 ITPR interacting domain containing 2 Mus musculus 226-229 29061640-6 2018 Furthermore, lenalidomide enhanced the antitumor activity and persistence of adoptively transferred CS1 CAR T cells in vivoConclusions: The study demonstrates that lenalidomide improves the anti-multiple myeloma properties of CS1-directed CAR T cells and provides a basis for a planned clinical trial using the combination of lenalidomide with engineered T cells against CS1 in relapsed myeloma. Lenalidomide 164-176 recoverin Mus musculus 239-242 29061640-6 2018 Furthermore, lenalidomide enhanced the antitumor activity and persistence of adoptively transferred CS1 CAR T cells in vivoConclusions: The study demonstrates that lenalidomide improves the anti-multiple myeloma properties of CS1-directed CAR T cells and provides a basis for a planned clinical trial using the combination of lenalidomide with engineered T cells against CS1 in relapsed myeloma. Lenalidomide 164-176 ITPR interacting domain containing 2 Mus musculus 226-229 29578180-0 2018 Effect of lenalidomide on the human gastric cancer cell line SGC7901/vincristine Notch signaling. Lenalidomide 10-22 notch receptor 2 Homo sapiens 81-86 29578180-1 2018 Aim of Study: To examine the function of lenalidomide (LEN) on the human multidrug resistance (MDR)-type gastric cancer line SGC7901/vincristine (VCR) via regulating Notch signaling. Lenalidomide 41-53 notch receptor 2 Homo sapiens 166-171 30185705-7 2018 Pharmacological targeting of Ikaros with lenalidomide ameliorated the response of MM cells to LPS in a CD180-dependent manner in vitro and in vivo. Lenalidomide 41-53 IKAROS family zinc finger 1 Mus musculus 29-35 30185705-7 2018 Pharmacological targeting of Ikaros with lenalidomide ameliorated the response of MM cells to LPS in a CD180-dependent manner in vitro and in vivo. Lenalidomide 41-53 CD180 antigen Mus musculus 103-108 27301994-0 2017 Durable remission in a patient with leptomeningeal relapse of a MYC/BCL6-positive double-hit DLBCL treated with lenalidomide monotherapy. Lenalidomide 112-124 MYC proto-oncogene, bHLH transcription factor Homo sapiens 64-67 28893618-0 2017 Lenalidomide modulates gene expression in human ABC-DLBCL cells by regulating IKAROS interaction with an intronic control region of SPIB. Lenalidomide 0-12 IKAROS family zinc finger 1 Homo sapiens 78-84 28893618-0 2017 Lenalidomide modulates gene expression in human ABC-DLBCL cells by regulating IKAROS interaction with an intronic control region of SPIB. Lenalidomide 0-12 Spi-B transcription factor Homo sapiens 132-136 28893618-2 2017 Lenalidomide, an immunomodulatory drug in trials for treatment of ABC-DLBCL, targets the transcription factor IKAROS for degradation by the cereblon E3 ubiquitin ligase complex. Lenalidomide 0-12 IKAROS family zinc finger 1 Homo sapiens 110-116 28893618-4 2017 Using cultured ABC-DLBCL cell lines, we found that high levels of SPI-B expression conferred resistance to lenalidomide. Lenalidomide 107-119 Spi-B transcription factor Homo sapiens 66-71 28893618-5 2017 Lenalidomide treatment of ABC-DLBCL cells resulted in downregulation of SPIB at the level of transcription. Lenalidomide 0-12 Spi-B transcription factor Homo sapiens 72-76 28893618-9 2017 These results show that the mechanism of action of lenalidomide in ABC-DLBCL cells involves downregulation of SPIB transcription by cereblon-induced degradation of IKAROS. Lenalidomide 51-63 Spi-B transcription factor Homo sapiens 110-114 28893618-9 2017 These results show that the mechanism of action of lenalidomide in ABC-DLBCL cells involves downregulation of SPIB transcription by cereblon-induced degradation of IKAROS. Lenalidomide 51-63 IKAROS family zinc finger 1 Homo sapiens 164-170 28983059-8 2017 Pre-incubation with lenalidomide significantly diminished suppression of erythropoietin production by S100A9 or tumor necrosis factor-alpha. Lenalidomide 20-32 erythropoietin Homo sapiens 73-87 28983059-8 2017 Pre-incubation with lenalidomide significantly diminished suppression of erythropoietin production by S100A9 or tumor necrosis factor-alpha. Lenalidomide 20-32 S100 calcium binding protein A9 Homo sapiens 102-139 28983059-9 2017 Moreover, in peripheral blood mononuclear cells from patients with myelodysplastic syndromes, lenalidomide significantly reduced steady-state S100A9 generation (P=0.01) and lipopolysaccharide-induced tumor necrosis factor-alpha elaboration (P=0.002). Lenalidomide 94-106 S100 calcium binding protein A9 Homo sapiens 142-148 28983059-9 2017 Moreover, in peripheral blood mononuclear cells from patients with myelodysplastic syndromes, lenalidomide significantly reduced steady-state S100A9 generation (P=0.01) and lipopolysaccharide-induced tumor necrosis factor-alpha elaboration (P=0.002). Lenalidomide 94-106 tumor necrosis factor Homo sapiens 200-227 28983059-11 2017 Moreover, baseline serum tumor necrosis factor-alpha concentration was significantly higher in responders to erythroid-stimulating agents (P=0.03), whereas lenalidomide responders had significantly lower tumor necrosis factor-alpha and higher S100A9 serum concentrations (P=0.03). Lenalidomide 156-168 tumor necrosis factor Homo sapiens 204-231 28983059-11 2017 Moreover, baseline serum tumor necrosis factor-alpha concentration was significantly higher in responders to erythroid-stimulating agents (P=0.03), whereas lenalidomide responders had significantly lower tumor necrosis factor-alpha and higher S100A9 serum concentrations (P=0.03). Lenalidomide 156-168 S100 calcium binding protein A9 Homo sapiens 243-249 27301994-0 2017 Durable remission in a patient with leptomeningeal relapse of a MYC/BCL6-positive double-hit DLBCL treated with lenalidomide monotherapy. Lenalidomide 112-124 BCL6 transcription repressor Homo sapiens 68-72 27301994-4 2017 Here we present the first case, to our knowledge, of a MYC/BCL6-positive double-hit diffuse large B-cell lymphoma relapsing in the leptomeninges that achieved an outstanding durable remission with single-agent lenalidomide following salvage chemotherapy. Lenalidomide 210-222 MYC proto-oncogene, bHLH transcription factor Homo sapiens 55-58 27301994-4 2017 Here we present the first case, to our knowledge, of a MYC/BCL6-positive double-hit diffuse large B-cell lymphoma relapsing in the leptomeninges that achieved an outstanding durable remission with single-agent lenalidomide following salvage chemotherapy. Lenalidomide 210-222 BCL6 transcription repressor Homo sapiens 59-63 28828689-3 2017 METHODS: In the previously completed phase 1 study RBP with a dose of 75 mg/m2 bendamustine days 1-2, prednisolone 100 mg days 1-4 and 25 mg lenalidomide days 1-21 was well tolerated. Lenalidomide 141-153 SURP and G-patch domain containing 1 Homo sapiens 51-54 29290933-0 2017 Prognostic impact of Ikaros expression in lenalidomide-treated multiple myeloma. Lenalidomide 42-54 IKAROS family zinc finger 1 Homo sapiens 21-27 28815645-2 2017 advanced hepatocellular carcinoma (HCC) AIM: To explore potential biomarkers of lenalidomide efficacy as second-line therapy for HCC. Lenalidomide 80-92 HCC Homo sapiens 35-38 28581522-10 2017 Finally, blockade of Rpn11 increases the cytotoxic activity of anti-MM agents lenalidomide, pomalidomide or dexamethasone. Lenalidomide 78-90 proteasome 26S subunit, non-ATPase 14 Homo sapiens 21-26 28958469-18 2017 PFS2 was significantly longer in the lenalidomide group than in the placebo group (median 57 5 months [47 7-NE] vs 32 7 months [26 4-49 0]; HR 0 46, 95% CI 0 29-0 70; p<0 01). Lenalidomide 37-49 GINS complex subunit 2 Homo sapiens 0-4 28815645-2 2017 advanced hepatocellular carcinoma (HCC) AIM: To explore potential biomarkers of lenalidomide efficacy as second-line therapy for HCC. Lenalidomide 80-92 HCC Homo sapiens 129-132 28815645-16 2017 CONCLUSIONS: Lenalidomide exhibited moderate activity as second-line therapy for advanced HCC. Lenalidomide 13-25 HCC Homo sapiens 90-93 28751557-2 2017 Clinical activity of lenalidomide relies on degradation of Ikaros and the consequent reduction in IRF4 expression, both required for myeloma cell survival and involved in the regulation of MYC transcription. Lenalidomide 21-33 IKAROS family zinc finger 1 Homo sapiens 59-65 28751557-2 2017 Clinical activity of lenalidomide relies on degradation of Ikaros and the consequent reduction in IRF4 expression, both required for myeloma cell survival and involved in the regulation of MYC transcription. Lenalidomide 21-33 interferon regulatory factor 4 Homo sapiens 98-102 28751557-2 2017 Clinical activity of lenalidomide relies on degradation of Ikaros and the consequent reduction in IRF4 expression, both required for myeloma cell survival and involved in the regulation of MYC transcription. Lenalidomide 21-33 MYC proto-oncogene, bHLH transcription factor Homo sapiens 189-192 28278708-0 2017 Myelodysplastic syndrome with concomitant del(5q) and JAK2 V617F mutation transformed to acute myeloid leukemia with an additional chromosomal abnormality after a long-term treatment with lenalidomide. Lenalidomide 188-200 Janus kinase 2 Homo sapiens 54-58 28681435-0 2017 Impact of lenalidomide-based induction therapy on the mobilization of CD34+ cells, blood graft cellular composition, and post-transplant recovery in myeloma patients: a prospective multicenter study. Lenalidomide 10-22 CD34 molecule Homo sapiens 70-74 28681435-2 2017 Previous studies have suggested a negative impact of lenalidomide on the mobilization of CD34+ cells. Lenalidomide 53-65 CD34 molecule Homo sapiens 89-93 28681435-8 2017 RESULTS: Patients in the lenalidomide arm had lower median peak CD34+ counts and approximately 40% lower CD34+ cell yields from the first apheresis session, but these differences were not significant. Lenalidomide 25-37 CD34 molecule Homo sapiens 64-68 28681435-8 2017 RESULTS: Patients in the lenalidomide arm had lower median peak CD34+ counts and approximately 40% lower CD34+ cell yields from the first apheresis session, but these differences were not significant. Lenalidomide 25-37 CD34 molecule Homo sapiens 105-109 28785100-0 2017 JAM-A overexpression is related to disease progression in diffuse large B-cell lymphoma and downregulated by lenalidomide. Lenalidomide 109-121 F11 receptor Homo sapiens 0-5 29228683-2 2017 While CD8+CD28- regulatory T-cells in patients with hematologic disorders display a known immune-escape mechanism, we show that lenalidomide can overcome the immunosuppressive impact of CD8+CD28- T-cells. Lenalidomide 128-140 CD28 molecule Homo sapiens 190-194 29228683-4 2017 We found that lenalidomide enhances the antigen-specific secretion of IFN-gamma and Granzyme B despite the addition of CD8+CD28- T-cells. Lenalidomide 14-26 interferon gamma Homo sapiens 70-79 29228683-4 2017 We found that lenalidomide enhances the antigen-specific secretion of IFN-gamma and Granzyme B despite the addition of CD8+CD28- T-cells. Lenalidomide 14-26 granzyme B Homo sapiens 84-94 29228683-5 2017 Furthermore, we showed that lenalidomide inhibits the IL-6 secretion of mononuclear cells, triggered by CD8+CD28- T-cells. Lenalidomide 28-40 interleukin 6 Homo sapiens 54-58 29228683-5 2017 Furthermore, we showed that lenalidomide inhibits the IL-6 secretion of mononuclear cells, triggered by CD8+CD28- T-cells. Lenalidomide 28-40 CD8a molecule Homo sapiens 104-107 29228683-5 2017 Furthermore, we showed that lenalidomide inhibits the IL-6 secretion of mononuclear cells, triggered by CD8+CD28- T-cells. Lenalidomide 28-40 CD28 molecule Homo sapiens 108-112 29228683-6 2017 The addition of IL-6 counteracts the action of lenalidomide based stimulation of IFN-gamma secretion and induction of T-cell maturation but not the secretion of Granzyme B. Lenalidomide 47-59 interleukin 6 Homo sapiens 16-20 29228683-6 2017 The addition of IL-6 counteracts the action of lenalidomide based stimulation of IFN-gamma secretion and induction of T-cell maturation but not the secretion of Granzyme B. Lenalidomide 47-59 interferon gamma Homo sapiens 81-90 29228683-8 2017 Analysis of the IL-6 modulating cereblon-binding protein KPNA2 showed the similar degradation capacity of lenalidomide and pomalidomide without explaining the divergent effects. Lenalidomide 106-118 interleukin 6 Homo sapiens 16-20 29228683-8 2017 Analysis of the IL-6 modulating cereblon-binding protein KPNA2 showed the similar degradation capacity of lenalidomide and pomalidomide without explaining the divergent effects. Lenalidomide 106-118 karyopherin subunit alpha 2 Homo sapiens 57-62 28709135-4 2017 Sustained inhibition of lymphatic vessels regeneration induced by lenalidomide or the soluble form of vascular endothelial growth factor receptor 3 (sVEGFR3) that neutralises lymphangiogenic vascular endothelial growth factor C (VEGF-C), significantly impaired antitumour efficacy of PDT. Lenalidomide 66-78 vascular endothelial growth factor C Mus musculus 191-227 28709135-6 2017 Lenalidomide also abrogated antitumour effects of the combination immunotherapy with PDT and anti-programmed death-ligand 1 (PD-L1) antibodies. Lenalidomide 0-12 CD274 antigen Mus musculus 125-130 28927072-1 2017 The mechanism of the anti-myeloma effect of the immunomodulatory drug lenalidomide relies upon the binding of lenalidomide or an analogue to cereblon (CRBN) ubiquitin ligase, which inhibits it and results in the degradation of Ikaros-family zinc finger proteins 1 and 3 (IKZF1 and IKZF3). Lenalidomide 70-82 cereblon Homo sapiens 151-155 28927072-1 2017 The mechanism of the anti-myeloma effect of the immunomodulatory drug lenalidomide relies upon the binding of lenalidomide or an analogue to cereblon (CRBN) ubiquitin ligase, which inhibits it and results in the degradation of Ikaros-family zinc finger proteins 1 and 3 (IKZF1 and IKZF3). Lenalidomide 70-82 IKAROS family zinc finger 1 Homo sapiens 227-269 28927072-1 2017 The mechanism of the anti-myeloma effect of the immunomodulatory drug lenalidomide relies upon the binding of lenalidomide or an analogue to cereblon (CRBN) ubiquitin ligase, which inhibits it and results in the degradation of Ikaros-family zinc finger proteins 1 and 3 (IKZF1 and IKZF3). Lenalidomide 70-82 IKAROS family zinc finger 1 Homo sapiens 271-276 28927072-1 2017 The mechanism of the anti-myeloma effect of the immunomodulatory drug lenalidomide relies upon the binding of lenalidomide or an analogue to cereblon (CRBN) ubiquitin ligase, which inhibits it and results in the degradation of Ikaros-family zinc finger proteins 1 and 3 (IKZF1 and IKZF3). Lenalidomide 70-82 IKAROS family zinc finger 3 Homo sapiens 281-286 28927072-1 2017 The mechanism of the anti-myeloma effect of the immunomodulatory drug lenalidomide relies upon the binding of lenalidomide or an analogue to cereblon (CRBN) ubiquitin ligase, which inhibits it and results in the degradation of Ikaros-family zinc finger proteins 1 and 3 (IKZF1 and IKZF3). Lenalidomide 110-122 cereblon Homo sapiens 151-155 28927072-1 2017 The mechanism of the anti-myeloma effect of the immunomodulatory drug lenalidomide relies upon the binding of lenalidomide or an analogue to cereblon (CRBN) ubiquitin ligase, which inhibits it and results in the degradation of Ikaros-family zinc finger proteins 1 and 3 (IKZF1 and IKZF3). Lenalidomide 110-122 IKAROS family zinc finger 1 Homo sapiens 227-269 28927072-1 2017 The mechanism of the anti-myeloma effect of the immunomodulatory drug lenalidomide relies upon the binding of lenalidomide or an analogue to cereblon (CRBN) ubiquitin ligase, which inhibits it and results in the degradation of Ikaros-family zinc finger proteins 1 and 3 (IKZF1 and IKZF3). Lenalidomide 110-122 IKAROS family zinc finger 1 Homo sapiens 271-276 28927072-1 2017 The mechanism of the anti-myeloma effect of the immunomodulatory drug lenalidomide relies upon the binding of lenalidomide or an analogue to cereblon (CRBN) ubiquitin ligase, which inhibits it and results in the degradation of Ikaros-family zinc finger proteins 1 and 3 (IKZF1 and IKZF3). Lenalidomide 110-122 IKAROS family zinc finger 3 Homo sapiens 281-286 28927072-3 2017 The present study revealed that arsenic trioxide upregulates the transcription and protein levels of CRBN, the anti-myeloma target of lenalidomide, thus potentiating the sensitivity of multiple myeloma cells to lenalidomide and enhancing the lenalidomide-dependent degradation of IKZF1 and IKZF3. Lenalidomide 134-146 cereblon Homo sapiens 101-105 28927072-3 2017 The present study revealed that arsenic trioxide upregulates the transcription and protein levels of CRBN, the anti-myeloma target of lenalidomide, thus potentiating the sensitivity of multiple myeloma cells to lenalidomide and enhancing the lenalidomide-dependent degradation of IKZF1 and IKZF3. Lenalidomide 134-146 IKAROS family zinc finger 1 Homo sapiens 280-285 28927072-3 2017 The present study revealed that arsenic trioxide upregulates the transcription and protein levels of CRBN, the anti-myeloma target of lenalidomide, thus potentiating the sensitivity of multiple myeloma cells to lenalidomide and enhancing the lenalidomide-dependent degradation of IKZF1 and IKZF3. Lenalidomide 134-146 IKAROS family zinc finger 3 Homo sapiens 290-295 28927072-3 2017 The present study revealed that arsenic trioxide upregulates the transcription and protein levels of CRBN, the anti-myeloma target of lenalidomide, thus potentiating the sensitivity of multiple myeloma cells to lenalidomide and enhancing the lenalidomide-dependent degradation of IKZF1 and IKZF3. Lenalidomide 211-223 cereblon Homo sapiens 101-105 28927072-3 2017 The present study revealed that arsenic trioxide upregulates the transcription and protein levels of CRBN, the anti-myeloma target of lenalidomide, thus potentiating the sensitivity of multiple myeloma cells to lenalidomide and enhancing the lenalidomide-dependent degradation of IKZF1 and IKZF3. Lenalidomide 211-223 cereblon Homo sapiens 101-105 28619709-5 2017 CRISPR-mediated depletion of endogenous FAM46C enhanced multiple myeloma cell growth, decreased Ig light chain and HSPA5/BIP expression, activated ERK and antiapoptotic signaling, and conferred relative resistance to dexamethasone and lenalidomide treatments. Lenalidomide 235-247 terminal nucleotidyltransferase 5C Mus musculus 40-46 28801515-0 2017 Lenalidomide-associated arterial thrombosis in a patient with JAK2 positive atypical myeloproliferative neoplasm. Lenalidomide 0-12 Janus kinase 2 Homo sapiens 62-66 29228683-0 2017 Lenalidomide overcomes the immunosuppression of regulatory CD8+CD28- T-cells. Lenalidomide 0-12 CD8a molecule Homo sapiens 59-62 29228683-0 2017 Lenalidomide overcomes the immunosuppression of regulatory CD8+CD28- T-cells. Lenalidomide 0-12 CD28 molecule Homo sapiens 63-67 29228683-2 2017 While CD8+CD28- regulatory T-cells in patients with hematologic disorders display a known immune-escape mechanism, we show that lenalidomide can overcome the immunosuppressive impact of CD8+CD28- T-cells. Lenalidomide 128-140 CD8a molecule Homo sapiens 186-189 28643330-0 2017 MUC1-C is a target in lenalidomide resistant multiple myeloma. Lenalidomide 22-34 mucin 1, cell surface associated Homo sapiens 0-4 28643330-1 2017 Lenalidomide (LEN) acts directly on multiple myeloma (MM) cells by inducing cereblon-mediated degradation of interferon regulatory factor 4, Ikaros (IKZF)1 and IKZF3, transcription factors that are essential for MM cell survival. Lenalidomide 0-12 interferon regulatory factor 4 Homo sapiens 109-139 28643330-1 2017 Lenalidomide (LEN) acts directly on multiple myeloma (MM) cells by inducing cereblon-mediated degradation of interferon regulatory factor 4, Ikaros (IKZF)1 and IKZF3, transcription factors that are essential for MM cell survival. Lenalidomide 0-12 IKAROS family zinc finger 1 Homo sapiens 141-147 28643330-1 2017 Lenalidomide (LEN) acts directly on multiple myeloma (MM) cells by inducing cereblon-mediated degradation of interferon regulatory factor 4, Ikaros (IKZF)1 and IKZF3, transcription factors that are essential for MM cell survival. Lenalidomide 0-12 IKAROS family zinc finger 1 Homo sapiens 149-155 28643330-1 2017 Lenalidomide (LEN) acts directly on multiple myeloma (MM) cells by inducing cereblon-mediated degradation of interferon regulatory factor 4, Ikaros (IKZF)1 and IKZF3, transcription factors that are essential for MM cell survival. Lenalidomide 0-12 IKAROS family zinc finger 3 Homo sapiens 160-165 28643330-1 2017 Lenalidomide (LEN) acts directly on multiple myeloma (MM) cells by inducing cereblon-mediated degradation of interferon regulatory factor 4, Ikaros (IKZF)1 and IKZF3, transcription factors that are essential for MM cell survival. Lenalidomide 14-17 interferon regulatory factor 4 Homo sapiens 109-139 28643330-1 2017 Lenalidomide (LEN) acts directly on multiple myeloma (MM) cells by inducing cereblon-mediated degradation of interferon regulatory factor 4, Ikaros (IKZF)1 and IKZF3, transcription factors that are essential for MM cell survival. Lenalidomide 14-17 IKAROS family zinc finger 1 Homo sapiens 141-147 28643330-1 2017 Lenalidomide (LEN) acts directly on multiple myeloma (MM) cells by inducing cereblon-mediated degradation of interferon regulatory factor 4, Ikaros (IKZF)1 and IKZF3, transcription factors that are essential for MM cell survival. Lenalidomide 14-17 IKAROS family zinc finger 1 Homo sapiens 149-155 28643330-1 2017 Lenalidomide (LEN) acts directly on multiple myeloma (MM) cells by inducing cereblon-mediated degradation of interferon regulatory factor 4, Ikaros (IKZF)1 and IKZF3, transcription factors that are essential for MM cell survival. Lenalidomide 14-17 IKAROS family zinc finger 3 Homo sapiens 160-165 28643330-4 2017 We show that the GO-203/LEN combination acts by synergistically increasing ROS and, in turn, suppressing beta-catenin. Lenalidomide 24-27 catenin beta 1 Homo sapiens 105-117 28643330-5 2017 LEN resistance has been linked to activation of the WNT/beta-catenin CD44 pathway. Lenalidomide 0-3 catenin beta 1 Homo sapiens 56-68 28643330-5 2017 LEN resistance has been linked to activation of the WNT/beta-catenin CD44 pathway. Lenalidomide 0-3 CD44 molecule (Indian blood group) Homo sapiens 69-73 28785100-6 2017 Lenalidomide downregulated JAM-A and downstream NODAL expression, resulting in inhibition of B-lymphoma cell invasion and epithelial-to-mesenchymal transition. Lenalidomide 0-12 F11 receptor Homo sapiens 27-32 28785100-6 2017 Lenalidomide downregulated JAM-A and downstream NODAL expression, resulting in inhibition of B-lymphoma cell invasion and epithelial-to-mesenchymal transition. Lenalidomide 0-12 nodal growth differentiation factor Homo sapiens 48-53 28785100-7 2017 In a murine xenograft model established with subcutaneous injection of JAM-A-overexpressing B-lymphoma cells, lenalidomide retarded tumor growth and prevented cell invasion to mesoendoderm-derived organs, consistent with the downregulation of JAM-A and NODAL expression. Lenalidomide 110-122 F11 receptor Mus musculus 71-76 28785100-7 2017 In a murine xenograft model established with subcutaneous injection of JAM-A-overexpressing B-lymphoma cells, lenalidomide retarded tumor growth and prevented cell invasion to mesoendoderm-derived organs, consistent with the downregulation of JAM-A and NODAL expression. Lenalidomide 110-122 F11 receptor Mus musculus 243-248 28785100-7 2017 In a murine xenograft model established with subcutaneous injection of JAM-A-overexpressing B-lymphoma cells, lenalidomide retarded tumor growth and prevented cell invasion to mesoendoderm-derived organs, consistent with the downregulation of JAM-A and NODAL expression. Lenalidomide 110-122 nodal Mus musculus 253-258 28785100-9 2017 Identified as a biomarker of stem cell property, JAM-A indicated the sensitivity of B-lymphoma cells to lenalidomide. Lenalidomide 104-116 F11 receptor Homo sapiens 49-54 28017969-9 2017 Our data suggest a prognostic role of IKZF1, IKZF3 and BSG expression levels in lenalidomide-treated multiple myeloma. Lenalidomide 80-92 IKAROS family zinc finger 1 Homo sapiens 38-43 28270494-11 2017 Combination of SJB and HDACi ACY-1215, bortezomib, lenalidomide, or pomalidomide triggers synergistic cytotoxicity.Conclusions: Our preclinical studies provide the framework for clinical evaluation of USP1 inhibitors, alone or in combination, as a potential novel multiple myeloma therapy. Lenalidomide 51-63 ubiquitin specific peptidase 1 Homo sapiens 201-205 28426350-11 2017 Conclusion Lenalidomide maintenance for 24 months after obtaining a CR or PR to R-CHOP significantly prolonged PFS in elderly patients with DLBCL. Lenalidomide 11-23 DNA damage inducible transcript 3 Homo sapiens 82-86 27967292-5 2017 P53 antagonism by lenalidomide or other therapeutics such as antisense oligonucleotides, repopulates erythroid precursors and enhances effective erythropoiesis. Lenalidomide 18-30 tumor protein p53 Homo sapiens 0-3 28903429-0 2017 Lenalidomide restores the osteogenic differentiation of bone marrow mesenchymal stem cells from multiple myeloma patients via deactivating Notch signaling pathway. Lenalidomide 0-12 notch receptor 1 Homo sapiens 139-144 28903429-7 2017 Furthermore, it is shown that the gene expression of Notch signaling molecules, including receptors, ligands and downstream factors are significantly decreased in MM-MSCs following lenalidomide treatment, compared with non-treated MM-MSCs. Lenalidomide 181-193 notch receptor 1 Homo sapiens 53-58 28903429-8 2017 Taken together, treatment with lenalidomide restores the osteogenic differentiation of MM-MSCs via deactivating Notch signaling pathway. Lenalidomide 31-43 notch receptor 1 Homo sapiens 112-117 28488026-0 2017 Carfilzomib and lenalidomide response related to VEGF and VEGFR2 germline polymorphisms. Lenalidomide 16-28 vascular endothelial growth factor A Homo sapiens 49-53 28488026-0 2017 Carfilzomib and lenalidomide response related to VEGF and VEGFR2 germline polymorphisms. Lenalidomide 16-28 kinase insert domain receptor Homo sapiens 58-64 28552667-9 2017 Also, results of the therapeutic mice model, in which SP2/0 cells- challenged mice were treated with 5mg/kg lenalidomide in combination with pcDNA3.1+NS3, reasonably agreed with those of the prophylactic model. Lenalidomide 108-120 Sp2 transcription factor Mus musculus 54-57 28017969-0 2017 IKZF1 expression is a prognostic marker in newly diagnosed standard-risk multiple myeloma treated with lenalidomide and intensive chemotherapy: a study of the German Myeloma Study Group (DSMM). Lenalidomide 103-115 IKAROS family zinc finger 1 Homo sapiens 0-5 28017969-2 2017 Lenalidomide binding to the Cereblon (CRBN) E3 ubiquitin ligase results in targeted ubiquitination and degradation of the lymphoid transcription factors Ikaros (IKZF1) and Aiolos (IKZF3) leading to growth inhibition of multiple myeloma cells. Lenalidomide 0-12 cereblon Homo sapiens 28-36 28017969-2 2017 Lenalidomide binding to the Cereblon (CRBN) E3 ubiquitin ligase results in targeted ubiquitination and degradation of the lymphoid transcription factors Ikaros (IKZF1) and Aiolos (IKZF3) leading to growth inhibition of multiple myeloma cells. Lenalidomide 0-12 cereblon Homo sapiens 38-42 28017969-2 2017 Lenalidomide binding to the Cereblon (CRBN) E3 ubiquitin ligase results in targeted ubiquitination and degradation of the lymphoid transcription factors Ikaros (IKZF1) and Aiolos (IKZF3) leading to growth inhibition of multiple myeloma cells. Lenalidomide 0-12 IKAROS family zinc finger 1 Homo sapiens 161-166 28017969-2 2017 Lenalidomide binding to the Cereblon (CRBN) E3 ubiquitin ligase results in targeted ubiquitination and degradation of the lymphoid transcription factors Ikaros (IKZF1) and Aiolos (IKZF3) leading to growth inhibition of multiple myeloma cells. Lenalidomide 0-12 IKAROS family zinc finger 3 Homo sapiens 180-185 28017969-3 2017 Recently, Basigin (BSG) was identified as another protein regulated by CRBN that is involved in the activity of lenalidomide. Lenalidomide 112-124 basigin (Ok blood group) Homo sapiens 10-17 28017969-3 2017 Recently, Basigin (BSG) was identified as another protein regulated by CRBN that is involved in the activity of lenalidomide. Lenalidomide 112-124 basigin (Ok blood group) Homo sapiens 19-22 28249894-4 2017 CD38 levels and the percentages of CD38high Tregs are increased by lenalidomide and pomalidomide. Lenalidomide 67-79 CD38 molecule Homo sapiens 0-4 28249894-4 2017 CD38 levels and the percentages of CD38high Tregs are increased by lenalidomide and pomalidomide. Lenalidomide 67-79 CD38 molecule Homo sapiens 35-39 28693194-9 2017 Therefore, the combination of warfarin and lenalidomide may cause a pharmacodynamic interaction, more likely by inhibiting the production of interleukin-6. Lenalidomide 43-55 interleukin 6 Homo sapiens 141-154 28958290-4 2017 In contrast, in non-del(5q) MDS, lenalidomide restores effective erythropoiesis via enhancement of erythropoietin (EPO) receptor-initiated transcriptional response arising from the assembly of signaling-competent receptor complexes within membrane lipid raft domains. Lenalidomide 33-45 erythropoietin Homo sapiens 99-113 28958290-4 2017 In contrast, in non-del(5q) MDS, lenalidomide restores effective erythropoiesis via enhancement of erythropoietin (EPO) receptor-initiated transcriptional response arising from the assembly of signaling-competent receptor complexes within membrane lipid raft domains. Lenalidomide 33-45 erythropoietin Homo sapiens 115-118 29296763-1 2017 A man with cytopenias, dysplasia, excess blasts, P53 and RUNX1 mutations, and ring chromosome 7 recovered after stopping lenalidomide. Lenalidomide 121-133 tumor protein p53 Homo sapiens 49-52 29296763-1 2017 A man with cytopenias, dysplasia, excess blasts, P53 and RUNX1 mutations, and ring chromosome 7 recovered after stopping lenalidomide. Lenalidomide 121-133 RUNX family transcription factor 1 Homo sapiens 57-62 28529032-0 2017 Synergistic Cytotoxicity of Lenalidomide and Dexamethasone in Mantle Cell Lymphoma via Cereblon-dependent Targeting of the IL-6/STAT3/PI3K Axis. Lenalidomide 28-40 interleukin 6 Homo sapiens 123-127 28529032-0 2017 Synergistic Cytotoxicity of Lenalidomide and Dexamethasone in Mantle Cell Lymphoma via Cereblon-dependent Targeting of the IL-6/STAT3/PI3K Axis. Lenalidomide 28-40 signal transducer and activator of transcription 3 Homo sapiens 128-133 28529032-2 2017 Cereblon is probably targeted by both lenalidomide and dexamethasone, which leads to synergistic cytotoxicity in MCL by inhibiting the interleukin-6/signal transducer and activator of transcription 3 (IL-6/STAT3), phosphatidylinositol 3-kinase (PI3K)/AKT and AKT2/Forkhead box O3 (FOXO3A)/BCL2-like 11 (BIM) pathways. Lenalidomide 38-50 interleukin 6 Homo sapiens 201-205 28529032-2 2017 Cereblon is probably targeted by both lenalidomide and dexamethasone, which leads to synergistic cytotoxicity in MCL by inhibiting the interleukin-6/signal transducer and activator of transcription 3 (IL-6/STAT3), phosphatidylinositol 3-kinase (PI3K)/AKT and AKT2/Forkhead box O3 (FOXO3A)/BCL2-like 11 (BIM) pathways. Lenalidomide 38-50 signal transducer and activator of transcription 3 Homo sapiens 206-211 28529032-2 2017 Cereblon is probably targeted by both lenalidomide and dexamethasone, which leads to synergistic cytotoxicity in MCL by inhibiting the interleukin-6/signal transducer and activator of transcription 3 (IL-6/STAT3), phosphatidylinositol 3-kinase (PI3K)/AKT and AKT2/Forkhead box O3 (FOXO3A)/BCL2-like 11 (BIM) pathways. Lenalidomide 38-50 phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit delta Homo sapiens 214-243 28529032-2 2017 Cereblon is probably targeted by both lenalidomide and dexamethasone, which leads to synergistic cytotoxicity in MCL by inhibiting the interleukin-6/signal transducer and activator of transcription 3 (IL-6/STAT3), phosphatidylinositol 3-kinase (PI3K)/AKT and AKT2/Forkhead box O3 (FOXO3A)/BCL2-like 11 (BIM) pathways. Lenalidomide 38-50 AKT serine/threonine kinase 1 Homo sapiens 251-254 28529032-2 2017 Cereblon is probably targeted by both lenalidomide and dexamethasone, which leads to synergistic cytotoxicity in MCL by inhibiting the interleukin-6/signal transducer and activator of transcription 3 (IL-6/STAT3), phosphatidylinositol 3-kinase (PI3K)/AKT and AKT2/Forkhead box O3 (FOXO3A)/BCL2-like 11 (BIM) pathways. Lenalidomide 38-50 AKT serine/threonine kinase 2 Homo sapiens 259-263 28529032-2 2017 Cereblon is probably targeted by both lenalidomide and dexamethasone, which leads to synergistic cytotoxicity in MCL by inhibiting the interleukin-6/signal transducer and activator of transcription 3 (IL-6/STAT3), phosphatidylinositol 3-kinase (PI3K)/AKT and AKT2/Forkhead box O3 (FOXO3A)/BCL2-like 11 (BIM) pathways. Lenalidomide 38-50 forkhead box O3 Homo sapiens 264-279 28529032-2 2017 Cereblon is probably targeted by both lenalidomide and dexamethasone, which leads to synergistic cytotoxicity in MCL by inhibiting the interleukin-6/signal transducer and activator of transcription 3 (IL-6/STAT3), phosphatidylinositol 3-kinase (PI3K)/AKT and AKT2/Forkhead box O3 (FOXO3A)/BCL2-like 11 (BIM) pathways. Lenalidomide 38-50 forkhead box O3 Homo sapiens 281-287 28529032-2 2017 Cereblon is probably targeted by both lenalidomide and dexamethasone, which leads to synergistic cytotoxicity in MCL by inhibiting the interleukin-6/signal transducer and activator of transcription 3 (IL-6/STAT3), phosphatidylinositol 3-kinase (PI3K)/AKT and AKT2/Forkhead box O3 (FOXO3A)/BCL2-like 11 (BIM) pathways. Lenalidomide 38-50 BCL2 like 11 Homo sapiens 289-301 28630385-10 2017 CONCLUSION: Treatment by lenalidomide and pcDNA3.1(+)/NS3 improves NK cytotoxicity up to 66.80% suggesting that lenalidomide can be used in parallel with such therapeutic vaccines as cancer vaccine or virus vaccines. Lenalidomide 112-124 KRAS proto-oncogene, GTPase Homo sapiens 54-57 28017969-3 2017 Recently, Basigin (BSG) was identified as another protein regulated by CRBN that is involved in the activity of lenalidomide. Lenalidomide 112-124 cereblon Homo sapiens 71-75 28017969-9 2017 Our data suggest a prognostic role of IKZF1, IKZF3 and BSG expression levels in lenalidomide-treated multiple myeloma. Lenalidomide 80-92 IKAROS family zinc finger 3 Homo sapiens 45-50 28017969-9 2017 Our data suggest a prognostic role of IKZF1, IKZF3 and BSG expression levels in lenalidomide-treated multiple myeloma. Lenalidomide 80-92 basigin (Ok blood group) Homo sapiens 55-58 28437394-2 2017 In particular, the substrate-receptor proteins of the cullin-ubiquitin ligase system play a key role in selective protein degradation, which is an essential component of the anti-myeloma activity of immunomodulatory drugs (IMiDs), such as lenalidomide. Lenalidomide 239-251 CDK2 associated cullin domain 1 Homo sapiens 54-60 28530236-4 2017 Using a pulse-chase SILAC mass spectrometry-based proteomics approach, we demonstrate that lenalidomide induces the ubiquitination and degradation of ZFP91. Lenalidomide 91-103 ZFP91 zinc finger protein, atypical E3 ubiquitin ligase Homo sapiens 150-155 28881567-5 2017 We show here that lenalidomide (Len) and pomalidomide (Pom) prevent down-regulation of MHC-I during lytic activation, and restore ICAM-1 and B7-2 surface expression in latently infected PEL cells. Lenalidomide 18-30 intercellular adhesion molecule 1 Homo sapiens 130-136 28881567-5 2017 We show here that lenalidomide (Len) and pomalidomide (Pom) prevent down-regulation of MHC-I during lytic activation, and restore ICAM-1 and B7-2 surface expression in latently infected PEL cells. Lenalidomide 18-30 CD86 molecule Homo sapiens 141-145 28881567-5 2017 We show here that lenalidomide (Len) and pomalidomide (Pom) prevent down-regulation of MHC-I during lytic activation, and restore ICAM-1 and B7-2 surface expression in latently infected PEL cells. Lenalidomide 32-35 intercellular adhesion molecule 1 Homo sapiens 130-136 28881567-5 2017 We show here that lenalidomide (Len) and pomalidomide (Pom) prevent down-regulation of MHC-I during lytic activation, and restore ICAM-1 and B7-2 surface expression in latently infected PEL cells. Lenalidomide 32-35 CD86 molecule Homo sapiens 141-145 26961690-0 2017 Lenalidomide potentiates CD4+CD25+Treg-related suppression of lymphoma B-cell proliferation. Lenalidomide 0-12 CD4 molecule Homo sapiens 25-28 26961690-2 2017 Here we demonstrate that the immunomodulatory drug lenalidomide potentiates suppression of lymphoma B-cell proliferation by freshly isolated CD4+CD25+Tregs, as well as suppression by Tregs expanded polyclonally in the presence of rapamycin from CD4+CD25+T cells or CD4+CD25+CD127loT cells. Lenalidomide 51-63 CD4 molecule Homo sapiens 141-144 26961690-2 2017 Here we demonstrate that the immunomodulatory drug lenalidomide potentiates suppression of lymphoma B-cell proliferation by freshly isolated CD4+CD25+Tregs, as well as suppression by Tregs expanded polyclonally in the presence of rapamycin from CD4+CD25+T cells or CD4+CD25+CD127loT cells. Lenalidomide 51-63 CD4 molecule Homo sapiens 245-248 26961690-2 2017 Here we demonstrate that the immunomodulatory drug lenalidomide potentiates suppression of lymphoma B-cell proliferation by freshly isolated CD4+CD25+Tregs, as well as suppression by Tregs expanded polyclonally in the presence of rapamycin from CD4+CD25+T cells or CD4+CD25+CD127loT cells. Lenalidomide 51-63 CD4 molecule Homo sapiens 245-248 26961690-8 2017 Freshly isolated lymphoma cells activated with multimeric CD40L and IL-4 to support their survival in vitro varied in their sensitivity to lenalidomide, and the regulatory effect of Tregs on such lymphoma cells ranged from suppression to help in individual patients. Lenalidomide 139-151 interleukin 4 Homo sapiens 68-72 28199990-9 2017 Given the importance of an effective immune response to counteract the MM progression and the promising approaches using anti-PD-1/PD-L1 strategies, we will discuss in this review how Lenalidomide could represent an adequate approach to re-establish the recognition against MM by exhausted NK cell. Lenalidomide 184-196 programmed cell death 1 Homo sapiens 126-130 28628013-12 2017 DC were treated with lenalidomide and the expression of surface markers MHC Class I, MHC Class II, CD80, CD86, CD 205, and CD40 was increased. Lenalidomide 21-33 CD80 antigen Mus musculus 99-103 28628013-12 2017 DC were treated with lenalidomide and the expression of surface markers MHC Class I, MHC Class II, CD80, CD86, CD 205, and CD40 was increased. Lenalidomide 21-33 CD86 antigen Mus musculus 105-109 28628013-12 2017 DC were treated with lenalidomide and the expression of surface markers MHC Class I, MHC Class II, CD80, CD86, CD 205, and CD40 was increased. Lenalidomide 21-33 CD40 antigen Mus musculus 123-127 28460478-6 2017 This study suggests that a combination of tumor antigen-loaded DC vaccination and lenalidomide synergistically enhanced antitumor immune response in the murine colon cancer model, by inhibiting the generation of immune suppressive cells and recovery of effector cells, and demonstrated superior polarization of Th1/Th2 balance in favor of Th1 immune response. Lenalidomide 82-94 negative elongation factor complex member C/D, Th1l Mus musculus 311-314 28460478-6 2017 This study suggests that a combination of tumor antigen-loaded DC vaccination and lenalidomide synergistically enhanced antitumor immune response in the murine colon cancer model, by inhibiting the generation of immune suppressive cells and recovery of effector cells, and demonstrated superior polarization of Th1/Th2 balance in favor of Th1 immune response. Lenalidomide 82-94 heart and neural crest derivatives expressed 2 Mus musculus 315-318 28460478-6 2017 This study suggests that a combination of tumor antigen-loaded DC vaccination and lenalidomide synergistically enhanced antitumor immune response in the murine colon cancer model, by inhibiting the generation of immune suppressive cells and recovery of effector cells, and demonstrated superior polarization of Th1/Th2 balance in favor of Th1 immune response. Lenalidomide 82-94 negative elongation factor complex member C/D, Th1l Mus musculus 339-342 28199990-0 2017 Activation of NK cells and disruption of PD-L1/PD-1 axis: two different ways for lenalidomide to block myeloma progression. Lenalidomide 81-93 CD274 molecule Homo sapiens 41-46 28199990-0 2017 Activation of NK cells and disruption of PD-L1/PD-1 axis: two different ways for lenalidomide to block myeloma progression. Lenalidomide 81-93 programmed cell death 1 Homo sapiens 47-51 28199990-7 2017 This occurs since Lenalidomide acts on several critical points: stimulates T cell proliferation and cytokine secretion; decreases the expression of the immune check-point inhibitor Programmed Death-1 (PD-1) on both T and NK cells in MM patients; decreases the expression of both PD-1 and PD-L1 on MM cells; promotes MM cell death and abrogates MM/stromal microenvironment cross-talk, a process known to promote the MM cell survival and proliferation. Lenalidomide 18-30 programmed cell death 1 Homo sapiens 201-205 28199990-7 2017 This occurs since Lenalidomide acts on several critical points: stimulates T cell proliferation and cytokine secretion; decreases the expression of the immune check-point inhibitor Programmed Death-1 (PD-1) on both T and NK cells in MM patients; decreases the expression of both PD-1 and PD-L1 on MM cells; promotes MM cell death and abrogates MM/stromal microenvironment cross-talk, a process known to promote the MM cell survival and proliferation. Lenalidomide 18-30 programmed cell death 1 Homo sapiens 279-283 28199990-7 2017 This occurs since Lenalidomide acts on several critical points: stimulates T cell proliferation and cytokine secretion; decreases the expression of the immune check-point inhibitor Programmed Death-1 (PD-1) on both T and NK cells in MM patients; decreases the expression of both PD-1 and PD-L1 on MM cells; promotes MM cell death and abrogates MM/stromal microenvironment cross-talk, a process known to promote the MM cell survival and proliferation. Lenalidomide 18-30 CD274 molecule Sus scrofa 288-293 27686868-8 2017 Concomitant pan-Raf/PI3K inhibition was also effective in carfilzomib- and lenalidomide-resistant MM models underscoring that this attractive therapeutic anti-MM strategy is suitable for immediate clinical translation. Lenalidomide 75-87 zinc fingers and homeoboxes 2 Homo sapiens 16-19 28199990-9 2017 Given the importance of an effective immune response to counteract the MM progression and the promising approaches using anti-PD-1/PD-L1 strategies, we will discuss in this review how Lenalidomide could represent an adequate approach to re-establish the recognition against MM by exhausted NK cell. Lenalidomide 184-196 CD274 molecule Homo sapiens 131-136 28087699-4 2017 Pharmacological inhibition or genetic ablation of the Abl-DDB1-DDA1 axis decreases the ubiquitination of CRL4 substrates, including IKZF1 and IKZF3, in lenalidomide-treated multiple myeloma cells. Lenalidomide 152-164 ABL proto-oncogene 1, non-receptor tyrosine kinase Homo sapiens 54-57 28358372-7 2017 Notably, inhibition of TNF-alpha and LRG1 activity by Lenalidomide, an inhibitor of TNF-alpha production, in ACLT mice attenuated degeneration of osteoarthritis articular cartilage. Lenalidomide 54-66 tumor necrosis factor Mus musculus 23-32 28358372-7 2017 Notably, inhibition of TNF-alpha and LRG1 activity by Lenalidomide, an inhibitor of TNF-alpha production, in ACLT mice attenuated degeneration of osteoarthritis articular cartilage. Lenalidomide 54-66 leucine-rich alpha-2-glycoprotein 1 Mus musculus 37-41 28358372-7 2017 Notably, inhibition of TNF-alpha and LRG1 activity by Lenalidomide, an inhibitor of TNF-alpha production, in ACLT mice attenuated degeneration of osteoarthritis articular cartilage. Lenalidomide 54-66 tumor necrosis factor Mus musculus 84-93 28358372-10 2017 Inhibition of TNF-alpha and LRG1 by Lenalidomide could be a potential therapeutic approach. Lenalidomide 36-48 tumor necrosis factor Mus musculus 14-23 28358372-10 2017 Inhibition of TNF-alpha and LRG1 by Lenalidomide could be a potential therapeutic approach. Lenalidomide 36-48 leucine-rich alpha-2-glycoprotein 1 Mus musculus 28-32 28087699-4 2017 Pharmacological inhibition or genetic ablation of the Abl-DDB1-DDA1 axis decreases the ubiquitination of CRL4 substrates, including IKZF1 and IKZF3, in lenalidomide-treated multiple myeloma cells. Lenalidomide 152-164 damage specific DNA binding protein 1 Homo sapiens 58-62 28087699-4 2017 Pharmacological inhibition or genetic ablation of the Abl-DDB1-DDA1 axis decreases the ubiquitination of CRL4 substrates, including IKZF1 and IKZF3, in lenalidomide-treated multiple myeloma cells. Lenalidomide 152-164 DET1 and DDB1 associated 1 Homo sapiens 63-67 28087699-4 2017 Pharmacological inhibition or genetic ablation of the Abl-DDB1-DDA1 axis decreases the ubiquitination of CRL4 substrates, including IKZF1 and IKZF3, in lenalidomide-treated multiple myeloma cells. Lenalidomide 152-164 IKAROS family zinc finger 1 Homo sapiens 132-137 28087699-4 2017 Pharmacological inhibition or genetic ablation of the Abl-DDB1-DDA1 axis decreases the ubiquitination of CRL4 substrates, including IKZF1 and IKZF3, in lenalidomide-treated multiple myeloma cells. Lenalidomide 152-164 IKAROS family zinc finger 3 Homo sapiens 142-147 28087699-5 2017 Importantly, panobinostat, a recently approved anti-myeloma drug, and dexamethasone enhance lenalidomide-induced substrate degradation and cytotoxicity by activating c-Abl, therefore providing a mechanism underlying their combination with lenalidomide to treat multiple myeloma. Lenalidomide 92-104 ABL proto-oncogene 1, non-receptor tyrosine kinase Homo sapiens 166-171 28087699-5 2017 Importantly, panobinostat, a recently approved anti-myeloma drug, and dexamethasone enhance lenalidomide-induced substrate degradation and cytotoxicity by activating c-Abl, therefore providing a mechanism underlying their combination with lenalidomide to treat multiple myeloma. Lenalidomide 239-251 ABL proto-oncogene 1, non-receptor tyrosine kinase Homo sapiens 166-171 28052520-0 2017 IKAROS expression in distinct bone marrow cell populations as a candidate biomarker for outcome with lenalidomide-dexamethasone therapy in multiple myeloma. Lenalidomide 101-113 IKAROS family zinc finger 1 Homo sapiens 0-6 27893171-4 2017 RESULTS: Bortezomib-based regimens upfront followed by lenalidomide-based regimens at first relapse resulted in significantly better second progression-free survival (2ndPFS), PFS2, and overall survival (OS) compared to the opposite sequence. Lenalidomide 55-67 GINS complex subunit 2 Homo sapiens 176-180 27884971-6 2017 TP53 mutations were found in seven of nine patients who developed acute leukemia, and were documented to be present in the earliest sample (n=1) and acquired during lenalidomide treatment (n=6). Lenalidomide 165-177 tumor protein p53 Homo sapiens 0-4 28028022-6 2017 Lenalidomide increased intracellular H2O2 levels by inhibiting thioredoxin reductase (TrxR) in cells expressing CRBN, causing accumulation of immunoglobulin light-chain dimers, significantly increasing endoplasmic reticulum stress and inducing cytotoxicity by activation of BH3-only protein Bim in MM. Lenalidomide 0-12 peroxiredoxin 5 Homo sapiens 63-84 27862100-10 2017 DC were treated with 5-AzaC and lenalidomide and the expression of surface markers MHC Class I, MHC Class II, CD80, CD86, CD 205, and CD40 was increased. Lenalidomide 32-44 CD40 antigen Mus musculus 134-138 27862100-12 2017 Secretion of IL-12 and IL-15 by DC increased significantly with addition of 5-AzaC or 5-AzaC and lenalidomide. Lenalidomide 97-109 interleukin 15 Mus musculus 23-28 28028022-6 2017 Lenalidomide increased intracellular H2O2 levels by inhibiting thioredoxin reductase (TrxR) in cells expressing CRBN, causing accumulation of immunoglobulin light-chain dimers, significantly increasing endoplasmic reticulum stress and inducing cytotoxicity by activation of BH3-only protein Bim in MM. Lenalidomide 0-12 peroxiredoxin 5 Homo sapiens 86-90 28028022-6 2017 Lenalidomide increased intracellular H2O2 levels by inhibiting thioredoxin reductase (TrxR) in cells expressing CRBN, causing accumulation of immunoglobulin light-chain dimers, significantly increasing endoplasmic reticulum stress and inducing cytotoxicity by activation of BH3-only protein Bim in MM. Lenalidomide 0-12 cereblon Homo sapiens 112-116 26956578-6 2016 A linear relationship with good correlation coefficient (r = 0.997, n = 9) was found between the peak area and lenalidomide concentrations in the range of 100 to 950 ng mL-1 The limits of detection and quantitation were 28 and 100 ng mL-1, respectively. Lenalidomide 111-123 L1 cell adhesion molecule Mus musculus 171-175 28057080-4 2017 To test this hypothesis, we combined the use of a single-chain antibody targeting alpha-syn modified for improved central nervous system penetration (CD5-D5) with an unconventional anti-inflammatory treatment (lenalidomide) in the myelin basic protein (MBP)-alpha-syn transgenic mouse model of MSA. Lenalidomide 210-222 myelin basic protein Mus musculus 231-251 28057080-4 2017 To test this hypothesis, we combined the use of a single-chain antibody targeting alpha-syn modified for improved central nervous system penetration (CD5-D5) with an unconventional anti-inflammatory treatment (lenalidomide) in the myelin basic protein (MBP)-alpha-syn transgenic mouse model of MSA. Lenalidomide 210-222 myelin basic protein Mus musculus 253-256 28057080-5 2017 While the use of either CD5-D5 or lenalidomide alone had positive effects on neuroinflammation and/or alpha-syn accumulation in this mouse model of MSA, the combination of both approaches yielded better results than each single treatment. Lenalidomide 34-46 synuclein, alpha Mus musculus 102-111 28484169-5 2017 Moreover, the CSNK1A1 gene is known to play a role in the mechanism of action of lenalidomide. Lenalidomide 81-93 casein kinase 1 alpha 1 Homo sapiens 14-21 28592763-4 2017 Ibrutinib or lenalidomide was shown to be more effective in activated B-cell (ABC) -type-DLBCL than in germinal center B-cell type-DLBCL. Lenalidomide 13-25 ATP binding cassette subfamily B member 6 (Langereis blood group) Homo sapiens 78-81 27889516-7 2017 These results suggest that LEN enhanced the function of DCs generated from patients with multiple myeloma by stimulating the capacity of allogeneic T cells, inhibiting the generation of immunosuppressive cells, inducing naive T cells toward Th1 polarization, and generating potent myeloma-specific cytotoxic T lymphocytes. Lenalidomide 27-30 negative elongation factor complex member C/D Homo sapiens 241-244 29212067-0 2017 Lenalidomide Maintenance after R-CHOP Therapy in Diffuse Large B-Cell Lymphoma: Can It Be a Standard of Care. Lenalidomide 0-12 DNA damage inducible transcript 3 Homo sapiens 33-37 27698446-0 2017 Expansion of Th1-like Vgamma9Vdelta2T cells by new-generation IMiDs, lenalidomide and pomalidomide, in combination with zoledronic acid. Lenalidomide 69-81 negative elongation factor complex member C/D Homo sapiens 13-16 26956578-8 2016 In conclusion, the proposed method is suitable for the accurate quantification of lenalidomide in human plasma with a wide linear range, from 100 to 950 ng mL-1 This is a valuable method for pharmacokinetic studies of lenalidomide in human subjects. Lenalidomide 82-94 L1 cell adhesion molecule Mus musculus 158-162 27677742-8 2016 Lenalidomide and trabectedin prevent TAM recruitment mainly through CCL2 blockade. Lenalidomide 0-12 C-C motif chemokine ligand 2 Homo sapiens 68-72 27492707-7 2016 Binding of thalidomide or lenalidomide increases the affinity of CRBN to the transcription factors IKZF1 and IKZF3 and casein kinase 1alpha (CK1alpha). Lenalidomide 26-38 cereblon Homo sapiens 65-69 27776107-10 2016 Treatment with lenalidomide increased RhoA activity and reversed the migration and activation defects of RASGRP1-deficient lymphocytes. Lenalidomide 15-27 ras homolog family member A Homo sapiens 38-42 27776107-10 2016 Treatment with lenalidomide increased RhoA activity and reversed the migration and activation defects of RASGRP1-deficient lymphocytes. Lenalidomide 15-27 RAS guanyl releasing protein 1 Homo sapiens 105-112 27881177-0 2016 High IKZF1/3 protein expression is a favorable prognostic factor for survival of relapsed/refractory multiple myeloma patients treated with lenalidomide. Lenalidomide 140-152 IKAROS family zinc finger 1 Homo sapiens 5-12 27855285-8 2017 This computational biology network approach identified GGH overexpression as a potential resistance factor to HMA treatment and paradoxical activation of beta-catenin (through Csnk1a1 inhibition) as a resistance factor to lenalidomide treatment. Lenalidomide 222-234 catenin beta 1 Homo sapiens 154-166 27881177-1 2016 The aim of this study is to assess nucleoprotein expression of IKZF1/3 in patients with relapsed/refractory multiple myeloma (MM) who received lenalidomide-based therapy and correlated them with their clinical outcomes. Lenalidomide 143-155 IKAROS family zinc finger 1 Homo sapiens 63-68 27881177-5 2016 IKZF3 expression also appears predicted a favorable response to the lenalidomide-based therapy. Lenalidomide 68-80 IKAROS family zinc finger 3 Homo sapiens 0-5 27389073-5 2016 We found that after six months of lenalidomide therapy, the number of CD34(+) HSPCs decreased. Lenalidomide 34-46 CD34 molecule Homo sapiens 70-74 27657380-8 2016 Apoptosis was associated with increased hallmarks of ER stress and activation of UPR sensors and was mediated by dephosphorylation of the AKT, MAPK/ERK, and STAT3 pathways.The combination of romidepsin and lenalidomide shows promise as a possible treatment for T-cell lymphoma. Lenalidomide 206-218 AKT serine/threonine kinase 1 Homo sapiens 138-141 27748917-0 2016 Lenalidomide induces apoptosis and inhibits angiogenesis via caspase-3 and VEGF in hepatocellular carcinoma cells. Lenalidomide 0-12 caspase 3 Homo sapiens 61-70 27748917-0 2016 Lenalidomide induces apoptosis and inhibits angiogenesis via caspase-3 and VEGF in hepatocellular carcinoma cells. Lenalidomide 0-12 vascular endothelial growth factor A Homo sapiens 75-79 27748917-7 2016 It was identified that treatment of cells with lenalidomide and thalidomide led to a dose-dependent inhibition of cell proliferation, and the two drugs were able to induce cells apoptosis and inhibit VEGF expression in HCC cells. Lenalidomide 47-59 vascular endothelial growth factor A Homo sapiens 200-204 27748917-9 2016 In conclusion, the results indicated that lenalidomide induces apoptosis and inhibits angiogenesis in HCC cells via caspase-3 and VEGF pathway, and these may provide a potential perspective for lenalidomide"s application in clinical. Lenalidomide 42-54 caspase 3 Homo sapiens 116-125 27748917-9 2016 In conclusion, the results indicated that lenalidomide induces apoptosis and inhibits angiogenesis in HCC cells via caspase-3 and VEGF pathway, and these may provide a potential perspective for lenalidomide"s application in clinical. Lenalidomide 42-54 vascular endothelial growth factor A Homo sapiens 130-134 27748917-9 2016 In conclusion, the results indicated that lenalidomide induces apoptosis and inhibits angiogenesis in HCC cells via caspase-3 and VEGF pathway, and these may provide a potential perspective for lenalidomide"s application in clinical. Lenalidomide 194-206 caspase 3 Homo sapiens 116-125 27748917-9 2016 In conclusion, the results indicated that lenalidomide induces apoptosis and inhibits angiogenesis in HCC cells via caspase-3 and VEGF pathway, and these may provide a potential perspective for lenalidomide"s application in clinical. Lenalidomide 194-206 vascular endothelial growth factor A Homo sapiens 130-134 26186254-1 2016 Clinical interest in the measurement of Cereblon (CRBN), the primary target of the IMiDs immunomodulatory drugs lenalidomide and pomalidomide, has been fueled by its essential requirement for antitumor or immunomodulatory activity of both drugs in multiple myeloma (MM). Lenalidomide 112-124 cereblon Homo sapiens 40-48 26186254-1 2016 Clinical interest in the measurement of Cereblon (CRBN), the primary target of the IMiDs immunomodulatory drugs lenalidomide and pomalidomide, has been fueled by its essential requirement for antitumor or immunomodulatory activity of both drugs in multiple myeloma (MM). Lenalidomide 112-124 cereblon Homo sapiens 50-54 27657380-8 2016 Apoptosis was associated with increased hallmarks of ER stress and activation of UPR sensors and was mediated by dephosphorylation of the AKT, MAPK/ERK, and STAT3 pathways.The combination of romidepsin and lenalidomide shows promise as a possible treatment for T-cell lymphoma. Lenalidomide 206-218 mitogen-activated protein kinase 1 Homo sapiens 148-151 27657380-8 2016 Apoptosis was associated with increased hallmarks of ER stress and activation of UPR sensors and was mediated by dephosphorylation of the AKT, MAPK/ERK, and STAT3 pathways.The combination of romidepsin and lenalidomide shows promise as a possible treatment for T-cell lymphoma. Lenalidomide 206-218 signal transducer and activator of transcription 3 Homo sapiens 157-162 27365142-1 2016 Cereblon (CRBN) has been identified as a primary target of immunomodulatory drugs and is considered a biomarker for the prediction of outcomes after thalidomide- or lenalidomide-based treatments. Lenalidomide 165-177 cereblon Homo sapiens 10-14 27465366-8 2016 Finally, using lenalidomide (an inhibitor of TNF-alpha), a lower viral load was found in animals with lenalidomide. Lenalidomide 15-27 tumor necrosis factor Homo sapiens 45-54 27601648-0 2016 Rabex-5 is a lenalidomide target molecule that negatively regulates TLR-induced type 1 IFN production. Lenalidomide 13-25 RAB guanine nucleotide exchange factor 1 Homo sapiens 0-7 27601648-4 2016 Treatment with lenalidomide prevented the association of Cereblon with Rabex-5. Lenalidomide 15-27 RAB guanine nucleotide exchange factor 1 Homo sapiens 71-78 27611189-7 2016 Furthermore, anti-CD38-IFNalpha(attenuated) is more efficacious than standard MM treatments (lenalidomide, bortezomib, dexamethasone) and exhibits strong synergy with lenalidomide and with bortezomib in xenograft models. Lenalidomide 93-105 CD38 molecule Homo sapiens 18-22 27611189-7 2016 Furthermore, anti-CD38-IFNalpha(attenuated) is more efficacious than standard MM treatments (lenalidomide, bortezomib, dexamethasone) and exhibits strong synergy with lenalidomide and with bortezomib in xenograft models. Lenalidomide 93-105 interferon alpha 1 Homo sapiens 23-31 27611189-7 2016 Furthermore, anti-CD38-IFNalpha(attenuated) is more efficacious than standard MM treatments (lenalidomide, bortezomib, dexamethasone) and exhibits strong synergy with lenalidomide and with bortezomib in xenograft models. Lenalidomide 167-179 CD38 molecule Homo sapiens 18-22 27611189-7 2016 Furthermore, anti-CD38-IFNalpha(attenuated) is more efficacious than standard MM treatments (lenalidomide, bortezomib, dexamethasone) and exhibits strong synergy with lenalidomide and with bortezomib in xenograft models. Lenalidomide 167-179 interferon alpha 1 Homo sapiens 23-31 27465366-8 2016 Finally, using lenalidomide (an inhibitor of TNF-alpha), a lower viral load was found in animals with lenalidomide. Lenalidomide 102-114 tumor necrosis factor Homo sapiens 45-54 27479651-6 2016 The expression of P-gp was induced in SKM-1 cells by selective pressure using vincristine (VCR), mitoxantrone (MTX), azacytidine (AzaC) and lenalidomide (LEN). Lenalidomide 140-152 ATP binding cassette subfamily B member 1 Homo sapiens 18-22 27133825-0 2016 Prevalence, clonal dynamics and clinical impact of TP53 mutations in patients with myelodysplastic syndrome with isolated deletion (5q) treated with lenalidomide: results from a prospective multicenter study of the german MDS study group (GMDS). Lenalidomide 149-161 tumor protein p53 Homo sapiens 51-55 27479651-6 2016 The expression of P-gp was induced in SKM-1 cells by selective pressure using vincristine (VCR), mitoxantrone (MTX), azacytidine (AzaC) and lenalidomide (LEN). Lenalidomide 140-152 sodium voltage-gated channel alpha subunit 4 Homo sapiens 38-43 27479651-6 2016 The expression of P-gp was induced in SKM-1 cells by selective pressure using vincristine (VCR), mitoxantrone (MTX), azacytidine (AzaC) and lenalidomide (LEN). Lenalidomide 154-157 ATP binding cassette subfamily B member 1 Homo sapiens 18-22 27479651-6 2016 The expression of P-gp was induced in SKM-1 cells by selective pressure using vincristine (VCR), mitoxantrone (MTX), azacytidine (AzaC) and lenalidomide (LEN). Lenalidomide 154-157 sodium voltage-gated channel alpha subunit 4 Homo sapiens 38-43 27089170-0 2016 ROBUST: Lenalidomide-R-CHOP versus placebo-R-CHOP in previously untreated ABC-type diffuse large B-cell lymphoma. Lenalidomide 8-20 DNA damage inducible transcript 3 Homo sapiens 23-27 27538006-0 2016 Erythema Nodosum-like Septal Panniculitis Secondary to Lenalidomide Therapy in a Patient With Janus Kinase 2-Positive Myelofibrosis. Lenalidomide 55-67 Janus kinase 2 Homo sapiens 94-108 27538006-2 2016 We present a case of a 66-year-old gentleman with JAK2-positive myelofibrosis who developed transient EN-like lesions on his trunk and upper and lower extremities approximately three weeks after starting lenalidomide therapy. Lenalidomide 204-216 Janus kinase 2 Homo sapiens 50-54 27343012-2 2016 Several key findings suggest diverse roles of CRBN, including its regulation of the large-conductance calcium- and voltage-activated potassium (BKCa) channels, regulation of thalidomide-binding proteins, and mediation of lenalidomide treatment in multiple myeloma. Lenalidomide 221-233 cereblon Homo sapiens 46-50 27286995-7 2016 Lenalidomide clearance declined with decreased CrCl. Lenalidomide 0-12 CRCL Homo sapiens 47-51 26823144-8 2016 Furthermore, treatment with an anti-MM agent, lenalidomide, caused ubiquitination and proteasomal degradation of Blimp-1, leading to the de-repression of a new Blimp-1 direct target, CULLIN 4A (CUL4A), and reduced Aiolos levels. Lenalidomide 46-58 PR/SET domain 1 Homo sapiens 113-120 26823144-8 2016 Furthermore, treatment with an anti-MM agent, lenalidomide, caused ubiquitination and proteasomal degradation of Blimp-1, leading to the de-repression of a new Blimp-1 direct target, CULLIN 4A (CUL4A), and reduced Aiolos levels. Lenalidomide 46-58 PR/SET domain 1 Homo sapiens 160-167 26823144-8 2016 Furthermore, treatment with an anti-MM agent, lenalidomide, caused ubiquitination and proteasomal degradation of Blimp-1, leading to the de-repression of a new Blimp-1 direct target, CULLIN 4A (CUL4A), and reduced Aiolos levels. Lenalidomide 46-58 cullin 4A Homo sapiens 183-192 26823144-8 2016 Furthermore, treatment with an anti-MM agent, lenalidomide, caused ubiquitination and proteasomal degradation of Blimp-1, leading to the de-repression of a new Blimp-1 direct target, CULLIN 4A (CUL4A), and reduced Aiolos levels. Lenalidomide 46-58 cullin 4A Homo sapiens 194-199 26823144-8 2016 Furthermore, treatment with an anti-MM agent, lenalidomide, caused ubiquitination and proteasomal degradation of Blimp-1, leading to the de-repression of a new Blimp-1 direct target, CULLIN 4A (CUL4A), and reduced Aiolos levels. Lenalidomide 46-58 IKAROS family zinc finger 3 Homo sapiens 214-220 26823144-9 2016 Accordingly, lenalidomide-induced cell death was partially rescued by reintroduction of Blimp-1 or knockdown of CUL4A. Lenalidomide 13-25 PR/SET domain 1 Homo sapiens 88-95 26823144-9 2016 Accordingly, lenalidomide-induced cell death was partially rescued by reintroduction of Blimp-1 or knockdown of CUL4A. Lenalidomide 13-25 cullin 4A Homo sapiens 112-117 26823144-11 2016 We also showed that interruption of Blimp-1/Aiolos regulatory pathways contributes to lenalidomide-mediated anti-MM activity. Lenalidomide 86-98 PR/SET domain 1 Homo sapiens 36-43 26823144-11 2016 We also showed that interruption of Blimp-1/Aiolos regulatory pathways contributes to lenalidomide-mediated anti-MM activity. Lenalidomide 86-98 IKAROS family zinc finger 3 Homo sapiens 44-50 27089170-2 2016 In Phase II studies, lenalidomide combined with R-CHOP (R(2)-CHOP) improved outcomes relative to historical R-CHOP in newly diagnosed DLBCL, particularly in non-GCB cases. Lenalidomide 21-33 DNA damage inducible transcript 3 Homo sapiens 61-65 27089170-2 2016 In Phase II studies, lenalidomide combined with R-CHOP (R(2)-CHOP) improved outcomes relative to historical R-CHOP in newly diagnosed DLBCL, particularly in non-GCB cases. Lenalidomide 21-33 DNA damage inducible transcript 3 Homo sapiens 61-65 27330758-0 2016 Durable response to lenalidomide in a patient with myelodysplastic syndrome associated with isolated 5q deletion and JAK2 V617F mutation despite discontinuation of treatment. Lenalidomide 20-32 Janus kinase 2 Homo sapiens 117-121 27330758-10 2016 The present case highlights the coexistence of the JAK2 V617F mutation in del(5q) MDS and suggests that lenalidomide treatment is beneficial and effective for these patients, leading to complete hematological, cytogenetic and molecular response. Lenalidomide 104-116 Janus kinase 2 Homo sapiens 51-55 27294874-5 2016 In contrast, deletion of GPR68 or inhibition of calcium and calpain activation suppressed LEN-induced cytotoxicity. Lenalidomide 90-93 G protein-coupled receptor 68 Homo sapiens 25-30 27294874-7 2016 Depletion of CAST restored responsiveness of LEN-resistant non-del(5q) MDS cells and AML cells, providing an explanation for the superior responses of patients with del(5q) MDS to LEN treatment. Lenalidomide 45-48 calpastatin Homo sapiens 13-17 27287425-0 2016 Lenalidomide Induces Interleukin-21 Production by T Cells and Enhances IL21-Mediated Cytotoxicity in Chronic Lymphocytic Leukemia B Cells. Lenalidomide 0-12 interleukin 21 Homo sapiens 21-35 27287425-0 2016 Lenalidomide Induces Interleukin-21 Production by T Cells and Enhances IL21-Mediated Cytotoxicity in Chronic Lymphocytic Leukemia B Cells. Lenalidomide 0-12 interleukin 21 Homo sapiens 71-75 27287425-3 2016 Lenalidomide also induced production of interleukin-21 (IL21) and its mRNA in T cells from patients with CLL. Lenalidomide 0-12 interleukin 21 Homo sapiens 40-54 27287425-3 2016 Lenalidomide also induced production of interleukin-21 (IL21) and its mRNA in T cells from patients with CLL. Lenalidomide 0-12 interleukin 21 Homo sapiens 56-60 27287425-4 2016 In addition, lenalidomide enhanced upregulation of functional IL21 receptor (IL21R) on the cell surface and increased receptor mRNA in vitro The in vitro combination of IL21 and lenalidomide enhanced IL21-mediated cytotoxicity toward CLL cells through a variety of mechanisms. Lenalidomide 13-25 interleukin 21 receptor Homo sapiens 62-75 27287425-4 2016 In addition, lenalidomide enhanced upregulation of functional IL21 receptor (IL21R) on the cell surface and increased receptor mRNA in vitro The in vitro combination of IL21 and lenalidomide enhanced IL21-mediated cytotoxicity toward CLL cells through a variety of mechanisms. Lenalidomide 13-25 interleukin 21 receptor Homo sapiens 77-82 27287425-4 2016 In addition, lenalidomide enhanced upregulation of functional IL21 receptor (IL21R) on the cell surface and increased receptor mRNA in vitro The in vitro combination of IL21 and lenalidomide enhanced IL21-mediated cytotoxicity toward CLL cells through a variety of mechanisms. Lenalidomide 13-25 interleukin 21 Homo sapiens 62-66 27287425-4 2016 In addition, lenalidomide enhanced upregulation of functional IL21 receptor (IL21R) on the cell surface and increased receptor mRNA in vitro The in vitro combination of IL21 and lenalidomide enhanced IL21-mediated cytotoxicity toward CLL cells through a variety of mechanisms. Lenalidomide 13-25 interleukin 21 Homo sapiens 77-81 27287425-4 2016 In addition, lenalidomide enhanced upregulation of functional IL21 receptor (IL21R) on the cell surface and increased receptor mRNA in vitro The in vitro combination of IL21 and lenalidomide enhanced IL21-mediated cytotoxicity toward CLL cells through a variety of mechanisms. Lenalidomide 178-190 interleukin 21 receptor Homo sapiens 62-75 27287425-4 2016 In addition, lenalidomide enhanced upregulation of functional IL21 receptor (IL21R) on the cell surface and increased receptor mRNA in vitro The in vitro combination of IL21 and lenalidomide enhanced IL21-mediated cytotoxicity toward CLL cells through a variety of mechanisms. Lenalidomide 178-190 interleukin 21 receptor Homo sapiens 77-82 27287425-4 2016 In addition, lenalidomide enhanced upregulation of functional IL21 receptor (IL21R) on the cell surface and increased receptor mRNA in vitro The in vitro combination of IL21 and lenalidomide enhanced IL21-mediated cytotoxicity toward CLL cells through a variety of mechanisms. Lenalidomide 178-190 interleukin 21 Homo sapiens 62-66 27287425-4 2016 In addition, lenalidomide enhanced upregulation of functional IL21 receptor (IL21R) on the cell surface and increased receptor mRNA in vitro The in vitro combination of IL21 and lenalidomide enhanced IL21-mediated cytotoxicity toward CLL cells through a variety of mechanisms. Lenalidomide 178-190 interleukin 21 Homo sapiens 77-81 27287425-6 2016 Collectively, we demonstrated an immune cell-tumor cell interaction through lenalidomide-mediated induction of IL21 and IL21R, with enhanced IL21-mediated cytotoxicity, which provides justification for this combination in clinical trials for patients with CLL. Lenalidomide 76-88 interleukin 21 Homo sapiens 111-115 27287425-6 2016 Collectively, we demonstrated an immune cell-tumor cell interaction through lenalidomide-mediated induction of IL21 and IL21R, with enhanced IL21-mediated cytotoxicity, which provides justification for this combination in clinical trials for patients with CLL. Lenalidomide 76-88 interleukin 21 receptor Homo sapiens 120-125 27287425-6 2016 Collectively, we demonstrated an immune cell-tumor cell interaction through lenalidomide-mediated induction of IL21 and IL21R, with enhanced IL21-mediated cytotoxicity, which provides justification for this combination in clinical trials for patients with CLL. Lenalidomide 76-88 interleukin 21 Homo sapiens 120-124 27089170-0 2016 ROBUST: Lenalidomide-R-CHOP versus placebo-R-CHOP in previously untreated ABC-type diffuse large B-cell lymphoma. Lenalidomide 8-20 ATP binding cassette subfamily B member 6 (Langereis blood group) Homo sapiens 74-77 26991062-3 2016 Lenalidomide increased von Willebrand factor (VWF), lowered VWF clearance and resolved bleeding. Lenalidomide 0-12 von Willebrand factor Homo sapiens 23-44 27224912-5 2016 Based on in vitro cell line-based experiments, overexpression of CARD11 mutants were demonstrated to confer resistance to the BCR-inhibitor ibrutinib and NF-kappaB-inhibitor lenalidomide. Lenalidomide 174-186 caspase recruitment domain family member 11 Homo sapiens 65-71 27197154-0 2016 Lenalidomide Stabilizes the Erythropoietin Receptor by Inhibiting the E3 Ubiquitin Ligase RNF41. Lenalidomide 0-12 ring finger protein 41 Homo sapiens 90-95 27197154-2 2016 To determine the mechanism by which lenalidomide promotes erythropoiesis, we investigated its action on erythropoietin receptor (EpoR) cellular dynamics. Lenalidomide 36-48 erythropoietin receptor Homo sapiens 129-133 27197154-3 2016 Lenalidomide upregulated expression and stability of JAK2-associated EpoR in UT7 erythroid cells and primary CD71+ erythroid progenitors. Lenalidomide 0-12 erythropoietin receptor Homo sapiens 69-73 27197154-5 2016 To elucidate this mechanism, we investigated the effects of lenalidomide on the E3 ubiquitin ligase RNF41. Lenalidomide 60-72 ring finger protein 41 Homo sapiens 100-105 27197154-6 2016 Lenalidomide promoted EpoR/RNF41 association and inhibited RNF41 auto-ubiquitination, accompanied by a reduction in EpoR ubiquitination. Lenalidomide 0-12 erythropoietin receptor Homo sapiens 22-26 27197154-6 2016 Lenalidomide promoted EpoR/RNF41 association and inhibited RNF41 auto-ubiquitination, accompanied by a reduction in EpoR ubiquitination. Lenalidomide 0-12 ring finger protein 41 Homo sapiens 27-32 27197154-6 2016 Lenalidomide promoted EpoR/RNF41 association and inhibited RNF41 auto-ubiquitination, accompanied by a reduction in EpoR ubiquitination. Lenalidomide 0-12 ring finger protein 41 Homo sapiens 59-64 27197154-6 2016 Lenalidomide promoted EpoR/RNF41 association and inhibited RNF41 auto-ubiquitination, accompanied by a reduction in EpoR ubiquitination. Lenalidomide 0-12 erythropoietin receptor Homo sapiens 116-120 27197154-10 2016 Immunohistochemical staining showed that RNF41 expression decreased in primary erythroid cells of lenalidomide-responding patients, suggesting that cellular RNF41 expression merits investigation as a biomarker for lenalidomide response. Lenalidomide 98-110 ring finger protein 41 Homo sapiens 41-46 27197154-10 2016 Immunohistochemical staining showed that RNF41 expression decreased in primary erythroid cells of lenalidomide-responding patients, suggesting that cellular RNF41 expression merits investigation as a biomarker for lenalidomide response. Lenalidomide 98-110 ring finger protein 41 Homo sapiens 157-162 27197154-10 2016 Immunohistochemical staining showed that RNF41 expression decreased in primary erythroid cells of lenalidomide-responding patients, suggesting that cellular RNF41 expression merits investigation as a biomarker for lenalidomide response. Lenalidomide 214-226 ring finger protein 41 Homo sapiens 41-46 27197154-11 2016 Our findings indicate that lenalidomide has E3 ubiquitin ligase inhibitory effects that extend to RNF41 and that inhibition of RNF41 auto-ubiquitination promotes membrane accumulation of signaling competent JAK2/EpoR complexes that augment Epo responsiveness. Lenalidomide 27-39 ring finger protein 41 Homo sapiens 98-103 27197154-11 2016 Our findings indicate that lenalidomide has E3 ubiquitin ligase inhibitory effects that extend to RNF41 and that inhibition of RNF41 auto-ubiquitination promotes membrane accumulation of signaling competent JAK2/EpoR complexes that augment Epo responsiveness. Lenalidomide 27-39 erythropoietin receptor Homo sapiens 212-216 26991062-16 2016 The biological mechanism(s) through which lenalidomide causes a sustained increase in plasma VWF levels in AVWS independently of paraprotein level requires further study, but is in part modulated through inhibition of enhanced VWF clearance. Lenalidomide 42-54 von Willebrand factor Homo sapiens 93-96 26991062-16 2016 The biological mechanism(s) through which lenalidomide causes a sustained increase in plasma VWF levels in AVWS independently of paraprotein level requires further study, but is in part modulated through inhibition of enhanced VWF clearance. Lenalidomide 42-54 von Willebrand factor Homo sapiens 227-230 27342494-0 2016 [Clinical Curative Efficacy of Lenalidomide Combined with Chemotherapy for Acute Leukemia and Its Impact on VEGF]. Lenalidomide 31-43 vascular endothelial growth factor A Homo sapiens 108-112 27342494-1 2016 OBJECTIVE: To investigate the clinical efficacy of regimen consisting of lenalidomide combined with chemotherapy for acute leukemia and its impact on vascular endothilial growth factor (vEGF) and basic fibroblast growth factor (bFGF), and to analyze the relationship lenalidomide with therapeutic efficacy of leukemia. Lenalidomide 73-85 vascular endothelial growth factor A Homo sapiens 150-184 27342494-1 2016 OBJECTIVE: To investigate the clinical efficacy of regimen consisting of lenalidomide combined with chemotherapy for acute leukemia and its impact on vascular endothilial growth factor (vEGF) and basic fibroblast growth factor (bFGF), and to analyze the relationship lenalidomide with therapeutic efficacy of leukemia. Lenalidomide 73-85 vascular endothelial growth factor A Homo sapiens 186-190 27342494-1 2016 OBJECTIVE: To investigate the clinical efficacy of regimen consisting of lenalidomide combined with chemotherapy for acute leukemia and its impact on vascular endothilial growth factor (vEGF) and basic fibroblast growth factor (bFGF), and to analyze the relationship lenalidomide with therapeutic efficacy of leukemia. Lenalidomide 73-85 fibroblast growth factor 2 Homo sapiens 196-226 27342494-1 2016 OBJECTIVE: To investigate the clinical efficacy of regimen consisting of lenalidomide combined with chemotherapy for acute leukemia and its impact on vascular endothilial growth factor (vEGF) and basic fibroblast growth factor (bFGF), and to analyze the relationship lenalidomide with therapeutic efficacy of leukemia. Lenalidomide 73-85 fibroblast growth factor 2 Homo sapiens 228-232 27342494-8 2016 CONCLUSION: The lenalidomide combined with chemotherapy can significantly decrease the expression level of VEGF and bFGF, and enhance the remission rate of patients with AML. Lenalidomide 16-28 vascular endothelial growth factor A Homo sapiens 107-111 27342494-8 2016 CONCLUSION: The lenalidomide combined with chemotherapy can significantly decrease the expression level of VEGF and bFGF, and enhance the remission rate of patients with AML. Lenalidomide 16-28 fibroblast growth factor 2 Homo sapiens 116-120 27142104-1 2016 BACKGROUND: Immunomodulatory drugs (IMiDs), such as lenalidomide, are therapeutically active compounds that bind and modulate the E3 ubiquitin ligase substrate recruiter cereblon, thereby affect steady-state levels of cereblon and cereblon binding partners, such as ikaros and aiolos, and induce many cellular responses, including cytotoxicity to multiple myeloma (MM) cells. Lenalidomide 52-64 Cbl proto-oncogene like 2 Homo sapiens 130-149 27142104-6 2016 Upon treatment of IMiD-sensitive MM cells with lenalidomide, the steady-state levels of cereblon were significantly increased, whereas levels of AGO2 were significantly decreased. Lenalidomide 47-59 argonaute RISC catalytic component 2 Homo sapiens 145-149 27232930-0 2016 Increased circulating VCAM-1 correlates with advanced disease and poor survival in patients with multiple myeloma: reduction by post-bortezomib and lenalidomide treatment. Lenalidomide 148-160 vascular cell adhesion molecule 1 Homo sapiens 22-28 26991062-3 2016 Lenalidomide increased von Willebrand factor (VWF), lowered VWF clearance and resolved bleeding. Lenalidomide 0-12 von Willebrand factor Homo sapiens 46-49 26991062-3 2016 Lenalidomide increased von Willebrand factor (VWF), lowered VWF clearance and resolved bleeding. Lenalidomide 0-12 von Willebrand factor Homo sapiens 60-63 26991062-7 2016 Results In both patients, lenalidomide treatment resulted in significant clinical improvement, and marked increases in plasma VWF antigen (VWF:Ag) and VWF ristocetin cofactor levels. Lenalidomide 26-38 von Willebrand factor Homo sapiens 126-129 26991062-7 2016 Results In both patients, lenalidomide treatment resulted in significant clinical improvement, and marked increases in plasma VWF antigen (VWF:Ag) and VWF ristocetin cofactor levels. Lenalidomide 26-38 von Willebrand factor Homo sapiens 139-142 26991062-7 2016 Results In both patients, lenalidomide treatment resulted in significant clinical improvement, and marked increases in plasma VWF antigen (VWF:Ag) and VWF ristocetin cofactor levels. Lenalidomide 26-38 von Willebrand factor Homo sapiens 139-142 26991062-9 2016 Furthermore, in one patient, plasma VWF levels remain normal for at least 14 months following discontinuation of lenalidomide treatment. Lenalidomide 113-125 von Willebrand factor Homo sapiens 36-39 26991062-12 2016 Importantly, lenalidomide treatment resulted in normalization of VWFpp/VWF:Ag ratios in both patients. Lenalidomide 13-25 von Willebrand factor Homo sapiens 65-68 26991062-13 2016 These novel data suggest that lenalidomide functions to attenuate enhanced VWF clearance in AVWS. Lenalidomide 30-42 von Willebrand factor Homo sapiens 75-78 26991062-14 2016 Interestingly, in a patient with MGUS, lenalidomide treatment was associated with a significant increase in plasma VWF levels, despite no major change in paraprotein level. Lenalidomide 39-51 von Willebrand factor Homo sapiens 115-118 27009084-7 2016 NVP-Bez235 combined with lenalidomide remarkably increased apoptosis through intrinsic pathway by upregulating Bim, Bax and downregulating Bcl-xL. Lenalidomide 25-37 BCL2-associated X protein Mus musculus 116-119 26119939-6 2016 Although microarray examination of PEL cells treated with lenalidomide revealed activation of interferon (IFN) signaling, blocking the IFN pathway did not block the anti-PEL activity of IMiDs. Lenalidomide 58-70 interferon alpha 1 Homo sapiens 94-104 26119939-6 2016 Although microarray examination of PEL cells treated with lenalidomide revealed activation of interferon (IFN) signaling, blocking the IFN pathway did not block the anti-PEL activity of IMiDs. Lenalidomide 58-70 interferon alpha 1 Homo sapiens 106-109 26119939-12 2016 Lenalidomide displayed synergistic cytotoxicity with several structurally distinct BRD4 inhibitors (JQ-1, IBET151 and PFI-1). Lenalidomide 0-12 bromodomain containing 4 Homo sapiens 83-87 27009084-7 2016 NVP-Bez235 combined with lenalidomide remarkably increased apoptosis through intrinsic pathway by upregulating Bim, Bax and downregulating Bcl-xL. Lenalidomide 25-37 BCL2-like 1 Mus musculus 139-145 27009084-11 2016 CONCLUSIONS: Our findings demonstrated the synergistic effect of low dose of NVP-Bez235 and lenalidomide in ABC-DLBCL, the underlying mechanism may be multifunctional, involving apoptosis, Akt and NF-kappaB inactivation and cell cycle arrest. Lenalidomide 92-104 thymoma viral proto-oncogene 1 Mus musculus 189-192 26437056-8 2016 Although CY-based mobilization was more effective, G-CSF alone was successful in a great majority of patients to reach the defined collection target after three cycles of lenalidomide-based induction. Lenalidomide 171-183 colony stimulating factor 3 Homo sapiens 51-56 26967821-7 2016 This analysis indicates that lenalidomide is active in patients with relapsed/refractory CLL with unfavorable genetic profiles, including TP53 inactivation or unmutated IGHV. Lenalidomide 29-41 tumor protein p53 Homo sapiens 138-142 26967821-7 2016 This analysis indicates that lenalidomide is active in patients with relapsed/refractory CLL with unfavorable genetic profiles, including TP53 inactivation or unmutated IGHV. Lenalidomide 29-41 immunoglobulin heavy variable 3-69-1 (pseudogene) Homo sapiens 169-173 26914976-4 2016 Typical hallmarks of IMiD compound activity, including IKZF3 (Aiolos) degradation, and the downregulation of interferon regulatory factor (IRF) 4 and MYC, seen in lenalidomide-sensitive H929 MM cell lines, were also observed in PomDex-treated lenalidomide-resistant H929 MM cells. Lenalidomide 163-175 interferon regulatory factor 4 Homo sapiens 109-145 26914976-4 2016 Typical hallmarks of IMiD compound activity, including IKZF3 (Aiolos) degradation, and the downregulation of interferon regulatory factor (IRF) 4 and MYC, seen in lenalidomide-sensitive H929 MM cell lines, were also observed in PomDex-treated lenalidomide-resistant H929 MM cells. Lenalidomide 163-175 MYC proto-oncogene, bHLH transcription factor Homo sapiens 150-153 26700310-0 2016 Potential crosstalk of the interleukin-6-heme oxygenase-1-dependent mechanism involved in resistance to lenalidomide in multiple myeloma cells. Lenalidomide 104-116 heme oxygenase 1 Homo sapiens 41-57 26700310-5 2016 Culture of U266, RPMI8226 and CD138(+) cells with exogenous IL-6 in vitro induced high HO-1 expression levels and allowed them to resist lenalidomide. Lenalidomide 137-149 interleukin 6 Homo sapiens 60-64 26700310-9 2016 Reduced HO-1 expression sensitized MM cells to lenalidomide. Lenalidomide 47-59 heme oxygenase 1 Homo sapiens 8-12 26700310-10 2016 Therefore, we postulated that IL-6 in the bone marrow microenvironment of MM patients stimulated high HO-1 expression in MM cells and their resistance to lenalidomide. Lenalidomide 154-166 interleukin 6 Homo sapiens 30-34 26659917-0 2016 The anti-tumoral effect of lenalidomide is increased in vivo by hypoxia-inducible factor (HIF)-1alpha inhibition in myeloma cells. Lenalidomide 27-39 hypoxia inducible factor 1 subunit alpha Homo sapiens 64-101 26657848-10 2016 In this study, we found that low-dose lenalidomide and pomalidomide up-regulate PU.1 expression through inducing demethylation of the PU.1 promoter. Lenalidomide 38-50 Spi-1 proto-oncogene Homo sapiens 80-84 27358895-13 2016 Lenalidomide is approved for treatment of anemia in low and intermediate risk MDS with del (5q), however based on a search of literature it seems that RUNX1 mutations are also more prominent in patients who have responded to Lenalidomide therapy. Lenalidomide 0-12 RUNX family transcription factor 1 Homo sapiens 151-156 27358895-13 2016 Lenalidomide is approved for treatment of anemia in low and intermediate risk MDS with del (5q), however based on a search of literature it seems that RUNX1 mutations are also more prominent in patients who have responded to Lenalidomide therapy. Lenalidomide 225-237 RUNX family transcription factor 1 Homo sapiens 151-156 27467960-7 2016 Furthermore, we found on T-cells from patients treated with lenalidomide a decreased CD45RA expression, indicating a maturated immunophenotype and a decreased expression of CD57, indicating functional T cells. Lenalidomide 60-72 beta-1,3-glucuronyltransferase 1 Homo sapiens 173-177 27467960-9 2016 Complementary to the results in vivo, lenalidomide decreased CD45RA expression on T cells in vitro. Lenalidomide 38-50 protein tyrosine phosphatase receptor type C Homo sapiens 61-65 26657848-10 2016 In this study, we found that low-dose lenalidomide and pomalidomide up-regulate PU.1 expression through inducing demethylation of the PU.1 promoter. Lenalidomide 38-50 Spi-1 proto-oncogene Homo sapiens 134-138 26504152-4 2016 Data discussed here from the pivotal phase 2 (MDS-003) and phase 3 (MDS-004) studies of lenalidomide demonstrate that lenalidomide treatment was associated with both short- and long-term benefits. Lenalidomide 88-100 proteasome assembly chaperone 2 Homo sapiens 46-53 26504152-4 2016 Data discussed here from the pivotal phase 2 (MDS-003) and phase 3 (MDS-004) studies of lenalidomide demonstrate that lenalidomide treatment was associated with both short- and long-term benefits. Lenalidomide 118-130 proteasome assembly chaperone 2 Homo sapiens 46-53 26231201-6 2015 Treatment of human myeloma cell lines such as MM1.S, OPM2, and U266 with thalidomide (100 muM) and its structural analog lenalidomide (10 muM) results in stabilization of CRBN and elevation of CRBN protein levels. Lenalidomide 121-133 latexin Homo sapiens 138-141 27980307-4 2016 Furthermore, some targeting drugs, such as lenalidomide, bortezomib and ibrutinib, directly or indirectly affect CD20 protein expression. Lenalidomide 43-55 keratin 20 Homo sapiens 113-117 26231201-6 2015 Treatment of human myeloma cell lines such as MM1.S, OPM2, and U266 with thalidomide (100 muM) and its structural analog lenalidomide (10 muM) results in stabilization of CRBN and elevation of CRBN protein levels. Lenalidomide 121-133 cereblon Homo sapiens 171-175 26231201-6 2015 Treatment of human myeloma cell lines such as MM1.S, OPM2, and U266 with thalidomide (100 muM) and its structural analog lenalidomide (10 muM) results in stabilization of CRBN and elevation of CRBN protein levels. Lenalidomide 121-133 cereblon Homo sapiens 193-197 26521987-1 2015 Recent studies have suggested that cereblon (CRBN) is essential for the anti-myeloma (MM) activity of immunomodulatory drugs (IMiDs), such as thalidomide and lenalidomide, and that dysregulation of Wnt/beta-catenin pathway may be one of possible reasons of lenalidomide resistance. Lenalidomide 158-170 cereblon Homo sapiens 45-49 26521987-1 2015 Recent studies have suggested that cereblon (CRBN) is essential for the anti-myeloma (MM) activity of immunomodulatory drugs (IMiDs), such as thalidomide and lenalidomide, and that dysregulation of Wnt/beta-catenin pathway may be one of possible reasons of lenalidomide resistance. Lenalidomide 257-269 cereblon Homo sapiens 45-49 26521987-8 2015 However, the occurrence of neutropenia during lenalidomide therapy was more frequent among the CTNNB1 (rs4135385) AA carriers (p=0.019), while the CTNNB1 (rs4533622) AA homozygosity characterized patients with high grade (3-4) neutropenia (p=0.044). Lenalidomide 46-58 catenin beta 1 Homo sapiens 95-101 26438514-1 2015 Lenalidomide acts by a novel drug mechanism-modulation of the substrate specificity of the CRL4(CRBN) E3 ubiquitin ligase. Lenalidomide 0-12 interleukin 17 receptor B Homo sapiens 91-95 26438514-1 2015 Lenalidomide acts by a novel drug mechanism-modulation of the substrate specificity of the CRL4(CRBN) E3 ubiquitin ligase. Lenalidomide 0-12 cereblon Homo sapiens 96-100 26438514-1 2015 Lenalidomide acts by a novel drug mechanism-modulation of the substrate specificity of the CRL4(CRBN) E3 ubiquitin ligase. Lenalidomide 0-12 Cbl proto-oncogene like 2 Homo sapiens 102-121 26438514-2 2015 In multiple myeloma, lenalidomide induces the ubiquitination of IKZF1 and IKZF3 by CRL4(CRBN). Lenalidomide 21-33 IKAROS family zinc finger 1 Homo sapiens 64-69 26438514-2 2015 In multiple myeloma, lenalidomide induces the ubiquitination of IKZF1 and IKZF3 by CRL4(CRBN). Lenalidomide 21-33 IKAROS family zinc finger 3 Homo sapiens 74-79 26438514-2 2015 In multiple myeloma, lenalidomide induces the ubiquitination of IKZF1 and IKZF3 by CRL4(CRBN). Lenalidomide 21-33 interleukin 17 receptor B Homo sapiens 83-87 26438514-2 2015 In multiple myeloma, lenalidomide induces the ubiquitination of IKZF1 and IKZF3 by CRL4(CRBN). Lenalidomide 21-33 cereblon Homo sapiens 88-92 26447613-0 2015 CD4+ T cells play a crucial role for lenalidomide in vivo anti-tumor activity in murine multiple myeloma. Lenalidomide 37-49 CD4 antigen Mus musculus 0-3 26447613-4 2015 Depletion of CD4+ T cells, but not NK cells, B cells, or CD8+ T cells, deprived lenalidomide of its therapeutic effects on 5TGM1-bearing immunocompetent mice. Lenalidomide 80-92 CD4 antigen Mus musculus 13-16 26447613-5 2015 Lenalidomide significantly increased the numbers of IFN-gamma-secreting CD4+ and CD8+ T cells but had no effects on NK cells and B cells in this mouse model. Lenalidomide 0-12 interferon gamma Mus musculus 52-61 26447613-5 2015 Lenalidomide significantly increased the numbers of IFN-gamma-secreting CD4+ and CD8+ T cells but had no effects on NK cells and B cells in this mouse model. Lenalidomide 0-12 CD4 antigen Mus musculus 72-75 26447613-6 2015 Lenalidomide slightly decreased the number of CD25+Foxp3+ T cells but increased perforin expression in CD8+ T cells in vivo. Lenalidomide 0-12 interleukin 2 receptor, alpha chain Mus musculus 46-50 26447613-8 2015 The CD4+ T cell subset may play a critical role in the lenalidomide-mediated anti-myeloma immune response in vivo. Lenalidomide 55-67 CD4 antigen Mus musculus 4-7 26535672-5 2015 We confirmed that CRBN may be a target of lenalidomide. Lenalidomide 42-54 cereblon Homo sapiens 18-22 26341959-3 2015 8226/AMY1 showed a survival advantage over mock control when treated with dexamethasone, bortezomib, and lenalidomide in vitro partly through inhibition of apoptosis induced by these reagents. Lenalidomide 105-117 amylase alpha 1A Homo sapiens 5-9 25979485-10 2015 Finally, lenalidomide with PD-1/PD-L1 blockade inhibits MDSC-mediated immune suppression. Lenalidomide 9-21 programmed cell death 1 Homo sapiens 27-31 25979485-10 2015 Finally, lenalidomide with PD-1/PD-L1 blockade inhibits MDSC-mediated immune suppression. Lenalidomide 9-21 CD274 molecule Homo sapiens 32-37 25979485-11 2015 CONCLUSIONS: Our data therefore demonstrate that checkpoint signaling plays an important role in providing the tumor-promoting, immune-suppressive microenvironment in multiple myeloma, and that PD-1/PD-L1 blockade induces anti-multiple myeloma immune response that can be enhanced by lenalidomide, providing the framework for clinical evaluation of combination therapy. Lenalidomide 284-296 programmed cell death 1 Homo sapiens 194-198 26429859-6 2015 Here, we show that a novel pan-HDACi AR-42 downregulates CD44, a glycoprotein that has been associated with lenalidomide and dexamethasone resistance in myeloma both in vitro and in vivo. Lenalidomide 108-120 CD44 molecule (Indian blood group) Homo sapiens 57-61 25979485-11 2015 CONCLUSIONS: Our data therefore demonstrate that checkpoint signaling plays an important role in providing the tumor-promoting, immune-suppressive microenvironment in multiple myeloma, and that PD-1/PD-L1 blockade induces anti-multiple myeloma immune response that can be enhanced by lenalidomide, providing the framework for clinical evaluation of combination therapy. Lenalidomide 284-296 CD274 molecule Homo sapiens 199-204 26429859-9 2015 Thus, our findings shed light on potential novel combinatorial therapeutic approaches modulating CD44 expression, which may help overcome lenalidomide resistance in myeloma patients. Lenalidomide 138-150 CD44 molecule (Indian blood group) Homo sapiens 97-101 26255966-4 2015 Lenalidomide inhibits retinal endothelial cell viability in normal and pathological condition, and inhibits VEGF-induced endothelial cell migration and tube formation in vitro. Lenalidomide 0-12 vascular endothelial growth factor A Homo sapiens 108-112 26131937-2 2015 Here, we demonstrate that lenalidomide induces the ubiquitination of casein kinase 1A1 (CK1alpha) by the E3 ubiquitin ligase CUL4-RBX1-DDB1-CRBN (known as CRL4(CRBN)), resulting in CK1alpha degradation. Lenalidomide 26-38 cereblon Mus musculus 140-144 26430725-0 2015 Rate of CRL4(CRBN) substrate Ikaros and Aiolos degradation underlies differential activity of lenalidomide and pomalidomide in multiple myeloma cells by regulation of c-Myc and IRF4. Lenalidomide 94-106 interleukin 17 receptor B Homo sapiens 8-12 26430725-0 2015 Rate of CRL4(CRBN) substrate Ikaros and Aiolos degradation underlies differential activity of lenalidomide and pomalidomide in multiple myeloma cells by regulation of c-Myc and IRF4. Lenalidomide 94-106 cereblon Homo sapiens 13-17 26430725-0 2015 Rate of CRL4(CRBN) substrate Ikaros and Aiolos degradation underlies differential activity of lenalidomide and pomalidomide in multiple myeloma cells by regulation of c-Myc and IRF4. Lenalidomide 94-106 IKAROS family zinc finger 1 Homo sapiens 29-35 26430725-0 2015 Rate of CRL4(CRBN) substrate Ikaros and Aiolos degradation underlies differential activity of lenalidomide and pomalidomide in multiple myeloma cells by regulation of c-Myc and IRF4. Lenalidomide 94-106 IKAROS family zinc finger 3 Homo sapiens 40-46 26430725-0 2015 Rate of CRL4(CRBN) substrate Ikaros and Aiolos degradation underlies differential activity of lenalidomide and pomalidomide in multiple myeloma cells by regulation of c-Myc and IRF4. Lenalidomide 94-106 MYC proto-oncogene, bHLH transcription factor Homo sapiens 167-172 26430725-0 2015 Rate of CRL4(CRBN) substrate Ikaros and Aiolos degradation underlies differential activity of lenalidomide and pomalidomide in multiple myeloma cells by regulation of c-Myc and IRF4. Lenalidomide 94-106 interferon regulatory factor 4 Homo sapiens 177-181 26430725-1 2015 Recent discoveries suggest that the critical events leading to the anti-proliferative activity of the IMiD immunomodulatory agents lenalidomide and pomalidomide in multiple myeloma (MM) cells are initiated by Cereblon-dependent ubiquitination and proteasomal degradation of substrate proteins Ikaros (IKZF1) and Aiolos (IKZF3). Lenalidomide 131-143 IKAROS family zinc finger 1 Homo sapiens 293-299 26430725-1 2015 Recent discoveries suggest that the critical events leading to the anti-proliferative activity of the IMiD immunomodulatory agents lenalidomide and pomalidomide in multiple myeloma (MM) cells are initiated by Cereblon-dependent ubiquitination and proteasomal degradation of substrate proteins Ikaros (IKZF1) and Aiolos (IKZF3). Lenalidomide 131-143 IKAROS family zinc finger 1 Homo sapiens 301-306 26430725-1 2015 Recent discoveries suggest that the critical events leading to the anti-proliferative activity of the IMiD immunomodulatory agents lenalidomide and pomalidomide in multiple myeloma (MM) cells are initiated by Cereblon-dependent ubiquitination and proteasomal degradation of substrate proteins Ikaros (IKZF1) and Aiolos (IKZF3). Lenalidomide 131-143 IKAROS family zinc finger 3 Homo sapiens 312-318 26430725-1 2015 Recent discoveries suggest that the critical events leading to the anti-proliferative activity of the IMiD immunomodulatory agents lenalidomide and pomalidomide in multiple myeloma (MM) cells are initiated by Cereblon-dependent ubiquitination and proteasomal degradation of substrate proteins Ikaros (IKZF1) and Aiolos (IKZF3). Lenalidomide 131-143 IKAROS family zinc finger 3 Homo sapiens 320-325 26430725-4 2015 In addition, we found that the half-maximal rate, rather than the final extent of Ikaros and Aiolos degradation, correlated to the relative efficacy of growth inhibition by lenalidomide or pomalidomide. Lenalidomide 173-185 IKAROS family zinc finger 1 Homo sapiens 82-88 26430725-4 2015 In addition, we found that the half-maximal rate, rather than the final extent of Ikaros and Aiolos degradation, correlated to the relative efficacy of growth inhibition by lenalidomide or pomalidomide. Lenalidomide 173-185 IKAROS family zinc finger 3 Homo sapiens 93-99 26408680-0 2015 Genes Involved in Maintaining the Bone Marrow Stroma Are Dysregulated in Patients with Myelofibrosis: Lenalidomide Treatment Up-regulates SOCS3. Lenalidomide 102-114 suppressor of cytokine signaling 3 Homo sapiens 138-143 26408680-5 2015 CONCLUSION: Patients with primary MF showed aberrant expression of several genes involved in maintaining BM homeostasis and our findings suggest that treatment with lenalidomide plus prednisone up-regulates SOCS3. Lenalidomide 165-177 suppressor of cytokine signaling 3 Homo sapiens 207-212 26450624-0 2015 Lenalidomide enhances the function of chimeric antigen receptor T cells against the epidermal growth factor receptor variant III by enhancing immune synapses. Lenalidomide 0-12 nuclear receptor subfamily 1, group I, member 3 Mus musculus 38-63 26450624-7 2015 An important and novel finding of our study was that a thalidomide derivative lenalidomide induced 3C10-CAR PBMC proliferation and enhanced the persistent antitumor effect of the cells in vivo. Lenalidomide 78-90 nuclear receptor subfamily 1, group I, member 3 Mus musculus 104-107 26450624-8 2015 Lenalidomide also exhibited enhanced immunological synapses between the effector cells and the target cells as determined by CD11a and F-actin polymerization. Lenalidomide 0-12 integrin alpha L Mus musculus 125-130 26450624-9 2015 Collectively, lentiviral-mediated transduction of CAR effectors targeting the EGFRvIII showed specific efficacy, and lenalidomide even intensified CAR cell therapy by enhanced formation of immunological synapses. Lenalidomide 117-129 nuclear receptor subfamily 1, group I, member 3 Mus musculus 147-150 26002042-0 2015 Selection of resistant acute myeloid leukemia SKM-1 and MOLM-13 cells by vincristine-, mitoxantrone- and lenalidomide-induced upregulation of P-glycoprotein activity and downregulation of CD33 cell surface exposure. Lenalidomide 105-117 sodium voltage-gated channel alpha subunit 4 Homo sapiens 46-51 26002042-0 2015 Selection of resistant acute myeloid leukemia SKM-1 and MOLM-13 cells by vincristine-, mitoxantrone- and lenalidomide-induced upregulation of P-glycoprotein activity and downregulation of CD33 cell surface exposure. Lenalidomide 105-117 ATP binding cassette subfamily B member 1 Homo sapiens 142-156 26002042-0 2015 Selection of resistant acute myeloid leukemia SKM-1 and MOLM-13 cells by vincristine-, mitoxantrone- and lenalidomide-induced upregulation of P-glycoprotein activity and downregulation of CD33 cell surface exposure. Lenalidomide 105-117 CD33 molecule Homo sapiens 188-192 26002042-5 2015 For this purpose, we established drug resistant variants of SKM-1 and MOLM-13 AML cell lines via the selection of parental cells for resistance to vincristine, mitoxantrone and lenalidomide. Lenalidomide 177-189 sodium voltage-gated channel alpha subunit 4 Homo sapiens 60-65 26378933-8 2015 Furthermore, our clinical analysis suggests that including immunomodulatory agents such as Lenalidomide in the treatment regimen may help to overcome this negative effect, providing an alternative consideration in treatment planning of MM patients with del(8)(p21). Lenalidomide 91-103 H3 histone pseudogene 16 Homo sapiens 260-263 25999435-0 2015 A Phase I Trial of the Anti-KIR Antibody IPH2101 and Lenalidomide in Patients with Relapsed/Refractory Multiple Myeloma. Lenalidomide 53-65 killer cell immunoglobulin like receptor, two Ig domains and long cytoplasmic tail 4 Homo sapiens 28-31 26051632-4 2015 Lenalidomide pre-treatment of MM cell lines reduced TReg generation and the concomitant TReg:TEff (CD4(+)CD25(+)FoxP3(-): effector T cells) ratio, as a consequence of reduced ICOSL transcription. Lenalidomide 0-12 inducible T cell costimulator ligand Homo sapiens 175-180 26169489-4 2015 The responses to lenalidomide seem to be inversely correlated to the pre-treatment EPO level. Lenalidomide 17-29 erythropoietin Homo sapiens 83-86 26195701-4 2015 Tumoricidal effects of lenalidomide are associated with reduced interferon regulatory factor 4, a downstream target of cereblon. Lenalidomide 23-35 interferon regulatory factor 4 Homo sapiens 64-94 26002965-1 2015 Cereblon (CRBN), a substrate receptor of the Cullin 4 RING E3 ubiquitin ligase complex, is the target of the immunomodulatory drugs lenalidomide and pomalidomide. Lenalidomide 132-144 cereblon Homo sapiens 10-14 26002964-3 2015 Here, we report that lenalidomide lowers the threshold for NK-cell activation, causing a 66% decrease in the 50% effective concentration (EC50) for activation through CD16, and a 38% decrease in EC50 for NK group 2 member D (NKG2D)-mediated activation, allowing NK cells to respond to lower doses of ligand. Lenalidomide 21-33 Fc gamma receptor IIIa Homo sapiens 167-171 26002964-3 2015 Here, we report that lenalidomide lowers the threshold for NK-cell activation, causing a 66% decrease in the 50% effective concentration (EC50) for activation through CD16, and a 38% decrease in EC50 for NK group 2 member D (NKG2D)-mediated activation, allowing NK cells to respond to lower doses of ligand. Lenalidomide 21-33 killer cell lectin like receptor K1 Homo sapiens 204-223 26535672-0 2015 Lenalidomide affect expression level of cereblon protein in multiple myeloma cell line RPMI8226. Lenalidomide 0-12 cereblon Homo sapiens 40-48 26535672-1 2015 We investigated the mechanisms of action of immuno-modulatory drug (lenalidomide) on the protein expression of cereblon (CRBN) and their therapeutic targets in the multiple myeloma cell line RPMI8226. Lenalidomide 68-80 cereblon Homo sapiens 111-119 26535672-1 2015 We investigated the mechanisms of action of immuno-modulatory drug (lenalidomide) on the protein expression of cereblon (CRBN) and their therapeutic targets in the multiple myeloma cell line RPMI8226. Lenalidomide 68-80 cereblon Homo sapiens 121-125 26535672-4 2015 However, the protein expression of CRBN decreased signifi-cantly after treatment with lenalidomide, while bortezomib had no effect on the expression of CRBN. Lenalidomide 86-98 cereblon Homo sapiens 35-39 25963624-0 2015 Effectiveness of lenalidomide in a patient with refractory anemia with ring sideroblasts and thrombocytosis with JAK2 (V617F) mutation. Lenalidomide 17-29 Janus kinase 2 Homo sapiens 113-117 26345573-0 2015 [Successful treatment of venous thromboembolism with a Factor Xa inhibitor, edoxaban, in patients with lenalidomide-treated multiple myeloma]. Lenalidomide 103-115 coagulation factor X Homo sapiens 55-64 26002964-3 2015 Here, we report that lenalidomide lowers the threshold for NK-cell activation, causing a 66% decrease in the 50% effective concentration (EC50) for activation through CD16, and a 38% decrease in EC50 for NK group 2 member D (NKG2D)-mediated activation, allowing NK cells to respond to lower doses of ligand. Lenalidomide 21-33 killer cell lectin like receptor K1 Homo sapiens 225-230 26002964-4 2015 In addition, lenalidomide augments NK-cell responses, causing a twofold increase in the proportion of primary NK cells producing interferon-gamma (IFN-gamma), and a 20-fold increase in the amount of IFN-gamma produced per cell. Lenalidomide 13-25 interferon gamma Homo sapiens 129-145 26002964-4 2015 In addition, lenalidomide augments NK-cell responses, causing a twofold increase in the proportion of primary NK cells producing interferon-gamma (IFN-gamma), and a 20-fold increase in the amount of IFN-gamma produced per cell. Lenalidomide 13-25 interferon gamma Homo sapiens 147-156 26002964-4 2015 In addition, lenalidomide augments NK-cell responses, causing a twofold increase in the proportion of primary NK cells producing interferon-gamma (IFN-gamma), and a 20-fold increase in the amount of IFN-gamma produced per cell. Lenalidomide 13-25 interferon gamma Homo sapiens 199-208 26002964-7 2015 Of particular clinical importance, lenalidomide also allowed NK cells to be activated by lower doses of rituximab, an anti-CD20 monoclonal antibody (mAb) widely used to treat B-cell malignancies. Lenalidomide 35-47 keratin 20 Homo sapiens 123-127 26002964-9 2015 Finally, superresolution microscopy revealed that lenalidomide increased the periodicity of cortical actin at immune synapses, resulting in an increase in the area of the actin mesh predicted to be penetrable to vesicles containing IFN-gamma. Lenalidomide 50-62 interferon gamma Homo sapiens 232-241 26131937-2 2015 Here, we demonstrate that lenalidomide induces the ubiquitination of casein kinase 1A1 (CK1alpha) by the E3 ubiquitin ligase CUL4-RBX1-DDB1-CRBN (known as CRL4(CRBN)), resulting in CK1alpha degradation. Lenalidomide 26-38 cereblon Mus musculus 160-164 26131937-4 2015 We found that mouse cells are resistant to lenalidomide but that changing a single amino acid in mouse Crbn to the corresponding human residue enables lenalidomide-dependent degradation of CK1alpha. Lenalidomide 43-55 cereblon Mus musculus 103-107 26131937-4 2015 We found that mouse cells are resistant to lenalidomide but that changing a single amino acid in mouse Crbn to the corresponding human residue enables lenalidomide-dependent degradation of CK1alpha. Lenalidomide 151-163 cereblon Mus musculus 103-107 26273440-0 2015 A case of myelodysplastic syndrome/myeloproliferative neoplasm (MDS/MPN) successfully treated with lenalidomide. Lenalidomide 99-111 serine protease 27 Homo sapiens 68-71 26273440-4 2015 We report a patient with MDS/MPN who responded well to lenalidomide therapy. Lenalidomide 55-67 serine protease 27 Homo sapiens 29-32 25915157-7 2015 Moreover, clarithromycin overcame stromal-mediated MM resistance to dexamethasone, suggesting that CYP3A4 inhibition plays a role in its ability to augment the activity of lenalidomide and dexamethasone as part of the BiRd regimen. Lenalidomide 172-184 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 99-105 25284710-0 2015 High level of full-length cereblon mRNA in lower risk myelodysplastic syndrome with isolated 5q deletion is implicated in the efficacy of lenalidomide. Lenalidomide 138-150 cereblon Homo sapiens 26-34 25284710-1 2015 Downregulation of cereblon (CRBN) gene expression is associated with resistance to the immunomodulatory drug lenalidomide and poor survival outcomes in multiple myeloma (MM) patients. Lenalidomide 109-121 cereblon Homo sapiens 18-26 25284710-1 2015 Downregulation of cereblon (CRBN) gene expression is associated with resistance to the immunomodulatory drug lenalidomide and poor survival outcomes in multiple myeloma (MM) patients. Lenalidomide 109-121 cereblon Homo sapiens 28-32 25284710-6 2015 High levels of CRBN mRNA were detected in all lenalidomide responders during the course of therapy. Lenalidomide 46-58 cereblon Homo sapiens 15-19 25284710-7 2015 A significant decrease of the CRBN mRNA level during lenalidomide treatment is associated with loss of response to treatment and disease progression. Lenalidomide 53-65 cereblon Homo sapiens 30-34 25284710-8 2015 These results suggest that, similar to the treatment of MM, high levels of full-length CRBN mRNA in lower risk 5q- patients are necessary for the efficacy of lenalidomide. Lenalidomide 158-170 cereblon Homo sapiens 87-91 25287904-3 2015 METHODS: We used miRNA expression microarrays to study changes in miRNA levels in peripheral blood CD14+ monocytes collected from patients before and during lenalidomide treatment and compared them with those from healthy donors. Lenalidomide 157-169 CD14 molecule Homo sapiens 99-103 25287904-4 2015 RESULTS: Before treatment, we observed strong upregulation of pro-apoptotic miR-34a and miR-34a* that diminished during lenalidomide exposure. Lenalidomide 120-132 microRNA 34a Homo sapiens 76-83 25287904-4 2015 RESULTS: Before treatment, we observed strong upregulation of pro-apoptotic miR-34a and miR-34a* that diminished during lenalidomide exposure. Lenalidomide 120-132 microRNA 34a Homo sapiens 88-95 25398450-0 2015 Preclinical Evidence for the Therapeutic Potential of CD38-Targeted Immuno-Chemotherapy in Multiple Myeloma Patients Refractory to Lenalidomide and Bortezomib. Lenalidomide 131-143 CD38 molecule Homo sapiens 54-58 27668268-0 2015 Lenalidomide Polarizes Th1-specific Anti-tumor Immune Response and Expands XBP1 Antigen-Specific Central Memory CD3+CD8+ T cells against Various Solid Tumors. Lenalidomide 0-12 negative elongation factor complex member C/D Homo sapiens 23-26 27668268-0 2015 Lenalidomide Polarizes Th1-specific Anti-tumor Immune Response and Expands XBP1 Antigen-Specific Central Memory CD3+CD8+ T cells against Various Solid Tumors. Lenalidomide 0-12 X-box binding protein 1 Homo sapiens 75-79 27668268-2 2015 METHODS: XBP1 antigen-specific cytotoxic T lymphocytes (XBP1-CTL) generated using immunogenic heteroclitic XBP1 US184-192 (YISPWILAV) and XBP1 SP367-375 (YLFPQLISV) peptides or various solid tumor cells over-expressing XBP1 target antigen were evaluated, either alone or in combination with lenalidomide, for phenotype and immune functional activity. Lenalidomide 291-303 X-box binding protein 1 Homo sapiens 9-13 27668268-2 2015 METHODS: XBP1 antigen-specific cytotoxic T lymphocytes (XBP1-CTL) generated using immunogenic heteroclitic XBP1 US184-192 (YISPWILAV) and XBP1 SP367-375 (YLFPQLISV) peptides or various solid tumor cells over-expressing XBP1 target antigen were evaluated, either alone or in combination with lenalidomide, for phenotype and immune functional activity. Lenalidomide 291-303 X-box binding protein 1 Homo sapiens 56-60 27668268-2 2015 METHODS: XBP1 antigen-specific cytotoxic T lymphocytes (XBP1-CTL) generated using immunogenic heteroclitic XBP1 US184-192 (YISPWILAV) and XBP1 SP367-375 (YLFPQLISV) peptides or various solid tumor cells over-expressing XBP1 target antigen were evaluated, either alone or in combination with lenalidomide, for phenotype and immune functional activity. Lenalidomide 291-303 X-box binding protein 1 Homo sapiens 56-60 27668268-2 2015 METHODS: XBP1 antigen-specific cytotoxic T lymphocytes (XBP1-CTL) generated using immunogenic heteroclitic XBP1 US184-192 (YISPWILAV) and XBP1 SP367-375 (YLFPQLISV) peptides or various solid tumor cells over-expressing XBP1 target antigen were evaluated, either alone or in combination with lenalidomide, for phenotype and immune functional activity. Lenalidomide 291-303 X-box binding protein 1 Homo sapiens 56-60 27668268-2 2015 METHODS: XBP1 antigen-specific cytotoxic T lymphocytes (XBP1-CTL) generated using immunogenic heteroclitic XBP1 US184-192 (YISPWILAV) and XBP1 SP367-375 (YLFPQLISV) peptides or various solid tumor cells over-expressing XBP1 target antigen were evaluated, either alone or in combination with lenalidomide, for phenotype and immune functional activity. Lenalidomide 291-303 X-box binding protein 1 Homo sapiens 56-60 27668268-3 2015 RESULTS: Lenalidomide treatment of XBP1-CTL increased the proportion of CD45RO+ memory CD3+CD8+ T cells, but not the total CD3+CD8+ T cells. Lenalidomide 9-21 X-box binding protein 1 Homo sapiens 35-39 27668268-4 2015 Lenalidomide upregulated critical T cell activation markers and costimulatory molecules (CD28, CD38, CD40L, CD69, ICOS), especially within the central memory CTL subset of XBP1-CTL, while decreasing TCRalphabeta and T cell checkpoint blockade (CTLA-4, PD-1). Lenalidomide 0-12 CD28 molecule Homo sapiens 89-93 27668268-4 2015 Lenalidomide upregulated critical T cell activation markers and costimulatory molecules (CD28, CD38, CD40L, CD69, ICOS), especially within the central memory CTL subset of XBP1-CTL, while decreasing TCRalphabeta and T cell checkpoint blockade (CTLA-4, PD-1). Lenalidomide 0-12 CD38 molecule Homo sapiens 95-99 27668268-4 2015 Lenalidomide upregulated critical T cell activation markers and costimulatory molecules (CD28, CD38, CD40L, CD69, ICOS), especially within the central memory CTL subset of XBP1-CTL, while decreasing TCRalphabeta and T cell checkpoint blockade (CTLA-4, PD-1). Lenalidomide 0-12 CD40 ligand Homo sapiens 101-106 27668268-4 2015 Lenalidomide upregulated critical T cell activation markers and costimulatory molecules (CD28, CD38, CD40L, CD69, ICOS), especially within the central memory CTL subset of XBP1-CTL, while decreasing TCRalphabeta and T cell checkpoint blockade (CTLA-4, PD-1). Lenalidomide 0-12 CD69 molecule Homo sapiens 108-112 27668268-4 2015 Lenalidomide upregulated critical T cell activation markers and costimulatory molecules (CD28, CD38, CD40L, CD69, ICOS), especially within the central memory CTL subset of XBP1-CTL, while decreasing TCRalphabeta and T cell checkpoint blockade (CTLA-4, PD-1). Lenalidomide 0-12 inducible T cell costimulator Homo sapiens 114-118 27668268-4 2015 Lenalidomide upregulated critical T cell activation markers and costimulatory molecules (CD28, CD38, CD40L, CD69, ICOS), especially within the central memory CTL subset of XBP1-CTL, while decreasing TCRalphabeta and T cell checkpoint blockade (CTLA-4, PD-1). Lenalidomide 0-12 X-box binding protein 1 Homo sapiens 172-176 27668268-4 2015 Lenalidomide upregulated critical T cell activation markers and costimulatory molecules (CD28, CD38, CD40L, CD69, ICOS), especially within the central memory CTL subset of XBP1-CTL, while decreasing TCRalphabeta and T cell checkpoint blockade (CTLA-4, PD-1). Lenalidomide 0-12 cytotoxic T-lymphocyte associated protein 4 Homo sapiens 244-250 27668268-5 2015 Lenalidomide increased the anti-tumor activities of XBP1-CTL memory subsets, which were associated with expression of Th1 transcriptional regulators (T-bet, Eomes) and Akt activation, thereby resulting in enhanced IFN-gamma production, granzyme B upregulation and specific CD28/CD38-positive and CTLA-4/PD-1-negative cell proliferation. Lenalidomide 0-12 X-box binding protein 1 Homo sapiens 52-56 27668268-5 2015 Lenalidomide increased the anti-tumor activities of XBP1-CTL memory subsets, which were associated with expression of Th1 transcriptional regulators (T-bet, Eomes) and Akt activation, thereby resulting in enhanced IFN-gamma production, granzyme B upregulation and specific CD28/CD38-positive and CTLA-4/PD-1-negative cell proliferation. Lenalidomide 0-12 negative elongation factor complex member C/D Homo sapiens 118-121 27668268-5 2015 Lenalidomide increased the anti-tumor activities of XBP1-CTL memory subsets, which were associated with expression of Th1 transcriptional regulators (T-bet, Eomes) and Akt activation, thereby resulting in enhanced IFN-gamma production, granzyme B upregulation and specific CD28/CD38-positive and CTLA-4/PD-1-negative cell proliferation. Lenalidomide 0-12 eomesodermin Homo sapiens 157-162 27668268-5 2015 Lenalidomide increased the anti-tumor activities of XBP1-CTL memory subsets, which were associated with expression of Th1 transcriptional regulators (T-bet, Eomes) and Akt activation, thereby resulting in enhanced IFN-gamma production, granzyme B upregulation and specific CD28/CD38-positive and CTLA-4/PD-1-negative cell proliferation. Lenalidomide 0-12 AKT serine/threonine kinase 1 Homo sapiens 168-171 27668268-5 2015 Lenalidomide increased the anti-tumor activities of XBP1-CTL memory subsets, which were associated with expression of Th1 transcriptional regulators (T-bet, Eomes) and Akt activation, thereby resulting in enhanced IFN-gamma production, granzyme B upregulation and specific CD28/CD38-positive and CTLA-4/PD-1-negative cell proliferation. Lenalidomide 0-12 interferon gamma Homo sapiens 214-223 27668268-5 2015 Lenalidomide increased the anti-tumor activities of XBP1-CTL memory subsets, which were associated with expression of Th1 transcriptional regulators (T-bet, Eomes) and Akt activation, thereby resulting in enhanced IFN-gamma production, granzyme B upregulation and specific CD28/CD38-positive and CTLA-4/PD-1-negative cell proliferation. Lenalidomide 0-12 granzyme B Homo sapiens 236-246 27668268-5 2015 Lenalidomide increased the anti-tumor activities of XBP1-CTL memory subsets, which were associated with expression of Th1 transcriptional regulators (T-bet, Eomes) and Akt activation, thereby resulting in enhanced IFN-gamma production, granzyme B upregulation and specific CD28/CD38-positive and CTLA-4/PD-1-negative cell proliferation. Lenalidomide 0-12 CD28 molecule Homo sapiens 273-277 27668268-5 2015 Lenalidomide increased the anti-tumor activities of XBP1-CTL memory subsets, which were associated with expression of Th1 transcriptional regulators (T-bet, Eomes) and Akt activation, thereby resulting in enhanced IFN-gamma production, granzyme B upregulation and specific CD28/CD38-positive and CTLA-4/PD-1-negative cell proliferation. Lenalidomide 0-12 CD38 molecule Homo sapiens 278-282 27668268-5 2015 Lenalidomide increased the anti-tumor activities of XBP1-CTL memory subsets, which were associated with expression of Th1 transcriptional regulators (T-bet, Eomes) and Akt activation, thereby resulting in enhanced IFN-gamma production, granzyme B upregulation and specific CD28/CD38-positive and CTLA-4/PD-1-negative cell proliferation. Lenalidomide 0-12 cytotoxic T-lymphocyte associated protein 4 Homo sapiens 296-302 27668268-6 2015 CONCLUSIONS: These studies suggest the potential benefit of lenalidomide treatment to boost anti-tumor activities of XBP1-specific CTL against a variety of solid tumors and enhance response to an XBP1-directing cancer vaccine regime. Lenalidomide 60-72 X-box binding protein 1 Homo sapiens 117-121 27668268-6 2015 CONCLUSIONS: These studies suggest the potential benefit of lenalidomide treatment to boost anti-tumor activities of XBP1-specific CTL against a variety of solid tumors and enhance response to an XBP1-directing cancer vaccine regime. Lenalidomide 60-72 X-box binding protein 1 Homo sapiens 196-200 25448491-0 2015 Increased serum tumor necrosis factor alpha levels in patients with lenalidomide-induced hypothyroidism. Lenalidomide 68-80 tumor necrosis factor Homo sapiens 16-43 25978432-1 2015 Immunomodulatory drugs (IMiDs) thalidomide, lenalidomide (Len) and pomalidomide trigger anti-tumor activities in multiple myeloma (MM) by targetting cereblon and thereby impacting IZF1/3, c-Myc and IRF4. Lenalidomide 44-56 MYC proto-oncogene, bHLH transcription factor Homo sapiens 188-193 25978432-1 2015 Immunomodulatory drugs (IMiDs) thalidomide, lenalidomide (Len) and pomalidomide trigger anti-tumor activities in multiple myeloma (MM) by targetting cereblon and thereby impacting IZF1/3, c-Myc and IRF4. Lenalidomide 44-56 interferon regulatory factor 4 Homo sapiens 198-202 25978432-1 2015 Immunomodulatory drugs (IMiDs) thalidomide, lenalidomide (Len) and pomalidomide trigger anti-tumor activities in multiple myeloma (MM) by targetting cereblon and thereby impacting IZF1/3, c-Myc and IRF4. Lenalidomide 58-61 MYC proto-oncogene, bHLH transcription factor Homo sapiens 188-193 25978432-1 2015 Immunomodulatory drugs (IMiDs) thalidomide, lenalidomide (Len) and pomalidomide trigger anti-tumor activities in multiple myeloma (MM) by targetting cereblon and thereby impacting IZF1/3, c-Myc and IRF4. Lenalidomide 58-61 interferon regulatory factor 4 Homo sapiens 198-202 25632047-1 2015 PURPOSE: Lenalidomide, an immunomodulatory agent that enhances antibody-dependent cell-mediated cytotoxicity, has the potential to synergize with rituximab, an anti-CD20 mAb. Lenalidomide 9-21 keratin 20 Homo sapiens 165-169 25643589-9 2015 Maintenance lenalidomide/azacitidine augmented the function of cytotoxic T lymphocytes, particularly in patients with NPM1 mutation. Lenalidomide 12-24 nucleophosmin 1 Homo sapiens 118-122 25643589-10 2015 The lenalidomide/azacitidine maintenance combination was effective in suppressing residual DNA (cytosine-5-)-methyltransferase 3 alpha (DNMT3A)-positive disease, resulting in sustained remission in patients with concurrent NPM1 mutation. Lenalidomide 4-16 DNA methyltransferase 3 alpha Homo sapiens 136-142 25643589-10 2015 The lenalidomide/azacitidine maintenance combination was effective in suppressing residual DNA (cytosine-5-)-methyltransferase 3 alpha (DNMT3A)-positive disease, resulting in sustained remission in patients with concurrent NPM1 mutation. Lenalidomide 4-16 nucleophosmin 1 Homo sapiens 223-227 24738975-0 2015 Intermittent granulocyte colony-stimulating factor for neutropenia management in patients with relapsed or refractory multiple myeloma treated with lenalidomide plus dexamethasone. Lenalidomide 148-160 colony stimulating factor 3 Homo sapiens 13-50 24738975-7 2015 Intermittent G-CSF may be an effective approach for lenalidomide dose-preservation, which may lead to improved outcomes, although it does not prevent infections or thrombocytopenia-related dose limitations. Lenalidomide 52-64 colony stimulating factor 3 Homo sapiens 13-18 25395420-5 2015 RSK2 knockdown induced cytotoxicity across a panel of MM cell lines and consistently increased sensitivity to lenalidomide. Lenalidomide 110-122 ribosomal protein S6 kinase A3 Homo sapiens 0-4 25395420-6 2015 Accordingly, 3 small molecular inhibitors of RSK2 demonstrated synergy with lenalidomide cytotoxicity in MM cells even in the presence of stromal contact. Lenalidomide 76-88 ribosomal protein S6 kinase A3 Homo sapiens 45-49 25395420-7 2015 Both RSK2 knockdown and small molecule inhibition downregulate interferon regulatory factor 4 and MYC, and provides an explanation for the synergy between lenalidomide and RSK2 inhibition. Lenalidomide 155-167 ribosomal protein S6 kinase A3 Homo sapiens 5-9 25395420-7 2015 Both RSK2 knockdown and small molecule inhibition downregulate interferon regulatory factor 4 and MYC, and provides an explanation for the synergy between lenalidomide and RSK2 inhibition. Lenalidomide 155-167 interferon regulatory factor 4 Homo sapiens 63-93 25395420-7 2015 Both RSK2 knockdown and small molecule inhibition downregulate interferon regulatory factor 4 and MYC, and provides an explanation for the synergy between lenalidomide and RSK2 inhibition. Lenalidomide 155-167 MYC proto-oncogene, bHLH transcription factor Homo sapiens 98-101 25987070-5 2015 These lenalidomide based regimens included cisplatin, doxorubicin, or GM-CSF. Lenalidomide 6-18 colony stimulating factor 2 Homo sapiens 70-76 25987070-6 2015 Pooled analysis suggested that, in all patients, the pooled PSA level decline of >=50% was 13.3% (13/98) in lenalidomide based regimens. Lenalidomide 111-123 aminopeptidase puromycin sensitive Homo sapiens 60-63 26075044-9 2015 Recurrent TP53 mutations have been associated with an increased risk of disease evolution and with decreased response to the drug lenalidomide in del(5q) MDS patients. Lenalidomide 130-142 tumor protein p53 Homo sapiens 10-14 25966683-5 2015 Lenalidomide has shown protective effects in an animal model of amyotrophic lateral sclerosis, and its mechanism of action involves modulation of cytokine production and inhibition of NF-kappaB signaling. Lenalidomide 0-12 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 184-193 25966683-9 2015 Central expression of pro-inflammatory cytokines was diminished in lenalidomide-treated transgenic animals, together with reduction in NF-kappaB activation. Lenalidomide 67-79 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 135-144 26688096-8 2015 Serial sampling and response to lenalidomide treatment showed that CSNK1A1 mutations were highly associated with the del(5q) clone. Lenalidomide 32-44 casein kinase 1 alpha 1 Homo sapiens 67-74 26688096-9 2015 Only one patient with a CSNK1A1 mutation showed complete cytogenetic response to lenalidomide. Lenalidomide 81-93 casein kinase 1 alpha 1 Homo sapiens 24-31 26688096-10 2015 Four (57%) of the seven patients carrying a CSNK1A1 mutation showed disease progression coupled with an increase in mutant allele burden (all four were on lenalidomide). Lenalidomide 155-167 casein kinase 1 alpha 1 Homo sapiens 44-51 25398834-6 2015 We found that lenalidomide induces high actin polymerization on CD14(+) monocytes through activation of small GTPases, RhoA, Rac1 and Rap1 that correlated with increased adhesion and impaired monocyte migration in response to CCL2, CCL3 and CXCL12. Lenalidomide 14-26 CD14 molecule Homo sapiens 64-68 25398834-6 2015 We found that lenalidomide induces high actin polymerization on CD14(+) monocytes through activation of small GTPases, RhoA, Rac1 and Rap1 that correlated with increased adhesion and impaired monocyte migration in response to CCL2, CCL3 and CXCL12. Lenalidomide 14-26 ras homolog family member A Homo sapiens 119-123 25398834-6 2015 We found that lenalidomide induces high actin polymerization on CD14(+) monocytes through activation of small GTPases, RhoA, Rac1 and Rap1 that correlated with increased adhesion and impaired monocyte migration in response to CCL2, CCL3 and CXCL12. Lenalidomide 14-26 Rac family small GTPase 1 Homo sapiens 125-129 25398834-6 2015 We found that lenalidomide induces high actin polymerization on CD14(+) monocytes through activation of small GTPases, RhoA, Rac1 and Rap1 that correlated with increased adhesion and impaired monocyte migration in response to CCL2, CCL3 and CXCL12. Lenalidomide 14-26 RAP1A, member of RAS oncogene family Homo sapiens 134-138 25398834-6 2015 We found that lenalidomide induces high actin polymerization on CD14(+) monocytes through activation of small GTPases, RhoA, Rac1 and Rap1 that correlated with increased adhesion and impaired monocyte migration in response to CCL2, CCL3 and CXCL12. Lenalidomide 14-26 C-C motif chemokine ligand 2 Homo sapiens 226-230 25398834-6 2015 We found that lenalidomide induces high actin polymerization on CD14(+) monocytes through activation of small GTPases, RhoA, Rac1 and Rap1 that correlated with increased adhesion and impaired monocyte migration in response to CCL2, CCL3 and CXCL12. Lenalidomide 14-26 C-C motif chemokine ligand 3 Homo sapiens 232-236 25398834-6 2015 We found that lenalidomide induces high actin polymerization on CD14(+) monocytes through activation of small GTPases, RhoA, Rac1 and Rap1 that correlated with increased adhesion and impaired monocyte migration in response to CCL2, CCL3 and CXCL12. Lenalidomide 14-26 C-X-C motif chemokine ligand 12 Homo sapiens 241-247 25398834-8 2015 Gene expression signature, induced by lenalidomide in nurse-like cells, indicated a reduction of pivotal pro-survival signals for chronic lymphocytic leukemia, such as CCL2, IGF1, CXCL12, HGF1, and supported a modulation towards M1 phenotype with high IL2 and low IL10, IL8 and CD163. Lenalidomide 38-50 C-C motif chemokine ligand 2 Homo sapiens 168-172 25398834-8 2015 Gene expression signature, induced by lenalidomide in nurse-like cells, indicated a reduction of pivotal pro-survival signals for chronic lymphocytic leukemia, such as CCL2, IGF1, CXCL12, HGF1, and supported a modulation towards M1 phenotype with high IL2 and low IL10, IL8 and CD163. Lenalidomide 38-50 insulin like growth factor 1 Homo sapiens 174-178 25398834-8 2015 Gene expression signature, induced by lenalidomide in nurse-like cells, indicated a reduction of pivotal pro-survival signals for chronic lymphocytic leukemia, such as CCL2, IGF1, CXCL12, HGF1, and supported a modulation towards M1 phenotype with high IL2 and low IL10, IL8 and CD163. Lenalidomide 38-50 C-X-C motif chemokine ligand 12 Homo sapiens 180-186 25398834-8 2015 Gene expression signature, induced by lenalidomide in nurse-like cells, indicated a reduction of pivotal pro-survival signals for chronic lymphocytic leukemia, such as CCL2, IGF1, CXCL12, HGF1, and supported a modulation towards M1 phenotype with high IL2 and low IL10, IL8 and CD163. Lenalidomide 38-50 interleukin 2 Homo sapiens 252-255 25398834-8 2015 Gene expression signature, induced by lenalidomide in nurse-like cells, indicated a reduction of pivotal pro-survival signals for chronic lymphocytic leukemia, such as CCL2, IGF1, CXCL12, HGF1, and supported a modulation towards M1 phenotype with high IL2 and low IL10, IL8 and CD163. Lenalidomide 38-50 interleukin 10 Homo sapiens 264-268 25398834-8 2015 Gene expression signature, induced by lenalidomide in nurse-like cells, indicated a reduction of pivotal pro-survival signals for chronic lymphocytic leukemia, such as CCL2, IGF1, CXCL12, HGF1, and supported a modulation towards M1 phenotype with high IL2 and low IL10, IL8 and CD163. Lenalidomide 38-50 C-X-C motif chemokine ligand 8 Homo sapiens 270-273 25398834-8 2015 Gene expression signature, induced by lenalidomide in nurse-like cells, indicated a reduction of pivotal pro-survival signals for chronic lymphocytic leukemia, such as CCL2, IGF1, CXCL12, HGF1, and supported a modulation towards M1 phenotype with high IL2 and low IL10, IL8 and CD163. Lenalidomide 38-50 CD163 molecule Homo sapiens 278-283 25617756-9 2015 In addition, TC11 and lenalidomide, another phthalimide derivative, directly bound to nucleophosmin 1 (NPM1), whose role in MM is unknown. Lenalidomide 22-34 nucleophosmin 1 Homo sapiens 86-101 25617756-9 2015 In addition, TC11 and lenalidomide, another phthalimide derivative, directly bound to nucleophosmin 1 (NPM1), whose role in MM is unknown. Lenalidomide 22-34 nucleophosmin 1 Homo sapiens 103-107 25984532-6 2015 Future efforts should aim at targeting the role of NF-kappaB resistance in clinical trials, where novel agents like lenalidomide and proteasome inhibitors with established activity in this perspective will be an important component in combination therapy, along with new monoclonal antibody, BTK-inhibitors, and other novel therapy agents. Lenalidomide 116-128 Bruton tyrosine kinase Homo sapiens 292-295 25287778-8 2015 In co-culture assays, TNF-alpha directly increased NK cell activation and myeloma cell death with elotuzumab or elotuzumab plus lenalidomide, and neutralizing TNF-alpha decreased NK cell activation and myeloma cell death with elotuzumab. Lenalidomide 128-140 tumor necrosis factor Homo sapiens 22-31 25448491-4 2015 We found a significantly higher rate of therapy-induced hypothyroidism in the DLBCL-len group (25.8% vs. 1.3%), and we found a statistically significant increase in serum tumor necrosis factor alpha in patients with lenalidomide-induced hypothyroidism. Lenalidomide 216-228 tumor necrosis factor Homo sapiens 171-198 26763359-0 2015 CD200 Expression on Plasma Cell Myeloma Cells is Associated with the Efficacies of Bortezomib, Lenalidomide and Thalidomide. Lenalidomide 95-107 CD200 molecule Homo sapiens 0-5 24721791-4 2014 Lenalidomide was particularly active in this subgroup of tumors, targeting IRF4 expression and plasmacytic differentiation program, thus overcoming bortezomib resistance. Lenalidomide 0-12 interferon regulatory factor 4 Mus musculus 75-79 25212607-2 2014 EXPERIMENTAL DESIGN: The HGF/cMET pathway was evaluated in ECs from patients with multiple myeloma (multiple myeloma ECs) at diagnosis, at relapse after bortezomib- or lenalidomide-based therapies, or on refractory phase to these drugs; in ECs from patients with MGUS (MGECs); and in those patients from the control group. Lenalidomide 168-180 hepatocyte growth factor Homo sapiens 25-28 25212607-2 2014 EXPERIMENTAL DESIGN: The HGF/cMET pathway was evaluated in ECs from patients with multiple myeloma (multiple myeloma ECs) at diagnosis, at relapse after bortezomib- or lenalidomide-based therapies, or on refractory phase to these drugs; in ECs from patients with MGUS (MGECs); and in those patients from the control group. Lenalidomide 168-180 MET proto-oncogene, receptor tyrosine kinase Homo sapiens 29-33 24813620-9 2014 CONCLUSIONS: These data support the clinical benefits and acceptable safety profile of lenalidomide in transfusion-dependent patients with IPSS-defined Low-/Int-1-risk MDS with isolated del(5q). Lenalidomide 87-99 Wnt family member 1 Homo sapiens 157-162 25547653-0 2015 Lenalidomide differently modulates CD20 antigen surface expression on chronic lymphocytic leukemia B-cells. Lenalidomide 0-12 keratin 20 Homo sapiens 35-39 25626322-3 2015 Lenalidomide stimulates T cells and NK-cell through production of Th1 type cytokines IL-2 and IFN-gamma from CD4+ helper-T cells. Lenalidomide 0-12 interferon gamma Homo sapiens 94-103 25626322-3 2015 Lenalidomide stimulates T cells and NK-cell through production of Th1 type cytokines IL-2 and IFN-gamma from CD4+ helper-T cells. Lenalidomide 0-12 CD4 molecule Homo sapiens 109-112 25469886-0 2014 Lenalidomide induces lipid raft assembly to enhance erythropoietin receptor signaling in myelodysplastic syndrome progenitors. Lenalidomide 0-12 erythropoietin receptor Homo sapiens 52-75 25469886-6 2014 Lenalidomide treatment rapidly induced lipid raft formation accompanied by EpoR recruitment into raft fractions together with STAT5, JAK2, and Lyn kinase. Lenalidomide 0-12 erythropoietin receptor Homo sapiens 75-79 25469886-6 2014 Lenalidomide treatment rapidly induced lipid raft formation accompanied by EpoR recruitment into raft fractions together with STAT5, JAK2, and Lyn kinase. Lenalidomide 0-12 signal transducer and activator of transcription 5A Homo sapiens 126-131 25469886-6 2014 Lenalidomide treatment rapidly induced lipid raft formation accompanied by EpoR recruitment into raft fractions together with STAT5, JAK2, and Lyn kinase. Lenalidomide 0-12 Janus kinase 2 Homo sapiens 133-137 25469886-6 2014 Lenalidomide treatment rapidly induced lipid raft formation accompanied by EpoR recruitment into raft fractions together with STAT5, JAK2, and Lyn kinase. Lenalidomide 0-12 LYN proto-oncogene, Src family tyrosine kinase Homo sapiens 143-146 25469886-8 2014 Lenalidomide treatment prior to Epo stimulation enhanced both JAK2 and STAT5 phosphorylation in UT7 and primary MDS erythroid progenitors, accompanied by increased STAT5 DNA binding in UT7 cells, and increased erythroid colony forming capacity in both UT7 and primary cells. Lenalidomide 0-12 Janus kinase 2 Homo sapiens 62-66 25469886-8 2014 Lenalidomide treatment prior to Epo stimulation enhanced both JAK2 and STAT5 phosphorylation in UT7 and primary MDS erythroid progenitors, accompanied by increased STAT5 DNA binding in UT7 cells, and increased erythroid colony forming capacity in both UT7 and primary cells. Lenalidomide 0-12 signal transducer and activator of transcription 5A Homo sapiens 71-76 25469886-8 2014 Lenalidomide treatment prior to Epo stimulation enhanced both JAK2 and STAT5 phosphorylation in UT7 and primary MDS erythroid progenitors, accompanied by increased STAT5 DNA binding in UT7 cells, and increased erythroid colony forming capacity in both UT7 and primary cells. Lenalidomide 0-12 signal transducer and activator of transcription 5A Homo sapiens 164-169 25550914-7 2014 In a subgroup analysis for therapy regimen, lenalidomide- and bortezomib-based therapies increased the PFS of patients with Amp (1q21) (HR=1.50, 95% CI=0.95-2.36, p=0.084) and t (4; 14) (HR=1.38, 95% CI=0.90-2.11, p=0.143). Lenalidomide 44-56 adenine phosphoribosyltransferase Homo sapiens 124-127 25281606-6 2014 In patients with a long REM1 (>=3 years), salvage treatment with either repeat FCR or lenalidomide-based therapy results in subsequent median survival exceeding 5 years; for these patients, FCR rechallenge represents a reasonable standard of care. Lenalidomide 89-101 RRAD and GEM like GTPase 1 Homo sapiens 24-28 24721791-5 2014 Moreover, repression of the IRF4 target gene MYC in bortezomib-resistant cells by gene knockdown or treatment with CPI203, a BET (bromodomain and extra terminal) bromodomain inhibitor, synergistically induced cell death when combined with lenalidomide. Lenalidomide 239-251 interferon regulatory factor 4 Mus musculus 28-32 24721791-5 2014 Moreover, repression of the IRF4 target gene MYC in bortezomib-resistant cells by gene knockdown or treatment with CPI203, a BET (bromodomain and extra terminal) bromodomain inhibitor, synergistically induced cell death when combined with lenalidomide. Lenalidomide 239-251 myelocytomatosis oncogene Mus musculus 45-48 24721791-7 2014 Together, these results suggest that exacerbated IRF4/MYC signaling is associated to bortezomib resistance in MCL in vivo and warrant clinical evaluation of lenalidomide plus BET inhibitor combination in MCL cases refractory to proteasome inhibition. Lenalidomide 157-169 interferon regulatory factor 4 Mus musculus 49-53 24721791-7 2014 Together, these results suggest that exacerbated IRF4/MYC signaling is associated to bortezomib resistance in MCL in vivo and warrant clinical evaluation of lenalidomide plus BET inhibitor combination in MCL cases refractory to proteasome inhibition. Lenalidomide 157-169 myelocytomatosis oncogene Mus musculus 54-57 25295231-11 2014 Through its inhibitory effect on PP2A, lenalidomide stabilizes MDM2 to restore p53 degradation in erythroid precursors, with subsequent arrest in G2/M. Lenalidomide 39-51 protein phosphatase 2 phosphatase activator Homo sapiens 33-37 25295231-11 2014 Through its inhibitory effect on PP2A, lenalidomide stabilizes MDM2 to restore p53 degradation in erythroid precursors, with subsequent arrest in G2/M. Lenalidomide 39-51 MDM2 proto-oncogene Homo sapiens 63-67 25295231-11 2014 Through its inhibitory effect on PP2A, lenalidomide stabilizes MDM2 to restore p53 degradation in erythroid precursors, with subsequent arrest in G2/M. Lenalidomide 39-51 tumor protein p53 Homo sapiens 79-82 24687382-1 2014 Cereblon (CRBN) is essential for the anti-myeloma (MM) activity of immunomodulatory drugs (IMiDs), such as thalidomide and lenalidomide. Lenalidomide 123-135 cereblon Homo sapiens 10-14 24990888-0 2014 Lenalidomide inhibits the proliferation of CLL cells via a cereblon/p21(WAF1/Cip1)-dependent mechanism independent of functional p53. Lenalidomide 0-12 cyclin dependent kinase inhibitor 1A Homo sapiens 68-81 24990888-2 2014 We examined the direct effect of lenalidomide on CLL-cell proliferation induced by CD154-expressing accessory cells in media containing interleukin-4 and -10. Lenalidomide 33-45 CD40 ligand Homo sapiens 83-88 24990888-3 2014 Treatment with lenalidomide significantly inhibited CLL-cell proliferation, an effect that was associated with the p53-independent upregulation of the cyclin-dependent kinase inhibitor, p21(WAF1/Cip1) (p21). Lenalidomide 15-27 tumor protein p53 Homo sapiens 115-118 24990888-3 2014 Treatment with lenalidomide significantly inhibited CLL-cell proliferation, an effect that was associated with the p53-independent upregulation of the cyclin-dependent kinase inhibitor, p21(WAF1/Cip1) (p21). Lenalidomide 15-27 cyclin dependent kinase inhibitor 1A Homo sapiens 186-199 24990888-3 2014 Treatment with lenalidomide significantly inhibited CLL-cell proliferation, an effect that was associated with the p53-independent upregulation of the cyclin-dependent kinase inhibitor, p21(WAF1/Cip1) (p21). Lenalidomide 15-27 cyclin dependent kinase inhibitor 1A Homo sapiens 186-189 24990888-4 2014 Silencing p21 with small interfering RNA impaired the capacity of lenalidomide to inhibit CLL-cell proliferation. Lenalidomide 66-78 cyclin dependent kinase inhibitor 1A Homo sapiens 10-13 24990888-5 2014 Silencing cereblon, a known molecular target of lenalidomide, impaired the capacity of lenalidomide to induce expression of p21, inhibit CD154-induced CLL-cell proliferation, or enhance the degradation of Ikaros family zinc finger proteins 1 and 3. Lenalidomide 48-60 cyclin dependent kinase inhibitor 1A Homo sapiens 124-127 24990888-5 2014 Silencing cereblon, a known molecular target of lenalidomide, impaired the capacity of lenalidomide to induce expression of p21, inhibit CD154-induced CLL-cell proliferation, or enhance the degradation of Ikaros family zinc finger proteins 1 and 3. Lenalidomide 48-60 CD40 ligand Homo sapiens 137-142 24990888-5 2014 Silencing cereblon, a known molecular target of lenalidomide, impaired the capacity of lenalidomide to induce expression of p21, inhibit CD154-induced CLL-cell proliferation, or enhance the degradation of Ikaros family zinc finger proteins 1 and 3. Lenalidomide 48-60 zinc finger protein 266 Homo sapiens 219-247 24990888-5 2014 Silencing cereblon, a known molecular target of lenalidomide, impaired the capacity of lenalidomide to induce expression of p21, inhibit CD154-induced CLL-cell proliferation, or enhance the degradation of Ikaros family zinc finger proteins 1 and 3. Lenalidomide 87-99 cyclin dependent kinase inhibitor 1A Homo sapiens 124-127 24990888-5 2014 Silencing cereblon, a known molecular target of lenalidomide, impaired the capacity of lenalidomide to induce expression of p21, inhibit CD154-induced CLL-cell proliferation, or enhance the degradation of Ikaros family zinc finger proteins 1 and 3. Lenalidomide 87-99 CD40 ligand Homo sapiens 137-142 24990888-5 2014 Silencing cereblon, a known molecular target of lenalidomide, impaired the capacity of lenalidomide to induce expression of p21, inhibit CD154-induced CLL-cell proliferation, or enhance the degradation of Ikaros family zinc finger proteins 1 and 3. Lenalidomide 87-99 zinc finger protein 266 Homo sapiens 219-247 24990888-6 2014 We isolated CLL cells from the blood of patients before and after short-term treatment with low-dose lenalidomide (5 mg per day) and found the leukemia cells were also induced to express p21 in vivo. Lenalidomide 101-113 cyclin dependent kinase inhibitor 1A Homo sapiens 187-190 24990888-7 2014 These results indicate that lenalidomide can directly inhibit proliferation of CLL cells in a cereblon/p21-dependent but p53-independent manner, at concentrations achievable in vivo, potentially contributing to the capacity of this drug to inhibit disease-progression in patients with CLL. Lenalidomide 28-40 cyclin dependent kinase inhibitor 1A Homo sapiens 103-106 24990888-7 2014 These results indicate that lenalidomide can directly inhibit proliferation of CLL cells in a cereblon/p21-dependent but p53-independent manner, at concentrations achievable in vivo, potentially contributing to the capacity of this drug to inhibit disease-progression in patients with CLL. Lenalidomide 28-40 tumor protein p53 Homo sapiens 121-124 25248908-0 2014 [Three cases of lenalidomide-resistant IgA myeloma for which a response was regained after the addition of clarithromycin]. Lenalidomide 16-28 CD79a molecule Homo sapiens 39-42 24569836-0 2014 SNS01-T modulation of eIF5A inhibits B-cell cancer progression and synergizes with bortezomib and lenalidomide. Lenalidomide 98-110 eukaryotic translation initiation factor 5A Homo sapiens 22-27 25108355-0 2014 Structure of the human Cereblon-DDB1-lenalidomide complex reveals basis for responsiveness to thalidomide analogs. Lenalidomide 37-49 damage specific DNA binding protein 1 Homo sapiens 32-36 25108355-1 2014 The Cul4-Rbx1-DDB1-Cereblon E3 ubiquitin ligase complex is the target of thalidomide, lenalidomide and pomalidomide, therapeutically important drugs for multiple myeloma and other B-cell malignancies. Lenalidomide 86-98 ring-box 1 Homo sapiens 9-13 25108355-1 2014 The Cul4-Rbx1-DDB1-Cereblon E3 ubiquitin ligase complex is the target of thalidomide, lenalidomide and pomalidomide, therapeutically important drugs for multiple myeloma and other B-cell malignancies. Lenalidomide 86-98 damage specific DNA binding protein 1 Homo sapiens 14-18 25108355-3 2014 Here we present the crystal structure of human CRBN bound to DDB1 and the drug lenalidomide. Lenalidomide 79-91 cereblon Homo sapiens 47-51 25108355-4 2014 A hydrophobic pocket in the thalidomide-binding domain (TBD) of CRBN accommodates the glutarimide moiety of lenalidomide, whereas the isoindolinone ring is exposed to solvent. Lenalidomide 108-120 cereblon Homo sapiens 64-68 25165875-0 2014 Combination of lenalidomide with vitamin D3 induces apoptosis in mantle cell lymphoma via demethylation of BIK. Lenalidomide 15-27 BCL2 interacting killer Homo sapiens 107-110 25043012-4 2014 Here we present crystal structures of the DDB1-CRBN complex bound to thalidomide, lenalidomide and pomalidomide. Lenalidomide 82-94 damage specific DNA binding protein 1 Homo sapiens 42-46 25043012-4 2014 Here we present crystal structures of the DDB1-CRBN complex bound to thalidomide, lenalidomide and pomalidomide. Lenalidomide 82-94 cereblon Homo sapiens 47-51 24757092-0 2014 Lenalidomide-based maintenance therapy reduces TNF receptor 2 on CD4 T cells and enhances immune effector function in acute myeloid leukemia patients. Lenalidomide 0-12 tumor necrosis factor Homo sapiens 47-50 24757092-0 2014 Lenalidomide-based maintenance therapy reduces TNF receptor 2 on CD4 T cells and enhances immune effector function in acute myeloid leukemia patients. Lenalidomide 0-12 CD4 molecule Homo sapiens 65-68 24757092-7 2014 Our data indicates that although treatment with lenalidomide and azacitidine increased cytokine production by effector T cells in all patients, durable clinical remissions may be observed in patients with a concomitant reduction in TNFR2+ T cells and TNFR2+ Tregs. Lenalidomide 48-60 TNF receptor superfamily member 1B Homo sapiens 232-237 24757092-7 2014 Our data indicates that although treatment with lenalidomide and azacitidine increased cytokine production by effector T cells in all patients, durable clinical remissions may be observed in patients with a concomitant reduction in TNFR2+ T cells and TNFR2+ Tregs. Lenalidomide 48-60 TNF receptor superfamily member 1B Homo sapiens 251-256 24757092-8 2014 In vitro studies further demonstrated that lenalidomide can reduce TNFR2 expression and can augment effector cytokine production by T cells, which can be further enhanced by azacitidine. Lenalidomide 43-55 TNF receptor superfamily member 1B Homo sapiens 67-72 24757092-9 2014 These results indicate that reduction of TNFR2+ T cells in AML postremission phase may result from combined azacitidine/lenalidomide therapy and may contribute to an improved clinical outcome. Lenalidomide 120-132 TNF receptor superfamily member 1B Homo sapiens 41-46 24687382-3 2014 Using immunohistochemical (IHC) staining on paraffin-embedded bone marrow sections, the expression of CRBN protein in myeloma cells (MCs) was assessed in 40 relapsed/refractory MM (RRMM) patients who received lenalidomide/dexamethasone (LD) and 45 and 22 newly diagnosed MM (NDMM) patients who received thalidomide/dexamethasone (TD) and melphalan/bortezomib/prednisolone (MVP), respectively. Lenalidomide 209-221 cereblon Homo sapiens 102-106 24712857-3 2014 Lenalidomide therapy was associated with increased amounts of a CD8(+) T cell subset, phenotypically staged between classical central memory T cells (TCM) and effector memory T cells (TEM), consequently termed TCM/TEM. Lenalidomide 0-12 CD8a molecule Homo sapiens 64-67 24840055-5 2014 demonstrates that, by binding to CRBN and altering its selectivity, lenalidomide potentiates the ubiquitination and proteolysis of 2 specific proteins, IKZF1 and IKZF3. Lenalidomide 68-80 cereblon Homo sapiens 33-37 24840055-5 2014 demonstrates that, by binding to CRBN and altering its selectivity, lenalidomide potentiates the ubiquitination and proteolysis of 2 specific proteins, IKZF1 and IKZF3. Lenalidomide 68-80 IKAROS family zinc finger 1 Homo sapiens 152-157 24840055-5 2014 demonstrates that, by binding to CRBN and altering its selectivity, lenalidomide potentiates the ubiquitination and proteolysis of 2 specific proteins, IKZF1 and IKZF3. Lenalidomide 68-80 IKAROS family zinc finger 3 Homo sapiens 162-167 24840055-7 2014 IKZF1 and IKZF3 are transcription factors that are necessary for multiple myeloma, and repression of these transcription factors is a likely mechanism for lenalidomide activity in this disease. Lenalidomide 155-167 IKAROS family zinc finger 1 Homo sapiens 0-5 24840055-7 2014 IKZF1 and IKZF3 are transcription factors that are necessary for multiple myeloma, and repression of these transcription factors is a likely mechanism for lenalidomide activity in this disease. Lenalidomide 155-167 IKAROS family zinc finger 3 Homo sapiens 10-15 24712857-5 2014 In addition, lenalidomide-treated patients showed higher abundance of CD14(+) myeloid cells co-expressing CD15. Lenalidomide 13-25 CD14 molecule Homo sapiens 70-74 24712857-5 2014 In addition, lenalidomide-treated patients showed higher abundance of CD14(+) myeloid cells co-expressing CD15. Lenalidomide 13-25 fucosyltransferase 4 Homo sapiens 106-110 24891322-7 2014 Chronic stimulation with LPS accelerated the development of MDS in mDia1 heterozygous and knockout mice that can be rescued by lenalidomide. Lenalidomide 127-139 diaphanous related formin 1 Mus musculus 67-72 24237448-7 2014 Lenalidomide impaired long-term thymic T-cell reconstitution, decreased CD4 + and CD8 + CD45RA + CCR7 - effector-terminal T-cell absolute counts and increased CD4 + CD25 + CD127 - /low regulatory T-cells. Lenalidomide 0-12 CD4 molecule Homo sapiens 72-75 24237448-7 2014 Lenalidomide impaired long-term thymic T-cell reconstitution, decreased CD4 + and CD8 + CD45RA + CCR7 - effector-terminal T-cell absolute counts and increased CD4 + CD25 + CD127 - /low regulatory T-cells. Lenalidomide 0-12 CD8a molecule Homo sapiens 82-85 24237448-7 2014 Lenalidomide impaired long-term thymic T-cell reconstitution, decreased CD4 + and CD8 + CD45RA + CCR7 - effector-terminal T-cell absolute counts and increased CD4 + CD25 + CD127 - /low regulatory T-cells. Lenalidomide 0-12 protein tyrosine phosphatase receptor type C Homo sapiens 88-92 24237448-7 2014 Lenalidomide impaired long-term thymic T-cell reconstitution, decreased CD4 + and CD8 + CD45RA + CCR7 - effector-terminal T-cell absolute counts and increased CD4 + CD25 + CD127 - /low regulatory T-cells. Lenalidomide 0-12 C-C motif chemokine receptor 7 Homo sapiens 97-101 24237448-7 2014 Lenalidomide impaired long-term thymic T-cell reconstitution, decreased CD4 + and CD8 + CD45RA + CCR7 - effector-terminal T-cell absolute counts and increased CD4 + CD25 + CD127 - /low regulatory T-cells. Lenalidomide 0-12 CD4 molecule Homo sapiens 88-91 24237448-7 2014 Lenalidomide impaired long-term thymic T-cell reconstitution, decreased CD4 + and CD8 + CD45RA + CCR7 - effector-terminal T-cell absolute counts and increased CD4 + CD25 + CD127 - /low regulatory T-cells. Lenalidomide 0-12 interferon stimulated exonuclease gene 20 Homo sapiens 165-169 24237448-7 2014 Lenalidomide impaired long-term thymic T-cell reconstitution, decreased CD4 + and CD8 + CD45RA + CCR7 - effector-terminal T-cell absolute counts and increased CD4 + CD25 + CD127 - /low regulatory T-cells. Lenalidomide 0-12 interleukin 7 receptor Homo sapiens 172-177 24739012-1 2014 Lenalidomide (Revlimid; Selleck Chemicals, Houston, TX, USA), an analogue of thalidomide, possesses potent cytokine modulatory capacity through inhibition of cytokines such as tumour necrosis factor (TNF)-alpha, a cytokine pivotal for the onset and development of complications in obesity and diabetes mellitus. Lenalidomide 0-12 tumor necrosis factor Mus musculus 176-210 24914135-2 2014 Using 2 different methodologies, we identified 244 CRBN binding proteins and established relevance to MM biology by changes in their abundance after exposure to lenalidomide. Lenalidomide 161-173 cereblon Homo sapiens 51-55 24914135-4 2014 After lenalidomide treatment, the abundance of 46 CRBN binding proteins decreased. Lenalidomide 6-18 cereblon Homo sapiens 50-54 25610725-0 2014 Lenalidomide induces degradation of IKZF1 and IKZF3. Lenalidomide 0-12 IKAROS family zinc finger 1 Homo sapiens 36-41 25610725-0 2014 Lenalidomide induces degradation of IKZF1 and IKZF3. Lenalidomide 0-12 IKAROS family zinc finger 3 Homo sapiens 46-51 25610725-1 2014 Lenalidomide and its analogs, thalidomide and pomalidomide, specifically inhibit growth of mature B-cell lymphomas, including multiple myeloma, and induce interleukin-2 (IL-2) release from T cells. Lenalidomide 0-12 interleukin 2 Homo sapiens 155-168 25610725-1 2014 Lenalidomide and its analogs, thalidomide and pomalidomide, specifically inhibit growth of mature B-cell lymphomas, including multiple myeloma, and induce interleukin-2 (IL-2) release from T cells. Lenalidomide 0-12 interleukin 2 Homo sapiens 170-174 24982344-7 2014 In RT4 xenografts, lenalidomide significantly reduced tumor size and CD31 expression, and increased expression of CC3 (p<0.05). Lenalidomide 19-31 platelet and endothelial cell adhesion molecule 1 Homo sapiens 69-73 24739012-1 2014 Lenalidomide (Revlimid; Selleck Chemicals, Houston, TX, USA), an analogue of thalidomide, possesses potent cytokine modulatory capacity through inhibition of cytokines such as tumour necrosis factor (TNF)-alpha, a cytokine pivotal for the onset and development of complications in obesity and diabetes mellitus. Lenalidomide 14-22 tumor necrosis factor Mus musculus 176-210 24739012-9 2014 Western blot analysis revealed elevated TNF-alpha, Bax, Bcl-2, Bip, caspase 8 and caspase 9 in ob/ob mice with various degrees of reversal by lenalidomide treatment. Lenalidomide 142-154 tumor necrosis factor Mus musculus 40-49 24739012-9 2014 Western blot analysis revealed elevated TNF-alpha, Bax, Bcl-2, Bip, caspase 8 and caspase 9 in ob/ob mice with various degrees of reversal by lenalidomide treatment. Lenalidomide 142-154 BCL2-associated X protein Mus musculus 51-54 24739012-9 2014 Western blot analysis revealed elevated TNF-alpha, Bax, Bcl-2, Bip, caspase 8 and caspase 9 in ob/ob mice with various degrees of reversal by lenalidomide treatment. Lenalidomide 142-154 B cell leukemia/lymphoma 2 Mus musculus 56-61 24739012-9 2014 Western blot analysis revealed elevated TNF-alpha, Bax, Bcl-2, Bip, caspase 8 and caspase 9 in ob/ob mice with various degrees of reversal by lenalidomide treatment. Lenalidomide 142-154 heat shock protein 5 Mus musculus 63-66 24739012-9 2014 Western blot analysis revealed elevated TNF-alpha, Bax, Bcl-2, Bip, caspase 8 and caspase 9 in ob/ob mice with various degrees of reversal by lenalidomide treatment. Lenalidomide 142-154 caspase 8 Mus musculus 68-77 24739012-9 2014 Western blot analysis revealed elevated TNF-alpha, Bax, Bcl-2, Bip, caspase 8 and caspase 9 in ob/ob mice with various degrees of reversal by lenalidomide treatment. Lenalidomide 142-154 caspase 9 Mus musculus 82-91 23922218-0 2014 Flow cytometry-based enumeration and functional characterization of CD8 T regulatory cells in patients with multiple myeloma before and after lenalidomide plus dexamethasone treatment. Lenalidomide 142-154 CD8a molecule Homo sapiens 68-71 24682512-3 2014 In this study, bone marrow biopsies from 85 patients treated with lenalidomide in the MDS-004 clinical trial were retrospectively assessed for p53 expression by immunohistochemistry in association with outcome. Lenalidomide 66-78 tumor protein p53 Homo sapiens 143-146 24742716-2 2014 Lenalidomide loaded chitosan nanoparticles (LND-CS-NPs) were in the size range of 220-295 nm and characterized by DLS, TEM, FT-IR, TGA and XRD. Lenalidomide 0-12 T-box transcription factor 1 Homo sapiens 131-134 24966686-0 2014 Myelodysplastic disorders carrying both isolated del(5q) and JAK2(V617F) mutation: concise review, with focus on lenalidomide therapy. Lenalidomide 113-125 Janus kinase 2 Homo sapiens 61-71 25189730-5 2014 The rationale of combination of danazol or lenalidomide with ruxolitinib is mainly based on mitigation of anti-JAK2-mediated myelosuppression. Lenalidomide 43-55 Janus kinase 2 Homo sapiens 111-115 24706190-7 2014 Moreover, bortezomib-induced up-regulation of CHOP was readily enhanced by lenalidomide in contact with stromal cells. Lenalidomide 75-87 DNA damage inducible transcript 3 Homo sapiens 46-50 24328678-0 2014 Immunomodulatory agents lenalidomide and pomalidomide co-stimulate T cells by inducing degradation of T cell repressors Ikaros and Aiolos via modulation of the E3 ubiquitin ligase complex CRL4(CRBN.). Lenalidomide 24-36 IKAROS family zinc finger 3 Homo sapiens 131-137 24659021-1 2014 PURPOSE: Lenalidomide, a weak substrate of P-glycoprotein (P-gp) in vitro, is an oral anticancer drug eliminated predominantly via renal excretion as unchanged compound. Lenalidomide 9-21 ATP binding cassette subfamily B member 1 Homo sapiens 43-57 24659021-1 2014 PURPOSE: Lenalidomide, a weak substrate of P-glycoprotein (P-gp) in vitro, is an oral anticancer drug eliminated predominantly via renal excretion as unchanged compound. Lenalidomide 9-21 ATP binding cassette subfamily B member 1 Homo sapiens 59-63 24659021-2 2014 The role of P-gp in lenalidomide disposition and the associated clinical relevance were evaluated. Lenalidomide 20-32 ATP binding cassette subfamily B member 1 Homo sapiens 12-16 24351336-0 2014 Increase in antibody-dependent cellular cytotoxicity (ADCC) in a patient with advanced colorectal carcinoma carrying a KRAS mutation under lenalidomide therapy. Lenalidomide 139-151 KRAS proto-oncogene, GTPase Homo sapiens 119-123 24351336-5 2014 In a heavily pretreated patient with advanced colorectal cancer carrying mutations in APC and KRAS genes, we show an early metabolic response and enhanced NK cell activity to monotherapy with lenalidomide. Lenalidomide 192-204 KRAS proto-oncogene, GTPase Homo sapiens 94-98 24627193-11 2014 TP53 mutations are important predictors of AML progression and possible resistance to lenalidomide. Lenalidomide 86-98 tumor protein p53 Homo sapiens 0-4 24606326-11 2014 The ORR and PFS in patients with low-affinity FCGR3A polymorphisms (F/F and F/V) suggest that lenalidomide may improve the activity of rituximab in these patients. Lenalidomide 94-106 Fc gamma receptor IIIa Homo sapiens 46-52 24627218-4 2014 Lenalidomide was also evaluated as an inhibitor of P-gp, BCRP, MRP2, OCT2, OAT1, OAT3, OATP1B1, OATP1B3 and bile salt export pump (BSEP). Lenalidomide 0-12 ATP binding cassette subfamily B member 1 Homo sapiens 51-55 24627218-4 2014 Lenalidomide was also evaluated as an inhibitor of P-gp, BCRP, MRP2, OCT2, OAT1, OAT3, OATP1B1, OATP1B3 and bile salt export pump (BSEP). Lenalidomide 0-12 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 57-61 24627218-4 2014 Lenalidomide was also evaluated as an inhibitor of P-gp, BCRP, MRP2, OCT2, OAT1, OAT3, OATP1B1, OATP1B3 and bile salt export pump (BSEP). Lenalidomide 0-12 ATP binding cassette subfamily C member 2 Homo sapiens 63-67 24627218-4 2014 Lenalidomide was also evaluated as an inhibitor of P-gp, BCRP, MRP2, OCT2, OAT1, OAT3, OATP1B1, OATP1B3 and bile salt export pump (BSEP). Lenalidomide 0-12 POU class 2 homeobox 2 Homo sapiens 69-73 24627218-4 2014 Lenalidomide was also evaluated as an inhibitor of P-gp, BCRP, MRP2, OCT2, OAT1, OAT3, OATP1B1, OATP1B3 and bile salt export pump (BSEP). Lenalidomide 0-12 solute carrier family 22 member 6 Homo sapiens 75-79 24627218-4 2014 Lenalidomide was also evaluated as an inhibitor of P-gp, BCRP, MRP2, OCT2, OAT1, OAT3, OATP1B1, OATP1B3 and bile salt export pump (BSEP). Lenalidomide 0-12 solute carrier family 22 member 8 Homo sapiens 81-85 24627218-4 2014 Lenalidomide was also evaluated as an inhibitor of P-gp, BCRP, MRP2, OCT2, OAT1, OAT3, OATP1B1, OATP1B3 and bile salt export pump (BSEP). Lenalidomide 0-12 solute carrier organic anion transporter family member 1B1 Homo sapiens 87-94 24627218-4 2014 Lenalidomide was also evaluated as an inhibitor of P-gp, BCRP, MRP2, OCT2, OAT1, OAT3, OATP1B1, OATP1B3 and bile salt export pump (BSEP). Lenalidomide 0-12 solute carrier organic anion transporter family member 1B3 Homo sapiens 96-103 24627218-4 2014 Lenalidomide was also evaluated as an inhibitor of P-gp, BCRP, MRP2, OCT2, OAT1, OAT3, OATP1B1, OATP1B3 and bile salt export pump (BSEP). Lenalidomide 0-12 ATP binding cassette subfamily B member 11 Homo sapiens 108-129 24627218-4 2014 Lenalidomide was also evaluated as an inhibitor of P-gp, BCRP, MRP2, OCT2, OAT1, OAT3, OATP1B1, OATP1B3 and bile salt export pump (BSEP). Lenalidomide 0-12 ATP binding cassette subfamily B member 11 Homo sapiens 131-135 24627218-5 2014 In addition, inhibition of UDP-glucuronosyltransferase 1A1 (UGT1A1) variants by lenalidomide was also assessed. Lenalidomide 80-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-58 24627218-5 2014 In addition, inhibition of UDP-glucuronosyltransferase 1A1 (UGT1A1) variants by lenalidomide was also assessed. Lenalidomide 80-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 24627218-7 2014 RESULTS: Results of these studies indicate that the lenalidomide is not a substrate for the transporters examined, except that it is weak substrate of P-gp. Lenalidomide 52-64 ATP binding cassette subfamily B member 1 Homo sapiens 151-155 24328678-2 2014 T cell co-stimulation by lenalidomide or pomalidomide is cereblon dependent: however, the CRL4(CRBN) substrates responsible for T cell co-stimulation have yet to be identified. Lenalidomide 25-37 interleukin 17 receptor B Homo sapiens 90-94 24328678-2 2014 T cell co-stimulation by lenalidomide or pomalidomide is cereblon dependent: however, the CRL4(CRBN) substrates responsible for T cell co-stimulation have yet to be identified. Lenalidomide 25-37 cereblon Homo sapiens 95-99 24328678-3 2014 Here we demonstrate that interaction of the transcription factors Ikaros (IKZF1, encoded by the IKZF1 gene) and Aiolos (IKZF3, encoded by the IKZF3 gene) with CRL4(CRBN) is induced by lenalidomide or pomalidomide. Lenalidomide 184-196 IKAROS family zinc finger 1 Homo sapiens 74-79 24328678-3 2014 Here we demonstrate that interaction of the transcription factors Ikaros (IKZF1, encoded by the IKZF1 gene) and Aiolos (IKZF3, encoded by the IKZF3 gene) with CRL4(CRBN) is induced by lenalidomide or pomalidomide. Lenalidomide 184-196 IKAROS family zinc finger 1 Homo sapiens 96-101 24328678-3 2014 Here we demonstrate that interaction of the transcription factors Ikaros (IKZF1, encoded by the IKZF1 gene) and Aiolos (IKZF3, encoded by the IKZF3 gene) with CRL4(CRBN) is induced by lenalidomide or pomalidomide. Lenalidomide 184-196 IKAROS family zinc finger 3 Homo sapiens 112-118 24328678-3 2014 Here we demonstrate that interaction of the transcription factors Ikaros (IKZF1, encoded by the IKZF1 gene) and Aiolos (IKZF3, encoded by the IKZF3 gene) with CRL4(CRBN) is induced by lenalidomide or pomalidomide. Lenalidomide 184-196 IKAROS family zinc finger 3 Homo sapiens 120-125 24328678-3 2014 Here we demonstrate that interaction of the transcription factors Ikaros (IKZF1, encoded by the IKZF1 gene) and Aiolos (IKZF3, encoded by the IKZF3 gene) with CRL4(CRBN) is induced by lenalidomide or pomalidomide. Lenalidomide 184-196 IKAROS family zinc finger 3 Homo sapiens 142-147 24328678-3 2014 Here we demonstrate that interaction of the transcription factors Ikaros (IKZF1, encoded by the IKZF1 gene) and Aiolos (IKZF3, encoded by the IKZF3 gene) with CRL4(CRBN) is induced by lenalidomide or pomalidomide. Lenalidomide 184-196 interleukin 17 receptor B Homo sapiens 159-163 24328678-3 2014 Here we demonstrate that interaction of the transcription factors Ikaros (IKZF1, encoded by the IKZF1 gene) and Aiolos (IKZF3, encoded by the IKZF3 gene) with CRL4(CRBN) is induced by lenalidomide or pomalidomide. Lenalidomide 184-196 cereblon Homo sapiens 164-168 24328678-7 2014 Importantly, Aiolos could serve as a proximal pharmacodynamic marker for lenalidomide and pomalidomide, as healthy human subjects administered lenalidomide demonstrated Aiolos degradation in their peripheral T cells. Lenalidomide 73-85 IKAROS family zinc finger 3 Homo sapiens 13-19 24328678-7 2014 Importantly, Aiolos could serve as a proximal pharmacodynamic marker for lenalidomide and pomalidomide, as healthy human subjects administered lenalidomide demonstrated Aiolos degradation in their peripheral T cells. Lenalidomide 143-155 IKAROS family zinc finger 3 Homo sapiens 13-19 24328678-7 2014 Importantly, Aiolos could serve as a proximal pharmacodynamic marker for lenalidomide and pomalidomide, as healthy human subjects administered lenalidomide demonstrated Aiolos degradation in their peripheral T cells. Lenalidomide 143-155 IKAROS family zinc finger 3 Homo sapiens 169-175 24328678-0 2014 Immunomodulatory agents lenalidomide and pomalidomide co-stimulate T cells by inducing degradation of T cell repressors Ikaros and Aiolos via modulation of the E3 ubiquitin ligase complex CRL4(CRBN.). Lenalidomide 24-36 interleukin 17 receptor B Homo sapiens 188-192 24328678-0 2014 Immunomodulatory agents lenalidomide and pomalidomide co-stimulate T cells by inducing degradation of T cell repressors Ikaros and Aiolos via modulation of the E3 ubiquitin ligase complex CRL4(CRBN.). Lenalidomide 24-36 cereblon Homo sapiens 193-197 24328678-1 2014 Cereblon (CRBN), the molecular target of lenalidomide and pomalidomide, is a substrate receptor of the cullin ring E3 ubiquitin ligase complex, CRL4(CRBN) . Lenalidomide 41-53 cereblon Homo sapiens 10-14 24328678-1 2014 Cereblon (CRBN), the molecular target of lenalidomide and pomalidomide, is a substrate receptor of the cullin ring E3 ubiquitin ligase complex, CRL4(CRBN) . Lenalidomide 41-53 CDK2 associated cullin domain 1 Homo sapiens 103-109 24328678-1 2014 Cereblon (CRBN), the molecular target of lenalidomide and pomalidomide, is a substrate receptor of the cullin ring E3 ubiquitin ligase complex, CRL4(CRBN) . Lenalidomide 41-53 interleukin 17 receptor B Homo sapiens 144-148 24328678-1 2014 Cereblon (CRBN), the molecular target of lenalidomide and pomalidomide, is a substrate receptor of the cullin ring E3 ubiquitin ligase complex, CRL4(CRBN) . Lenalidomide 41-53 cereblon Homo sapiens 149-153 24236538-7 2014 This cytotoxic effect and BCL2 downregulation were further potentiated when AT-101 was combined with lenalidomide/dexamethasone (LDA). Lenalidomide 101-113 BCL2 apoptosis regulator Homo sapiens 26-30 24297864-7 2014 The HIF-1alpha inhibition by either siRNA or panobinostat impaired the MMECs angiogenesis-related functions both in vitro and in vivo and restored MMEC sensitivity to bortezomib and lenalidomide. Lenalidomide 182-194 hypoxia inducible factor 1 subunit alpha Homo sapiens 4-14 24297864-9 2014 CONCLUSIONS: The HIF-1alpha protein in MMECs may induce angiogenesis and resistance to bortezomib and lenalidomide and may be a plausible target for the antiangiogenic management of patients with well-defined relapsed/refractory multiple myeloma. Lenalidomide 102-114 hypoxia inducible factor 1 subunit alpha Homo sapiens 17-27 24211319-3 2014 In multiple regression analysis, age, platelet count at time of mobilization, type of GF utilized, and extent of exposure to lenalidomide independently correlated with peripheral blood (PB)-CD34+ and were integrated in a predicting score (PS) for poor mobilizers, defined as PB-CD34+ < 20/mm(3) 4 days after initiation of GF. Lenalidomide 125-137 CD34 molecule Homo sapiens 190-194 24211319-3 2014 In multiple regression analysis, age, platelet count at time of mobilization, type of GF utilized, and extent of exposure to lenalidomide independently correlated with peripheral blood (PB)-CD34+ and were integrated in a predicting score (PS) for poor mobilizers, defined as PB-CD34+ < 20/mm(3) 4 days after initiation of GF. Lenalidomide 125-137 CD34 molecule Homo sapiens 278-282 24253022-8 2014 Immunotherapy against MM is also being explored, and probably the most attractive example of this approach is the combination of the anti-CS1 MoAb elotuzumab with lenalidomide and dexamethasone, which has produced exciting results in the relapsed/refractory setting. Lenalidomide 163-175 chorionic somatomammotropin hormone 1 Homo sapiens 138-141 24335103-9 2014 Data from correlative studies on peripheral blood samples suggest that the effects of lenalidomide could be associated with decreased circulating CD25(+) T cells and CD4(+) T-cell numbers. Lenalidomide 86-98 interleukin 2 receptor subunit alpha Homo sapiens 146-150 24335103-9 2014 Data from correlative studies on peripheral blood samples suggest that the effects of lenalidomide could be associated with decreased circulating CD25(+) T cells and CD4(+) T-cell numbers. Lenalidomide 86-98 CD4 molecule Homo sapiens 166-169 24212063-7 2014 Patients who respond to lenalidomide showed a more pronounced decrease of VEGF and bFGF than did patients with stable or progressive disease (p = 0.007 and p = 0.005). Lenalidomide 24-36 vascular endothelial growth factor A Homo sapiens 74-78 24212063-7 2014 Patients who respond to lenalidomide showed a more pronounced decrease of VEGF and bFGF than did patients with stable or progressive disease (p = 0.007 and p = 0.005). Lenalidomide 24-36 fibroblast growth factor 2 Homo sapiens 83-87 24178620-0 2014 Antitumoral activity of lenalidomide in in vitro and in vivo models of mantle cell lymphoma involves the destabilization of cyclin D1/p27KIP1 complexes. Lenalidomide 24-36 cyclin D1 Homo sapiens 124-133 23760401-0 2014 Evidence of a role for CD44 and cell adhesion in mediating resistance to lenalidomide in multiple myeloma: therapeutic implications. Lenalidomide 73-85 CD44 molecule (Indian blood group) Homo sapiens 23-27 23760401-2 2014 Lenalidomide-resistant models were found to overexpress the hyaluronan (HA)-binding protein CD44, a downstream Wnt/beta-catenin transcriptional target. Lenalidomide 0-12 CD44 molecule (Indian blood group) Homo sapiens 92-96 23760401-2 2014 Lenalidomide-resistant models were found to overexpress the hyaluronan (HA)-binding protein CD44, a downstream Wnt/beta-catenin transcriptional target. Lenalidomide 0-12 catenin beta 1 Homo sapiens 115-127 23760401-3 2014 Consistent with a role of CD44 in cell adhesion-mediated drug resistance (CAM-DR), lenalidomide-resistant myeloma cells were more adhesive to bone marrow stroma and HA-coated plates. Lenalidomide 83-95 CD44 molecule (Indian blood group) Homo sapiens 26-30 23760401-4 2014 Blockade of CD44 with monoclonal antibodies, free HA or CD44 knockdown reduced adhesion and sensitized to lenalidomide. Lenalidomide 106-118 CD44 molecule (Indian blood group) Homo sapiens 12-16 23760401-4 2014 Blockade of CD44 with monoclonal antibodies, free HA or CD44 knockdown reduced adhesion and sensitized to lenalidomide. Lenalidomide 106-118 CD44 molecule (Indian blood group) Homo sapiens 56-60 23760401-5 2014 Wnt/beta-catenin suppression by FH535 enhanced the activity of lenalidomide, as did interleukin-6 neutralization with siltuximab. Lenalidomide 63-75 catenin beta 1 Homo sapiens 4-16 23760401-6 2014 Notably, all-trans retinoic acid (ATRA) downregulated total beta-catenin, cell-surface and total CD44, reduced adhesion of lenalidomide-resistant myeloma cells and enhanced the activity of lenalidomide in a lenalidomide-resistant in vivo murine xenograft model. Lenalidomide 189-201 catenin (cadherin associated protein), beta 1 Mus musculus 60-72 23760401-6 2014 Notably, all-trans retinoic acid (ATRA) downregulated total beta-catenin, cell-surface and total CD44, reduced adhesion of lenalidomide-resistant myeloma cells and enhanced the activity of lenalidomide in a lenalidomide-resistant in vivo murine xenograft model. Lenalidomide 189-201 catenin (cadherin associated protein), beta 1 Mus musculus 60-72 23760401-8 2014 Taken together, our findings support the hypotheses that CD44 and CAM-DR contribute to lenalidomide resistance in multiple myeloma, that CD44 should be evaluated as a putative biomarker of sensitivity to lenalidomide, and that ATRA or other approaches that target CD44 may overcome clinical lenalidomide resistance. Lenalidomide 87-99 CD44 molecule (Indian blood group) Homo sapiens 57-61 24292623-0 2014 The myeloma drug lenalidomide promotes the cereblon-dependent destruction of Ikaros proteins. Lenalidomide 17-29 IKAROS family zinc finger 1 Homo sapiens 77-83 24292623-5 2014 Here we show that lenalidomide-bound cereblon acquires the ability to target for proteasomal degradation two specific B cell transcription factors, Ikaros family zinc finger proteins 1 and 3 (IKZF1 and IKZF3). Lenalidomide 18-30 IKAROS family zinc finger 1 Homo sapiens 148-154 24292623-5 2014 Here we show that lenalidomide-bound cereblon acquires the ability to target for proteasomal degradation two specific B cell transcription factors, Ikaros family zinc finger proteins 1 and 3 (IKZF1 and IKZF3). Lenalidomide 18-30 zinc finger protein 266 Homo sapiens 162-190 24292623-5 2014 Here we show that lenalidomide-bound cereblon acquires the ability to target for proteasomal degradation two specific B cell transcription factors, Ikaros family zinc finger proteins 1 and 3 (IKZF1 and IKZF3). Lenalidomide 18-30 IKAROS family zinc finger 1 Homo sapiens 192-197 24292623-5 2014 Here we show that lenalidomide-bound cereblon acquires the ability to target for proteasomal degradation two specific B cell transcription factors, Ikaros family zinc finger proteins 1 and 3 (IKZF1 and IKZF3). Lenalidomide 18-30 IKAROS family zinc finger 3 Homo sapiens 202-207 24292623-6 2014 Analysis of myeloma cell lines revealed that loss of IKZF1 and IKZF3 is both necessary and sufficient for lenalidomide"s therapeutic effect, suggesting that the antitumor and teratogenic activities of thalidomide-like drugs are dissociable. Lenalidomide 106-118 IKAROS family zinc finger 1 Homo sapiens 53-58 24292623-6 2014 Analysis of myeloma cell lines revealed that loss of IKZF1 and IKZF3 is both necessary and sufficient for lenalidomide"s therapeutic effect, suggesting that the antitumor and teratogenic activities of thalidomide-like drugs are dissociable. Lenalidomide 106-118 IKAROS family zinc finger 3 Homo sapiens 63-68 24292625-0 2014 Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells. Lenalidomide 0-12 IKAROS family zinc finger 1 Homo sapiens 45-50 24292625-0 2014 Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells. Lenalidomide 0-12 IKAROS family zinc finger 3 Homo sapiens 55-60 24292625-2 2014 Using quantitative proteomics, we found that lenalidomide causes selective ubiquitination and degradation of two lymphoid transcription factors, IKZF1 and IKZF3, by the CRBN-CRL4 ubiquitin ligase. Lenalidomide 45-57 IKAROS family zinc finger 1 Homo sapiens 145-150 24292625-2 2014 Using quantitative proteomics, we found that lenalidomide causes selective ubiquitination and degradation of two lymphoid transcription factors, IKZF1 and IKZF3, by the CRBN-CRL4 ubiquitin ligase. Lenalidomide 45-57 IKAROS family zinc finger 3 Homo sapiens 155-160 24292625-4 2014 A single amino acid substitution of IKZF3 conferred resistance to lenalidomide-induced degradation and rescued lenalidomide-induced inhibition of cell growth. Lenalidomide 66-78 IKAROS family zinc finger 3 Homo sapiens 36-41 24292625-4 2014 A single amino acid substitution of IKZF3 conferred resistance to lenalidomide-induced degradation and rescued lenalidomide-induced inhibition of cell growth. Lenalidomide 111-123 IKAROS family zinc finger 3 Homo sapiens 36-41 24292625-5 2014 Similarly, we found that lenalidomide-induced interleukin-2 production in T cells is due to depletion of IKZF1 and IKZF3. Lenalidomide 25-37 interleukin 2 Homo sapiens 46-59 24292625-5 2014 Similarly, we found that lenalidomide-induced interleukin-2 production in T cells is due to depletion of IKZF1 and IKZF3. Lenalidomide 25-37 IKAROS family zinc finger 1 Homo sapiens 105-110 24292625-5 2014 Similarly, we found that lenalidomide-induced interleukin-2 production in T cells is due to depletion of IKZF1 and IKZF3. Lenalidomide 25-37 IKAROS family zinc finger 3 Homo sapiens 115-120 24178620-0 2014 Antitumoral activity of lenalidomide in in vitro and in vivo models of mantle cell lymphoma involves the destabilization of cyclin D1/p27KIP1 complexes. Lenalidomide 24-36 cyclin dependent kinase inhibitor 1B Homo sapiens 134-141 24178620-2 2014 We investigated whether the expression and subcellular localization of cyclin D1, a major cell-cycle regulator overexpressed in MCL, and the cyclin-dependent kinase inhibitor p27(KIP1), could identify MCL cases sensitive to lenalidomide, and whether the compound could modulate cyclin D1/p27(KIP1) complexes in MCL cells. Lenalidomide 224-236 cyclin D1 Homo sapiens 71-80 24178620-4 2014 Activity of lenalidomide in vitro and its effect on cyclin D1/p27(KIP1) complexes were evaluated by real-time PCR, immunoprecipitation, immunofluorescence, and Western blot. Lenalidomide 12-24 cyclin D1 Homo sapiens 52-61 24178620-4 2014 Activity of lenalidomide in vitro and its effect on cyclin D1/p27(KIP1) complexes were evaluated by real-time PCR, immunoprecipitation, immunofluorescence, and Western blot. Lenalidomide 12-24 interferon alpha inducible protein 27 Homo sapiens 62-65 24178620-4 2014 Activity of lenalidomide in vitro and its effect on cyclin D1/p27(KIP1) complexes were evaluated by real-time PCR, immunoprecipitation, immunofluorescence, and Western blot. Lenalidomide 12-24 cyclin dependent kinase inhibitor 1B Homo sapiens 66-70 24178620-9 2014 Lenalidomide mechanism of action relied on cyclin D1 downregulation and disruption of cyclin D1/p27(KIP1) complexes, followed by cytosolic accumulation of p27(KIP1), cell proliferation arrest, apoptosis, and angiogenesis inhibition. Lenalidomide 0-12 cyclin D1 Homo sapiens 43-52 24178620-9 2014 Lenalidomide mechanism of action relied on cyclin D1 downregulation and disruption of cyclin D1/p27(KIP1) complexes, followed by cytosolic accumulation of p27(KIP1), cell proliferation arrest, apoptosis, and angiogenesis inhibition. Lenalidomide 0-12 cyclin D1 Homo sapiens 86-95 24178620-9 2014 Lenalidomide mechanism of action relied on cyclin D1 downregulation and disruption of cyclin D1/p27(KIP1) complexes, followed by cytosolic accumulation of p27(KIP1), cell proliferation arrest, apoptosis, and angiogenesis inhibition. Lenalidomide 0-12 interferon alpha inducible protein 27 Homo sapiens 96-99 24178620-9 2014 Lenalidomide mechanism of action relied on cyclin D1 downregulation and disruption of cyclin D1/p27(KIP1) complexes, followed by cytosolic accumulation of p27(KIP1), cell proliferation arrest, apoptosis, and angiogenesis inhibition. Lenalidomide 0-12 cyclin dependent kinase inhibitor 1B Homo sapiens 100-104 24178620-9 2014 Lenalidomide mechanism of action relied on cyclin D1 downregulation and disruption of cyclin D1/p27(KIP1) complexes, followed by cytosolic accumulation of p27(KIP1), cell proliferation arrest, apoptosis, and angiogenesis inhibition. Lenalidomide 0-12 interferon alpha inducible protein 27 Homo sapiens 155-158 24178620-9 2014 Lenalidomide mechanism of action relied on cyclin D1 downregulation and disruption of cyclin D1/p27(KIP1) complexes, followed by cytosolic accumulation of p27(KIP1), cell proliferation arrest, apoptosis, and angiogenesis inhibition. Lenalidomide 0-12 cyclin dependent kinase inhibitor 1B Homo sapiens 159-163 24178620-10 2014 CONCLUSIONS: These results highlight a mechanism of action of lenalidomide in MCL cases with increased tumorigenicity in vivo, which is mediated by the dissociation of cyclin D1/p27(KIP1) complexes, and subsequent proliferation blockade and apoptosis induction. Lenalidomide 62-74 cyclin D1 Homo sapiens 168-177 24178620-10 2014 CONCLUSIONS: These results highlight a mechanism of action of lenalidomide in MCL cases with increased tumorigenicity in vivo, which is mediated by the dissociation of cyclin D1/p27(KIP1) complexes, and subsequent proliferation blockade and apoptosis induction. Lenalidomide 62-74 interferon alpha inducible protein 27 Homo sapiens 178-181 24178620-10 2014 CONCLUSIONS: These results highlight a mechanism of action of lenalidomide in MCL cases with increased tumorigenicity in vivo, which is mediated by the dissociation of cyclin D1/p27(KIP1) complexes, and subsequent proliferation blockade and apoptosis induction. Lenalidomide 62-74 cyclin dependent kinase inhibitor 1B Homo sapiens 182-186 23573828-7 2014 Our data thus indicate a functional role for the TRAIL/TRAIL-R system in lenalidomide-mediated NK-cell activity against MM cells, but also show that dasatinib is unsuitable to support or boost this effect. Lenalidomide 73-85 TNF superfamily member 10 Homo sapiens 49-54 24018623-5 2014 Advances in the understanding of the pathogenesis of the disease have suggested that lenalidomide targets aberrant signaling pathways caused by haplosufficiency of specific genes in a commonly deleted region on chromosome 5 (e.g., SPARC, RPS14, Cdc25C, and PP2A). Lenalidomide 85-97 secreted protein acidic and cysteine rich Homo sapiens 231-236 24018623-5 2014 Advances in the understanding of the pathogenesis of the disease have suggested that lenalidomide targets aberrant signaling pathways caused by haplosufficiency of specific genes in a commonly deleted region on chromosome 5 (e.g., SPARC, RPS14, Cdc25C, and PP2A). Lenalidomide 85-97 ribosomal protein S14 Homo sapiens 238-243 24018623-5 2014 Advances in the understanding of the pathogenesis of the disease have suggested that lenalidomide targets aberrant signaling pathways caused by haplosufficiency of specific genes in a commonly deleted region on chromosome 5 (e.g., SPARC, RPS14, Cdc25C, and PP2A). Lenalidomide 85-97 cell division cycle 25C Homo sapiens 245-251 24018623-5 2014 Advances in the understanding of the pathogenesis of the disease have suggested that lenalidomide targets aberrant signaling pathways caused by haplosufficiency of specific genes in a commonly deleted region on chromosome 5 (e.g., SPARC, RPS14, Cdc25C, and PP2A). Lenalidomide 85-97 protein phosphatase 2 phosphatase activator Homo sapiens 257-261 25313353-6 2014 The effect of lenalidomide on NK cells was secondary to the induction of IL-2 production by CD4 T cells. Lenalidomide 14-26 interleukin 2 Homo sapiens 73-77 25313353-9 2014 Lenalidomide also upregulated CD20 expression on leukemia cells and, accordingly, it had a synergistic effect with rituximab on promoting antibody-dependent cell-mediated cytotoxicity against primary leukemia cells. Lenalidomide 0-12 keratin 20 Homo sapiens 30-34 24206017-0 2014 Measuring cereblon as a biomarker of response or resistance to lenalidomide and pomalidomide requires use of standardized reagents and understanding of gene complexity. Lenalidomide 63-75 cereblon Homo sapiens 10-18 24206017-1 2014 Cereblon, a member of the cullin 4 ring ligase complex (CRL4), is the molecular target of the immunomodulatory drugs (IMiDs) lenalidomide and pomalidomide and is required for the antiproliferative activity of these agents in multiple myeloma (MM) and immunomodulatory activity in T cells. Lenalidomide 125-137 cereblon Homo sapiens 0-8 24206017-1 2014 Cereblon, a member of the cullin 4 ring ligase complex (CRL4), is the molecular target of the immunomodulatory drugs (IMiDs) lenalidomide and pomalidomide and is required for the antiproliferative activity of these agents in multiple myeloma (MM) and immunomodulatory activity in T cells. Lenalidomide 125-137 interleukin 17 receptor B Homo sapiens 56-60 24206017-2 2014 Cereblon"s central role as a target of lenalidomide and pomalidomide suggests potential utility as a predictive biomarker of response or resistance to IMiD therapy. Lenalidomide 39-51 cereblon Homo sapiens 0-8 24206017-7 2014 In cell lines made resistant to lenalidomide and pomalidomide, cereblon protein is greatly reduced. Lenalidomide 32-44 cereblon Homo sapiens 63-71 23573828-7 2014 Our data thus indicate a functional role for the TRAIL/TRAIL-R system in lenalidomide-mediated NK-cell activity against MM cells, but also show that dasatinib is unsuitable to support or boost this effect. Lenalidomide 73-85 TNF superfamily member 10 Homo sapiens 55-60 23636891-4 2013 Modulation of TNF-alpha expression, either by stimulation using myeloid-related protein (MRP8) or lipopolysaccharide or inhibition using lenalidomide on astrocytes, leads to similar dynamic changes in miR-155 expression. Lenalidomide 137-149 tumor necrosis factor Homo sapiens 14-23 23992230-4 2013 We examined HMCL protein expression levels of the lenalidomide target cereblon (CRBN) and its downstream target interferon regulatory factor-4 (IRF4), which have previously been shown to be predictive of lenalidomide response in HMCLs. Lenalidomide 50-62 cereblon Homo sapiens 80-84 23992230-4 2013 We examined HMCL protein expression levels of the lenalidomide target cereblon (CRBN) and its downstream target interferon regulatory factor-4 (IRF4), which have previously been shown to be predictive of lenalidomide response in HMCLs. Lenalidomide 204-216 cereblon Homo sapiens 80-84 23992230-4 2013 We examined HMCL protein expression levels of the lenalidomide target cereblon (CRBN) and its downstream target interferon regulatory factor-4 (IRF4), which have previously been shown to be predictive of lenalidomide response in HMCLs. Lenalidomide 204-216 interferon regulatory factor 4 Homo sapiens 112-142 23992230-4 2013 We examined HMCL protein expression levels of the lenalidomide target cereblon (CRBN) and its downstream target interferon regulatory factor-4 (IRF4), which have previously been shown to be predictive of lenalidomide response in HMCLs. Lenalidomide 204-216 interferon regulatory factor 4 Homo sapiens 144-148 23992230-10 2013 While CRBN and IRF4 have been shown to be associated with response to lenalidomide in patients, these findings do not translate back to HMCLs, which could be attributable to factors present in the bone marrow microenvironment. Lenalidomide 70-82 cereblon Homo sapiens 6-10 23992230-10 2013 While CRBN and IRF4 have been shown to be associated with response to lenalidomide in patients, these findings do not translate back to HMCLs, which could be attributable to factors present in the bone marrow microenvironment. Lenalidomide 70-82 interferon regulatory factor 4 Homo sapiens 15-19 24210217-6 2013 Bortezomib, thalidomide or lenalidomide extended OS of FGFR3 and/or c-MAF-positive patients. Lenalidomide 27-39 fibroblast growth factor receptor 3 Homo sapiens 55-60 24210217-6 2013 Bortezomib, thalidomide or lenalidomide extended OS of FGFR3 and/or c-MAF-positive patients. Lenalidomide 27-39 MAF bZIP transcription factor Homo sapiens 68-73 24244261-0 2013 Phase II open-label study to assess efficacy and safety of lenalidomide in combination with cetuximab in KRAS-mutant metastatic colorectal cancer. Lenalidomide 59-71 KRAS proto-oncogene, GTPase Homo sapiens 105-109 24244261-1 2013 UNLABELLED: This study aimed to assess the efficacy and safety of combination treatment with lenalidomide and cetuximab in KRAS-mutant metastatic colorectal cancer patients. Lenalidomide 93-105 KRAS proto-oncogene, GTPase Homo sapiens 123-127 24244687-0 2013 Immunomodulatory effects in a phase II study of lenalidomide combined with cetuximab in refractory KRAS-mutant metastatic colorectal cancer patients. Lenalidomide 48-60 KRAS proto-oncogene, GTPase Homo sapiens 99-103 24244687-3 2013 In addition, lenalidomide decreased the percentage of circulating CD19(+) B cells 2.6-fold (p<0.0001). Lenalidomide 13-25 CD19 molecule Homo sapiens 66-70 24189071-6 2013 Disease progression is attenuated in a Rhoh(-/-) mouse model of chronic lymphocytic leukemia and treatment of primary human chronic lymphocytic leukemia cells with Lenalidomide results in reduced RhoH protein levels. Lenalidomide 164-176 ras homolog family member H Mus musculus 39-43 24189071-6 2013 Disease progression is attenuated in a Rhoh(-/-) mouse model of chronic lymphocytic leukemia and treatment of primary human chronic lymphocytic leukemia cells with Lenalidomide results in reduced RhoH protein levels. Lenalidomide 164-176 ras homolog family member H Homo sapiens 196-200 23545991-1 2013 Lenalidomide-rituximab therapy is effective in grade 1-2 follicular and mantle cell lymphoma, but its efficacy in diffuse large B-cell lymphoma (DLBCL), transformed large cell lymphoma (TL) and grade 3 follicular lymphoma (FLG3) is unknown. Lenalidomide 0-12 hornerin Homo sapiens 223-227 24482747-4 2013 In particular, lenalidomide has been shown to stimulate the cytotoxic functions of T lymphocytes and natural killer cells, to limit the immunosuppressive impact of regulatory T cells, and to modulate the secretion of a wide range of cytokines, including tumor necrosis factor alpha, interferon gamma as well as interleukin (IL)-6, IL-10, and IL-12. Lenalidomide 15-27 tumor necrosis factor Homo sapiens 254-281 24482747-4 2013 In particular, lenalidomide has been shown to stimulate the cytotoxic functions of T lymphocytes and natural killer cells, to limit the immunosuppressive impact of regulatory T cells, and to modulate the secretion of a wide range of cytokines, including tumor necrosis factor alpha, interferon gamma as well as interleukin (IL)-6, IL-10, and IL-12. Lenalidomide 15-27 interferon gamma Homo sapiens 283-299 24482747-4 2013 In particular, lenalidomide has been shown to stimulate the cytotoxic functions of T lymphocytes and natural killer cells, to limit the immunosuppressive impact of regulatory T cells, and to modulate the secretion of a wide range of cytokines, including tumor necrosis factor alpha, interferon gamma as well as interleukin (IL)-6, IL-10, and IL-12. Lenalidomide 15-27 interleukin 10 Homo sapiens 331-336 23982484-0 2013 Lenalidomide overcomes suppression of human natural killer cell anti-tumor functions by neuroblastoma microenvironment-associated IL-6 and TGFbeta1. Lenalidomide 0-12 interleukin 6 Homo sapiens 130-134 23982484-0 2013 Lenalidomide overcomes suppression of human natural killer cell anti-tumor functions by neuroblastoma microenvironment-associated IL-6 and TGFbeta1. Lenalidomide 0-12 transforming growth factor beta 1 Homo sapiens 139-147 23982484-8 2013 Lenalidomide blocks IL-6 and TGFbeta1 activation of these signaling pathways and inhibits their suppression of NK cells. Lenalidomide 0-12 interleukin 6 Homo sapiens 20-24 23982484-8 2013 Lenalidomide blocks IL-6 and TGFbeta1 activation of these signaling pathways and inhibits their suppression of NK cells. Lenalidomide 0-12 transforming growth factor beta 1 Homo sapiens 29-37 23982484-11 2013 CONCLUSION: Immunotherapy with anti-tumor cell antibodies may be improved by lenalidomide, which enhances activation of NK cells and inhibits their suppression by IL-6 and TGFbeta1. Lenalidomide 77-89 interleukin 6 Homo sapiens 163-167 23982484-11 2013 CONCLUSION: Immunotherapy with anti-tumor cell antibodies may be improved by lenalidomide, which enhances activation of NK cells and inhibits their suppression by IL-6 and TGFbeta1. Lenalidomide 77-89 transforming growth factor beta 1 Homo sapiens 172-180 24170980-0 2013 Effects of MRP8, LPS, and lenalidomide on the expressions of TNF-alpha , brain-enriched, and inflammation-related microRNAs in the primary astrocyte culture. Lenalidomide 26-38 tumor necrosis factor Rattus norvegicus 61-70 24170980-4 2013 Further, we inhibited the expression of TNF- alpha in the astrocytes by using TNF- alpha inhibitor (lenalidomide) and tested for the first time the effect of this inhibition on the expressions of the same tested miRNAs. Lenalidomide 100-112 tumor necrosis factor Rattus norvegicus 40-50 24170980-4 2013 Further, we inhibited the expression of TNF- alpha in the astrocytes by using TNF- alpha inhibitor (lenalidomide) and tested for the first time the effect of this inhibition on the expressions of the same tested miRNAs. Lenalidomide 100-112 tumor necrosis factor Rattus norvegicus 78-88 24170980-6 2013 TNF- alpha inhibition with lenalidomide leads to opposite expressions of the tested miRNAs. Lenalidomide 27-39 tumor necrosis factor Rattus norvegicus 0-10 24043769-0 2013 TP53 suppression promotes erythropoiesis in del(5q) MDS, suggesting a targeted therapeutic strategy in lenalidomide-resistant patients. Lenalidomide 103-115 tumor protein p53 Homo sapiens 0-4 24043769-7 2013 We conclude that targeted suppression of p53 could support effective erythropoiesis in lenalidomide-resistant del(5q) MDS. Lenalidomide 87-99 tumor protein p53 Homo sapiens 41-44 23893930-0 2013 Flow cytometry based enumeration and functional characterization of CD8 T regulatory cells in patients with multiple myeloma before and after lenalidomide plus dexamethasone treatment. Lenalidomide 142-154 CD8a molecule Homo sapiens 68-71 23334269-7 2013 The data support the current International Multiple Myeloma Working Group guidelines recommending the use of cyclophosphamide and G-CSF based mobilization for patients previously exposed to lenalidomide. Lenalidomide 190-202 colony stimulating factor 3 Homo sapiens 130-135 23417027-4 2013 Our findings demonstrate that lenalidomide acts directly on bone marrow stromal cells via an Akt-mediated increase in Jun N-terminal kinase-dependent signaling resulting in activin A secretion, with consequent inhibition of osteoblastogenesis. Lenalidomide 30-42 AKT serine/threonine kinase 1 Homo sapiens 93-96 23614682-10 2013 This study reveals the importance of a low baseline platelet count, karyotypic complexity and TP53 mutational status for response to lenalidomide treatment. Lenalidomide 133-145 tumor protein p53 Homo sapiens 94-98 23614682-11 2013 It supports the molecular study of TP53 in MDS patients treated with lenalidomide. Lenalidomide 69-81 tumor protein p53 Homo sapiens 35-39 23252419-0 2013 Response to lenalidomide of a patient with t(2;3)(p23;q29) and JAK2 non-mutated refractory anemia with ring sideroblasts and thrombocytosis. Lenalidomide 12-24 Janus kinase 2 Homo sapiens 63-67 23434730-0 2013 A G polymorphism in the CRBN gene acts as a biomarker of response to treatment with lenalidomide in low/int-1 risk MDS without del(5q). Lenalidomide 84-96 cereblon Homo sapiens 24-28 23692564-9 2013 In conclusion, RBP with lenalidomide 25 mg/d, days 1-21 and bendamustine 75 mg/m(2) days 1-2 is well tolerated in patients with relapsed/refractory MM. Lenalidomide 24-36 SURP and G-patch domain containing 1 Homo sapiens 15-18 23609417-0 2013 Association of Th1 and Th2 cytokines with transient inflammatory reaction during lenalidomide plus dexamethasone therapy in multiple myeloma. Lenalidomide 81-93 negative elongation factor complex member C/D Homo sapiens 15-18 23565715-4 2013 CRBN expression levels correlated significantly with response to lenalidomide treatment (r = 0 48; P < 0 001). Lenalidomide 65-77 cereblon Homo sapiens 0-4 23565715-7 2013 Overall, a statistically significant association between baseline CRBN expression and response in MM patients treated with lenalidomide is shown. Lenalidomide 123-135 cereblon Homo sapiens 66-70 23506134-0 2013 Low RPS14 expression in MDS without 5q - aberration confers higher apoptosis rate of nucleated erythrocytes and predicts prolonged survival and possible response to lenalidomide in lower risk non-5q- patients. Lenalidomide 165-177 ribosomal protein S14 Homo sapiens 4-9 23506134-14 2013 Additionally, lower RPS14 predicts prolonged survival and possible response to lenalidomide in lower risk MDS patients. Lenalidomide 79-91 ribosomal protein S14 Homo sapiens 20-25 23434730-0 2013 A G polymorphism in the CRBN gene acts as a biomarker of response to treatment with lenalidomide in low/int-1 risk MDS without del(5q). Lenalidomide 84-96 Wnt family member 1 Homo sapiens 104-109 23609417-6 2013 Furthermore, Len enhanced the production of both Th1 and Th2 cytokines in normal peripheral blood mononuclear cells and in patient bone marrow mononuclear cells containing primary myeloma cells and lymphocytes. Lenalidomide 13-16 negative elongation factor complex member C/D Homo sapiens 49-52 23138185-0 2013 Lenalidomide-induced upregulation of CXCR4 in CD34+ hematopoietic cells, a potential mechanism of decreased hematopoietic progenitor mobilization. Lenalidomide 0-12 C-X-C motif chemokine receptor 4 Homo sapiens 37-42 23138185-0 2013 Lenalidomide-induced upregulation of CXCR4 in CD34+ hematopoietic cells, a potential mechanism of decreased hematopoietic progenitor mobilization. Lenalidomide 0-12 CD34 molecule Homo sapiens 46-50 23632478-8 2013 CONCLUSION: Lenalidomide favours a uniform TNF-alpha and IL-8 inflammatory and IGF-1 secretory profile of myeloma cells, an observation that raises important questions for therapeutic approaches incorporating the agent. Lenalidomide 12-24 insulin like growth factor 1 Homo sapiens 79-84 23565715-0 2013 High expression of cereblon (CRBN) is associated with improved clinical response in patients with multiple myeloma treated with lenalidomide and dexamethasone. Lenalidomide 128-140 cereblon Homo sapiens 19-27 23565715-0 2013 High expression of cereblon (CRBN) is associated with improved clinical response in patients with multiple myeloma treated with lenalidomide and dexamethasone. Lenalidomide 128-140 cereblon Homo sapiens 29-33 23565715-1 2013 Cereblon (CRBN) has recently been identified as a target for immunomodulatory drugs (IMiDs) and its downregulation has been linked to resistance to lenalidomide. Lenalidomide 148-160 cereblon Homo sapiens 0-8 23565715-1 2013 Cereblon (CRBN) has recently been identified as a target for immunomodulatory drugs (IMiDs) and its downregulation has been linked to resistance to lenalidomide. Lenalidomide 148-160 cereblon Homo sapiens 10-14 23565715-2 2013 Here, we studied CRBN expression by real time polymerase chain reaction in 49 bone marrow samples of newly diagnosed patients with multiple myeloma treated with lenalidomide and dexamethasone. Lenalidomide 161-173 cereblon Homo sapiens 17-21 23325833-0 2013 Chronic lymphocytic leukemia cells induce defective LFA-1-directed T-cell motility by altering Rho GTPase signaling that is reversible with lenalidomide. Lenalidomide 140-152 integrin subunit alpha L Homo sapiens 52-57 23696885-8 2013 When Len is given in combination with Dex, PCA may be induced on endothelial cells and monocytes through TF expression and PS exposure. Lenalidomide 5-8 coagulation factor III, tissue factor Homo sapiens 105-107 23632478-8 2013 CONCLUSION: Lenalidomide favours a uniform TNF-alpha and IL-8 inflammatory and IGF-1 secretory profile of myeloma cells, an observation that raises important questions for therapeutic approaches incorporating the agent. Lenalidomide 12-24 tumor necrosis factor Homo sapiens 43-52 23632478-8 2013 CONCLUSION: Lenalidomide favours a uniform TNF-alpha and IL-8 inflammatory and IGF-1 secretory profile of myeloma cells, an observation that raises important questions for therapeutic approaches incorporating the agent. Lenalidomide 12-24 C-X-C motif chemokine ligand 8 Homo sapiens 57-61 23178378-0 2013 Immunomodulatory drugs lenalidomide and pomalidomide inhibit multiple myeloma-induced osteoclast formation and the RANKL/OPG ratio in the myeloma microenvironment targeting the expression of adhesion molecules. Lenalidomide 23-35 TNF superfamily member 11 Homo sapiens 115-120 23178378-0 2013 Immunomodulatory drugs lenalidomide and pomalidomide inhibit multiple myeloma-induced osteoclast formation and the RANKL/OPG ratio in the myeloma microenvironment targeting the expression of adhesion molecules. Lenalidomide 23-35 TNF receptor superfamily member 11b Homo sapiens 121-124 23178378-3 2013 We found that in vivo concentrations of both lenalidomide (LEN) and pomalidomide (POM) significantly blunted RANKL upregulation normalizing the RANKL/OPG ratio in human osteoprogenitor cells (PreOBs) when co-cultured with MM cells and also inhibited CCL3 production by MM cells. Lenalidomide 45-57 TNF superfamily member 11 Homo sapiens 109-114 23178378-3 2013 We found that in vivo concentrations of both lenalidomide (LEN) and pomalidomide (POM) significantly blunted RANKL upregulation normalizing the RANKL/OPG ratio in human osteoprogenitor cells (PreOBs) when co-cultured with MM cells and also inhibited CCL3 production by MM cells. Lenalidomide 45-57 TNF superfamily member 11 Homo sapiens 144-149 23252516-0 2013 Lenalidomide efficacy in activated B-cell-like subtype diffuse large B-cell lymphoma is dependent upon IRF4 and cereblon expression. Lenalidomide 0-12 interferon regulatory factor 4 Homo sapiens 103-107 22929976-0 2013 Correlation of clinical response and response duration with miR-145 induction by lenalidomide in CD34(+) cells from patients with del(5q) myelodysplastic syndrome. Lenalidomide 81-93 microRNA 145 Homo sapiens 60-67 22929976-0 2013 Correlation of clinical response and response duration with miR-145 induction by lenalidomide in CD34(+) cells from patients with del(5q) myelodysplastic syndrome. Lenalidomide 81-93 CD34 molecule Homo sapiens 97-101 22929976-1 2013 We examined whether lenalidomide exposure up-regulates miRNAs and mRNAs, previously shown to play a role in the disease phenotype of del(5q) myelodysplastic syndrome, in pre-treatment CD34(+) marrow cells. Lenalidomide 20-32 CD34 molecule Homo sapiens 184-188 22929976-7 2013 Lenalidomide selectively abrogated progenitor activity in cells depleted of miR-143 and miR-145 supporting a key role for miR-143/145 in the sensitivity to lenalidomide of del(5q) myelodysplastic syndrome patients. Lenalidomide 0-12 microRNA 143 Homo sapiens 76-83 22929976-7 2013 Lenalidomide selectively abrogated progenitor activity in cells depleted of miR-143 and miR-145 supporting a key role for miR-143/145 in the sensitivity to lenalidomide of del(5q) myelodysplastic syndrome patients. Lenalidomide 0-12 microRNA 145 Homo sapiens 88-95 22929976-7 2013 Lenalidomide selectively abrogated progenitor activity in cells depleted of miR-143 and miR-145 supporting a key role for miR-143/145 in the sensitivity to lenalidomide of del(5q) myelodysplastic syndrome patients. Lenalidomide 0-12 microRNA 143 Homo sapiens 122-129 22525275-0 2013 Lenalidomide promotes p53 degradation by inhibiting MDM2 auto-ubiquitination in myelodysplastic syndrome with chromosome 5q deletion. Lenalidomide 0-12 tumor protein p53 Homo sapiens 22-25 22525275-0 2013 Lenalidomide promotes p53 degradation by inhibiting MDM2 auto-ubiquitination in myelodysplastic syndrome with chromosome 5q deletion. Lenalidomide 0-12 MDM2 proto-oncogene Homo sapiens 52-56 22525275-4 2013 More importantly, we show that lenalidomide (Len) acts to stabilize MDM2, thereby accelerating p53 degradation. Lenalidomide 31-43 MDM2 proto-oncogene Homo sapiens 68-72 22525275-4 2013 More importantly, we show that lenalidomide (Len) acts to stabilize MDM2, thereby accelerating p53 degradation. Lenalidomide 31-43 tumor protein p53 Homo sapiens 95-98 22525275-4 2013 More importantly, we show that lenalidomide (Len) acts to stabilize MDM2, thereby accelerating p53 degradation. Lenalidomide 45-48 MDM2 proto-oncogene Homo sapiens 68-72 22525275-4 2013 More importantly, we show that lenalidomide (Len) acts to stabilize MDM2, thereby accelerating p53 degradation. Lenalidomide 45-48 tumor protein p53 Homo sapiens 95-98 23178378-3 2013 We found that in vivo concentrations of both lenalidomide (LEN) and pomalidomide (POM) significantly blunted RANKL upregulation normalizing the RANKL/OPG ratio in human osteoprogenitor cells (PreOBs) when co-cultured with MM cells and also inhibited CCL3 production by MM cells. Lenalidomide 45-57 TNF receptor superfamily member 11b Homo sapiens 150-153 23178378-3 2013 We found that in vivo concentrations of both lenalidomide (LEN) and pomalidomide (POM) significantly blunted RANKL upregulation normalizing the RANKL/OPG ratio in human osteoprogenitor cells (PreOBs) when co-cultured with MM cells and also inhibited CCL3 production by MM cells. Lenalidomide 45-57 C-C motif chemokine ligand 3 Homo sapiens 250-254 23252516-6 2013 IRF4 inhibition by lenalidomide induced downregulation of B-cell receptor (BCR)-dependent NF-kappaB. Lenalidomide 19-31 interferon regulatory factor 4 Homo sapiens 0-4 23252516-7 2013 Whereas IRF4-specific small, interfering RNA mimicked the effects of lenalidomide reducing NF-kappaB activation, IRF4 overexpression enhanced NF-kappaB activation and conferred resistance to lenalidomide. Lenalidomide 69-81 interferon regulatory factor 4 Homo sapiens 8-12 23252516-7 2013 Whereas IRF4-specific small, interfering RNA mimicked the effects of lenalidomide reducing NF-kappaB activation, IRF4 overexpression enhanced NF-kappaB activation and conferred resistance to lenalidomide. Lenalidomide 191-203 interferon regulatory factor 4 Homo sapiens 113-117 23252516-8 2013 These findings indicate the crucial role of IRF4 inhibition in lenalidomide efficacy in ABC cells. Lenalidomide 63-75 interferon regulatory factor 4 Homo sapiens 44-48 23252516-9 2013 Furthermore, lenalidomide-induced IRF4 downregulation required the expression of cereblon, a molecular target of lenalidomide. Lenalidomide 13-25 interferon regulatory factor 4 Homo sapiens 34-38 23252516-9 2013 Furthermore, lenalidomide-induced IRF4 downregulation required the expression of cereblon, a molecular target of lenalidomide. Lenalidomide 113-125 interferon regulatory factor 4 Homo sapiens 34-38 23252516-10 2013 Taken together, these findings suggest that lenalidomide has direct antitumour activity against DLBCL cells, preferentially ABC-DLBCL cells, by blocking IRF4 expression and the BCR-NF-kappaB signalling pathway in a cereblon-dependent manner. Lenalidomide 44-56 interferon regulatory factor 4 Homo sapiens 153-157 23233657-1 2013 Recently, cereblon (CRBN) expression was found to be essential for the activity of thalidomide and lenalidomide. Lenalidomide 99-111 cereblon Homo sapiens 20-24 23420263-8 2013 In addition, array-based gene expression analysis revealed that lenalidomide regulated the expression of several genes associated with cell survival, apoptosis and development, including BH3-interacting domain death agonist (BID), v-fos FBJ murine osteosarcoma viral oncogene homolog (FOS) and NK2 homeobox1 (NKX2-1). Lenalidomide 64-76 BH3 interacting domain death agonist Mus musculus 225-228 23420263-8 2013 In addition, array-based gene expression analysis revealed that lenalidomide regulated the expression of several genes associated with cell survival, apoptosis and development, including BH3-interacting domain death agonist (BID), v-fos FBJ murine osteosarcoma viral oncogene homolog (FOS) and NK2 homeobox1 (NKX2-1). Lenalidomide 64-76 Fos proto-oncogene, AP-1 transcription factor subunit Homo sapiens 233-236 23420263-8 2013 In addition, array-based gene expression analysis revealed that lenalidomide regulated the expression of several genes associated with cell survival, apoptosis and development, including BH3-interacting domain death agonist (BID), v-fos FBJ murine osteosarcoma viral oncogene homolog (FOS) and NK2 homeobox1 (NKX2-1). Lenalidomide 64-76 FBJ osteosarcoma oncogene Mus musculus 285-288 23420263-8 2013 In addition, array-based gene expression analysis revealed that lenalidomide regulated the expression of several genes associated with cell survival, apoptosis and development, including BH3-interacting domain death agonist (BID), v-fos FBJ murine osteosarcoma viral oncogene homolog (FOS) and NK2 homeobox1 (NKX2-1). Lenalidomide 64-76 NK2 homeobox 1 Mus musculus 294-307 23420263-8 2013 In addition, array-based gene expression analysis revealed that lenalidomide regulated the expression of several genes associated with cell survival, apoptosis and development, including BH3-interacting domain death agonist (BID), v-fos FBJ murine osteosarcoma viral oncogene homolog (FOS) and NK2 homeobox1 (NKX2-1). Lenalidomide 64-76 NK2 homeobox 1 Mus musculus 309-315 23420263-9 2013 BID and FOS, which are known apoptosis activators, were upregulated by lenalidomide treatment, whereas NKX2-1, which is used as an immunohistochemistry marker for NSCLC, was downregulated. Lenalidomide 71-83 BH3 interacting domain death agonist Homo sapiens 0-3 23420263-9 2013 BID and FOS, which are known apoptosis activators, were upregulated by lenalidomide treatment, whereas NKX2-1, which is used as an immunohistochemistry marker for NSCLC, was downregulated. Lenalidomide 71-83 Fos proto-oncogene, AP-1 transcription factor subunit Homo sapiens 8-11 23100311-6 2013 Furthermore, we show that lenalidomide, a drug approved for myelodysplastic syndromes and multiple myeloma, enhances translation of the C/EBPalpha-p30 isoform, resulting in higher miR-181a levels. Lenalidomide 26-38 centromere protein V Homo sapiens 147-150 23100311-6 2013 Furthermore, we show that lenalidomide, a drug approved for myelodysplastic syndromes and multiple myeloma, enhances translation of the C/EBPalpha-p30 isoform, resulting in higher miR-181a levels. Lenalidomide 26-38 microRNA 181a-2 Mus musculus 180-188 23100311-8 2013 Similarly, lenalidomide exhibits antitumorigenic activity paralleled by increased miR-181a expression. Lenalidomide 11-23 microRNA 181a-2 Mus musculus 82-90 23100311-10 2013 Altogether, our data provide a potential explanation for the improved clinical outcomes observed in CEBPA-mutated CN-AML patients, and suggest that lenalidomide treatment enhancing the C/EBPalpha-p30 protein levels and in turn miR-181a may sensitize AML blasts to chemotherapy. Lenalidomide 148-160 CCAAT enhancer binding protein alpha Homo sapiens 100-105 23100311-10 2013 Altogether, our data provide a potential explanation for the improved clinical outcomes observed in CEBPA-mutated CN-AML patients, and suggest that lenalidomide treatment enhancing the C/EBPalpha-p30 protein levels and in turn miR-181a may sensitize AML blasts to chemotherapy. Lenalidomide 148-160 CCAAT enhancer binding protein alpha Homo sapiens 185-195 23100311-10 2013 Altogether, our data provide a potential explanation for the improved clinical outcomes observed in CEBPA-mutated CN-AML patients, and suggest that lenalidomide treatment enhancing the C/EBPalpha-p30 protein levels and in turn miR-181a may sensitize AML blasts to chemotherapy. Lenalidomide 148-160 centromere protein V Homo sapiens 196-199 23100311-10 2013 Altogether, our data provide a potential explanation for the improved clinical outcomes observed in CEBPA-mutated CN-AML patients, and suggest that lenalidomide treatment enhancing the C/EBPalpha-p30 protein levels and in turn miR-181a may sensitize AML blasts to chemotherapy. Lenalidomide 148-160 microRNA 181a-2 Mus musculus 227-235 22975686-0 2013 BTK inhibitor ibrutinib is cytotoxic to myeloma and potently enhances bortezomib and lenalidomide activities through NF-kappaB. Lenalidomide 85-97 Bruton tyrosine kinase Homo sapiens 0-3 24163824-3 2013 This review discusses the potential mechanism of action and efficacy of lenalidomide, alone or in combination, in treatment of CLL and its toxic effects such as tumor lysis syndrome (TLS) and tumor flare reaction (TFR), that make its management different from other hematologic malignancies. Lenalidomide 72-84 transferrin receptor Homo sapiens 214-217 22733396-4 2013 We demonstrate that exposure to lenalidomide in the context of T-cell expansion with direct ligation of CD3/CD28 complex results in polarization toward a Th1 phenotype characterized by increased IFN-gamma, but not IL-10 expression. Lenalidomide 32-44 CD28 molecule Homo sapiens 108-112 22733396-4 2013 We demonstrate that exposure to lenalidomide in the context of T-cell expansion with direct ligation of CD3/CD28 complex results in polarization toward a Th1 phenotype characterized by increased IFN-gamma, but not IL-10 expression. Lenalidomide 32-44 interferon gamma Homo sapiens 195-204 22733396-4 2013 We demonstrate that exposure to lenalidomide in the context of T-cell expansion with direct ligation of CD3/CD28 complex results in polarization toward a Th1 phenotype characterized by increased IFN-gamma, but not IL-10 expression. Lenalidomide 32-44 interleukin 10 Homo sapiens 214-219 22522886-1 2012 Lenalidomide is a potent immunomodulatory agent capable of downregulating proinflammatory cytokines such as tumor necrosis factor-alpha (TNF-alpha) and upregulating anti-inflammatory cytokines. Lenalidomide 0-12 tumor necrosis factor Mus musculus 108-135 22552008-0 2012 Cereblon is a direct protein target for immunomodulatory and antiproliferative activities of lenalidomide and pomalidomide. Lenalidomide 93-105 cereblon Homo sapiens 0-8 22552008-3 2012 Our studies demonstrate that thalidomide, lenalidomide and another immunomodulatory drug, pomalidomide, bound endogenous CRBN and recombinant CRBN-DNA damage binding protein-1 (DDB1) complexes. Lenalidomide 42-54 cereblon Homo sapiens 121-125 22552008-3 2012 Our studies demonstrate that thalidomide, lenalidomide and another immunomodulatory drug, pomalidomide, bound endogenous CRBN and recombinant CRBN-DNA damage binding protein-1 (DDB1) complexes. Lenalidomide 42-54 damage specific DNA binding protein 1 Homo sapiens 142-175 22552008-3 2012 Our studies demonstrate that thalidomide, lenalidomide and another immunomodulatory drug, pomalidomide, bound endogenous CRBN and recombinant CRBN-DNA damage binding protein-1 (DDB1) complexes. Lenalidomide 42-54 damage specific DNA binding protein 1 Homo sapiens 177-181 22552008-4 2012 CRBN mediated antiproliferative activities of lenalidomide and pomalidomide in myeloma cells, as well as lenalidomide- and pomalidomide-induced cytokine production in T cells. Lenalidomide 46-58 cereblon Homo sapiens 0-4 22552008-5 2012 Lenalidomide and pomalidomide inhibited autoubiquitination of CRBN in HEK293T cells expressing thalidomide-binding competent wild-type CRBN, but not thalidomide-binding defective CRBN(YW/AA). Lenalidomide 0-12 cereblon Homo sapiens 62-66 22552008-5 2012 Lenalidomide and pomalidomide inhibited autoubiquitination of CRBN in HEK293T cells expressing thalidomide-binding competent wild-type CRBN, but not thalidomide-binding defective CRBN(YW/AA). Lenalidomide 0-12 cereblon Homo sapiens 135-139 22552008-5 2012 Lenalidomide and pomalidomide inhibited autoubiquitination of CRBN in HEK293T cells expressing thalidomide-binding competent wild-type CRBN, but not thalidomide-binding defective CRBN(YW/AA). Lenalidomide 0-12 cereblon Homo sapiens 135-139 22552008-7 2012 Long-term selection for lenalidomide resistance in H929 myeloma cell lines was accompanied by a reduction in CRBN, while in DF15R myeloma cells resistant to both pomalidomide and lenalidomide, CRBN protein was undetectable. Lenalidomide 24-36 cereblon Homo sapiens 109-113 22552008-7 2012 Long-term selection for lenalidomide resistance in H929 myeloma cell lines was accompanied by a reduction in CRBN, while in DF15R myeloma cells resistant to both pomalidomide and lenalidomide, CRBN protein was undetectable. Lenalidomide 24-36 cereblon Homo sapiens 193-197 22552008-8 2012 Our biophysical, biochemical and gene silencing studies show that CRBN is a proximate, therapeutically important molecular target of lenalidomide and pomalidomide. Lenalidomide 133-145 cereblon Homo sapiens 66-70 22623160-3 2012 The combination of Enzastaurin and Lenalidomide, at doses as low as 1 muM, showed strong synergism against indolent lymphomas by reducing cell growth, producing an increase in G0-G1 phase followed by significant decrease in S phase, increasing apoptosis, and inhibiting PI3K/AKT, PKC and MAPK/ERK pathways. Lenalidomide 35-47 latexin Homo sapiens 70-73 22623160-3 2012 The combination of Enzastaurin and Lenalidomide, at doses as low as 1 muM, showed strong synergism against indolent lymphomas by reducing cell growth, producing an increase in G0-G1 phase followed by significant decrease in S phase, increasing apoptosis, and inhibiting PI3K/AKT, PKC and MAPK/ERK pathways. Lenalidomide 35-47 AKT serine/threonine kinase 1 Homo sapiens 275-278 22623160-3 2012 The combination of Enzastaurin and Lenalidomide, at doses as low as 1 muM, showed strong synergism against indolent lymphomas by reducing cell growth, producing an increase in G0-G1 phase followed by significant decrease in S phase, increasing apoptosis, and inhibiting PI3K/AKT, PKC and MAPK/ERK pathways. Lenalidomide 35-47 proline rich transmembrane protein 2 Homo sapiens 280-283 22623160-3 2012 The combination of Enzastaurin and Lenalidomide, at doses as low as 1 muM, showed strong synergism against indolent lymphomas by reducing cell growth, producing an increase in G0-G1 phase followed by significant decrease in S phase, increasing apoptosis, and inhibiting PI3K/AKT, PKC and MAPK/ERK pathways. Lenalidomide 35-47 mitogen-activated protein kinase 1 Homo sapiens 293-296 22608605-1 2012 UNLABELLED: We used microarray profiling to investigate the direct effects of lenalidomide on gene expression in isolated CD14(+) monocytes from 6 patients with del(5q). Lenalidomide 78-90 CD14 molecule Homo sapiens 122-126 22608605-2 2012 Our data demonstrate that changes in genes involved the tumor necrosis factor (TNF) signaling pathway and the bone marrow stroma, suggesting that treatment with lenalidomide may help restore the damaged niche and suppress the TNF signaling pathway. Lenalidomide 161-173 tumor necrosis factor Homo sapiens 56-77 22608605-2 2012 Our data demonstrate that changes in genes involved the tumor necrosis factor (TNF) signaling pathway and the bone marrow stroma, suggesting that treatment with lenalidomide may help restore the damaged niche and suppress the TNF signaling pathway. Lenalidomide 161-173 tumor necrosis factor Homo sapiens 79-82 22608605-2 2012 Our data demonstrate that changes in genes involved the tumor necrosis factor (TNF) signaling pathway and the bone marrow stroma, suggesting that treatment with lenalidomide may help restore the damaged niche and suppress the TNF signaling pathway. Lenalidomide 161-173 tumor necrosis factor Homo sapiens 226-229 22608605-4 2012 MATERIALS AND METHODS: We used gene expression profiling to study the effect of lenalidomide therapy in peripheral blood CD14(+) monocytes of 6 patients with del(5q) and MDS. Lenalidomide 80-92 CD14 molecule Homo sapiens 121-125 22608605-5 2012 RESULTS: After lenalidomide treatment, genes involved in the tumor necrosis factor (TNF) signaling pathway that were upregulated in the patients before treatment decreased to the healthy control baseline expression level. Lenalidomide 15-27 tumor necrosis factor Homo sapiens 61-82 22608605-5 2012 RESULTS: After lenalidomide treatment, genes involved in the tumor necrosis factor (TNF) signaling pathway that were upregulated in the patients before treatment decreased to the healthy control baseline expression level. Lenalidomide 15-27 tumor necrosis factor Homo sapiens 84-87 23100311-0 2013 Lenalidomide-mediated enhanced translation of C/EBPalpha-p30 protein up-regulates expression of the antileukemic microRNA-181a in acute myeloid leukemia. Lenalidomide 0-12 CCAAT enhancer binding protein alpha Homo sapiens 46-56 23100311-0 2013 Lenalidomide-mediated enhanced translation of C/EBPalpha-p30 protein up-regulates expression of the antileukemic microRNA-181a in acute myeloid leukemia. Lenalidomide 0-12 centromere protein V Homo sapiens 57-60 23100311-6 2013 Furthermore, we show that lenalidomide, a drug approved for myelodysplastic syndromes and multiple myeloma, enhances translation of the C/EBPalpha-p30 isoform, resulting in higher miR-181a levels. Lenalidomide 26-38 CCAAT enhancer binding protein alpha Homo sapiens 136-146 22933333-9 2012 Our findings also suggest that sequencing of the TP53 gene should be included in the study of patients with del(5q) as a single abnormality or in complex karyotype before lenalidomide treatment. Lenalidomide 171-183 tumor protein p53 Homo sapiens 49-53 23012246-7 2012 RSK2(Ser227) inhibition resulting from BI-D1870 treatment restored lenalidomide-induced direct cytotoxicity of myeloma cells from interleukin-6-mediated cell protection, showed no cross-resistance to bortezomib, and exerted additive/synergistic antiproliferative effects in conjunction with the mTOR, histone deacetylase, and BH3-mimicking BCL2/BCLX(L) inhibitors. Lenalidomide 67-79 ribosomal protein S6 kinase A3 Homo sapiens 0-4 23012246-7 2012 RSK2(Ser227) inhibition resulting from BI-D1870 treatment restored lenalidomide-induced direct cytotoxicity of myeloma cells from interleukin-6-mediated cell protection, showed no cross-resistance to bortezomib, and exerted additive/synergistic antiproliferative effects in conjunction with the mTOR, histone deacetylase, and BH3-mimicking BCL2/BCLX(L) inhibitors. Lenalidomide 67-79 interleukin 6 Homo sapiens 130-143 23012246-7 2012 RSK2(Ser227) inhibition resulting from BI-D1870 treatment restored lenalidomide-induced direct cytotoxicity of myeloma cells from interleukin-6-mediated cell protection, showed no cross-resistance to bortezomib, and exerted additive/synergistic antiproliferative effects in conjunction with the mTOR, histone deacetylase, and BH3-mimicking BCL2/BCLX(L) inhibitors. Lenalidomide 67-79 mechanistic target of rapamycin kinase Homo sapiens 295-299 23012246-7 2012 RSK2(Ser227) inhibition resulting from BI-D1870 treatment restored lenalidomide-induced direct cytotoxicity of myeloma cells from interleukin-6-mediated cell protection, showed no cross-resistance to bortezomib, and exerted additive/synergistic antiproliferative effects in conjunction with the mTOR, histone deacetylase, and BH3-mimicking BCL2/BCLX(L) inhibitors. Lenalidomide 67-79 BCL2 apoptosis regulator Homo sapiens 340-344 23012246-7 2012 RSK2(Ser227) inhibition resulting from BI-D1870 treatment restored lenalidomide-induced direct cytotoxicity of myeloma cells from interleukin-6-mediated cell protection, showed no cross-resistance to bortezomib, and exerted additive/synergistic antiproliferative effects in conjunction with the mTOR, histone deacetylase, and BH3-mimicking BCL2/BCLX(L) inhibitors. Lenalidomide 67-79 BCL2 like 1 Homo sapiens 345-349 22936658-1 2012 The erythropoietic effects of lenalidomide are cytokine dependent, suggesting that the erythroid hematologic improvement (HI-E) rate may be augmented by combined treatment (CT) with recombinant human erythropoietin (rhu-EPO) in myelodysplastic syndrome (MDS). Lenalidomide 30-42 erythropoietin Homo sapiens 200-214 22936658-1 2012 The erythropoietic effects of lenalidomide are cytokine dependent, suggesting that the erythroid hematologic improvement (HI-E) rate may be augmented by combined treatment (CT) with recombinant human erythropoietin (rhu-EPO) in myelodysplastic syndrome (MDS). Lenalidomide 30-42 erythropoietin Homo sapiens 220-223 22522886-1 2012 Lenalidomide is a potent immunomodulatory agent capable of downregulating proinflammatory cytokines such as tumor necrosis factor-alpha (TNF-alpha) and upregulating anti-inflammatory cytokines. Lenalidomide 0-12 tumor necrosis factor Mus musculus 137-146 22522886-9 2012 ob/ob mice displayed elevated serum TNF-alpha and IL-6 levels, fat composition and glucose intolerance, the effects of which except glucose intolerance and fat composition were attenuated by lenalidomide. Lenalidomide 191-203 tumor necrosis factor Mus musculus 36-45 22522886-9 2012 ob/ob mice displayed elevated serum TNF-alpha and IL-6 levels, fat composition and glucose intolerance, the effects of which except glucose intolerance and fat composition were attenuated by lenalidomide. Lenalidomide 191-203 interleukin 6 Mus musculus 50-54 22766978-0 2012 Multiple myeloma cells expressing low levels of CD138 have an immature phenotype and reduced sensitivity to lenalidomide. Lenalidomide 120-132 syndecan 1 Homo sapiens 48-53 22150117-0 2012 Severe dermatologic adverse reactions after exposure to lenalidomide in multiple myeloma patients with a positive HLA-DRB1*1501 and HLA-DQB1*0602. Lenalidomide 56-68 major histocompatibility complex, class II, DR beta 1 Homo sapiens 114-122 22150117-0 2012 Severe dermatologic adverse reactions after exposure to lenalidomide in multiple myeloma patients with a positive HLA-DRB1*1501 and HLA-DQB1*0602. Lenalidomide 56-68 major histocompatibility complex, class II, DQ beta 1 Homo sapiens 132-140 22766978-10 2012 CD138- cells sorted from the KYMM-2 cell line also showed high BCL6, low IRF4 expression and decreased sensitivity to lenalidomide compared with CD138+ cells. Lenalidomide 130-142 syndecan 1 Homo sapiens 0-5 22766978-11 2012 Our observations suggest that low CD138 expression relates to i) poor prognosis, ii) immature phenotype and iii) low sensitivity to lenalidomide. Lenalidomide 156-168 syndecan 1 Homo sapiens 34-39 22640031-2 2012 Preclinical data showed that the Akt inhibitor, perifosine, sensitized MM cells to lenalidomide and dexamethasone, providing the rationale for this Phase I, multicentre, single-arm study to assess the safety and determine the maximum-tolerated dose (MTD) of perifosine-lenalidomide-dexamethasone in relapsed and relapsed/refractory MM. Lenalidomide 83-95 AKT serine/threonine kinase 1 Homo sapiens 33-36 22640031-2 2012 Preclinical data showed that the Akt inhibitor, perifosine, sensitized MM cells to lenalidomide and dexamethasone, providing the rationale for this Phase I, multicentre, single-arm study to assess the safety and determine the maximum-tolerated dose (MTD) of perifosine-lenalidomide-dexamethasone in relapsed and relapsed/refractory MM. Lenalidomide 269-281 AKT serine/threonine kinase 1 Homo sapiens 33-36 21750922-5 2012 Despite their inhibition of CFU formation, perifosine, bortezomib and lenalidomide induced only slight or moderate cytotoxicity in CD34(+) selected HPC, as assessed using different assays such as flow cytometry-based detection of activated caspases and immunohistochemistry studies (e.g., Ki-67 staining). Lenalidomide 70-82 CD34 molecule Homo sapiens 131-135 22774993-0 2012 Lenalidomide enhancement of human T cell functions in human immunodeficiency virus (HIV)-infected and HIV-negative CD4 T lymphocytopenic patients. Lenalidomide 0-12 CD4 molecule Homo sapiens 115-118 22774993-5 2012 Significant enhancement of chemotaxis to S1P and CCL21 was induced by 100-1000 nM lenalidomide only for normal T cells at a low CD4 : CD8 ratio. Lenalidomide 82-94 C-C motif chemokine ligand 21 Homo sapiens 49-54 22774993-5 2012 Significant enhancement of chemotaxis to S1P and CCL21 was induced by 100-1000 nM lenalidomide only for normal T cells at a low CD4 : CD8 ratio. Lenalidomide 82-94 CD4 molecule Homo sapiens 128-131 22774993-5 2012 Significant enhancement of chemotaxis to S1P and CCL21 was induced by 100-1000 nM lenalidomide only for normal T cells at a low CD4 : CD8 ratio. Lenalidomide 82-94 CD8a molecule Homo sapiens 134-137 22774993-7 2012 Lenalidomide at 30-1000 nM significantly enhanced chemotaxis to S1P and IL-2 generation for T cells from HIV-negative CD4 T lymphocytopenic patients as from HIV-infected patients, with less effect on CCL21-elicited chemotaxis and none for IFN-gamma generation. Lenalidomide 0-12 interleukin 2 Homo sapiens 72-76 22774993-7 2012 Lenalidomide at 30-1000 nM significantly enhanced chemotaxis to S1P and IL-2 generation for T cells from HIV-negative CD4 T lymphocytopenic patients as from HIV-infected patients, with less effect on CCL21-elicited chemotaxis and none for IFN-gamma generation. Lenalidomide 0-12 CD4 molecule Homo sapiens 118-121 22774993-7 2012 Lenalidomide at 30-1000 nM significantly enhanced chemotaxis to S1P and IL-2 generation for T cells from HIV-negative CD4 T lymphocytopenic patients as from HIV-infected patients, with less effect on CCL21-elicited chemotaxis and none for IFN-gamma generation. Lenalidomide 0-12 interferon gamma Homo sapiens 239-248 22474168-4 2012 Unexpectedly, A2A and beta2AR agonists also synergize with melphalan, lenalidomide, bortezomib, and doxorubicin. Lenalidomide 70-82 immunoglobulin kappa variable 2D-29 Homo sapiens 14-17 22474168-4 2012 Unexpectedly, A2A and beta2AR agonists also synergize with melphalan, lenalidomide, bortezomib, and doxorubicin. Lenalidomide 70-82 adrenoceptor beta 2 Homo sapiens 22-29 22617388-6 2012 Furthermore, we explored the expression of HO-1 in multiple myeloma cells in response to the key anti-myeloma drugs bortezomib and lenalidomide. Lenalidomide 131-143 heme oxygenase 1 Homo sapiens 43-47 22617388-8 2012 Moreover, we also observe that HO-1 is increased in lenalidomide-resistant MM cell lines. Lenalidomide 52-64 heme oxygenase 1 Homo sapiens 31-35 22698399-3 2012 Lenalidomide kills ABC DLBCL cells by augmenting interferon beta (IFNbeta) production, owing to the oncogenic MYD88 mutations in these lymphomas. Lenalidomide 0-12 interferon beta 1 Homo sapiens 49-64 22698399-3 2012 Lenalidomide kills ABC DLBCL cells by augmenting interferon beta (IFNbeta) production, owing to the oncogenic MYD88 mutations in these lymphomas. Lenalidomide 0-12 interferon beta 1 Homo sapiens 66-73 22488443-5 2012 Given the high serum and plasma levels of VEGF observed in POEMS patients, the use of anti-angiogenetic drugs such as thalidomide and lenalidomide and other drugs with anti-VEGF and anti-TNF effect such as bortezomib have been considered to treat this syndrome. Lenalidomide 134-146 vascular endothelial growth factor A Homo sapiens 42-46 22488443-5 2012 Given the high serum and plasma levels of VEGF observed in POEMS patients, the use of anti-angiogenetic drugs such as thalidomide and lenalidomide and other drugs with anti-VEGF and anti-TNF effect such as bortezomib have been considered to treat this syndrome. Lenalidomide 134-146 vascular endothelial growth factor A Homo sapiens 173-177 22488443-5 2012 Given the high serum and plasma levels of VEGF observed in POEMS patients, the use of anti-angiogenetic drugs such as thalidomide and lenalidomide and other drugs with anti-VEGF and anti-TNF effect such as bortezomib have been considered to treat this syndrome. Lenalidomide 134-146 tumor necrosis factor Homo sapiens 187-190 22698399-2 2012 Lenalidomide is an active agent in the activated B cell-like (ABC) subtype of diffuse large B cell lymphoma (DLBCL), but its mechanism of action is unknown. Lenalidomide 0-12 ATP binding cassette subfamily B member 6 (Langereis blood group) Homo sapiens 62-65 22698399-3 2012 Lenalidomide kills ABC DLBCL cells by augmenting interferon beta (IFNbeta) production, owing to the oncogenic MYD88 mutations in these lymphomas. Lenalidomide 0-12 ATP binding cassette subfamily B member 6 (Langereis blood group) Homo sapiens 19-22 21968939-5 2012 RESULTS: Lenalidomide decreased the IC(50) of docetaxel by up to 50% (P < 0.05) and also decreased invasion in PC3, LNCaP, and DU145 cells and anchorage independent growth in PC3 cells (P < 0.01). Lenalidomide 9-21 chromobox 8 Homo sapiens 114-117 21968939-5 2012 RESULTS: Lenalidomide decreased the IC(50) of docetaxel by up to 50% (P < 0.05) and also decreased invasion in PC3, LNCaP, and DU145 cells and anchorage independent growth in PC3 cells (P < 0.01). Lenalidomide 9-21 chromobox 8 Homo sapiens 178-181 21968939-6 2012 Apoptosis in lenalidomide/docetaxel-treated cells was increased by 2.2-fold over single agent docetaxel and a corresponding increase in p53, p38, and BAD activation was observed in Western blots (P < 0.001). Lenalidomide 13-25 tumor protein p53 Homo sapiens 136-139 21968939-6 2012 Apoptosis in lenalidomide/docetaxel-treated cells was increased by 2.2-fold over single agent docetaxel and a corresponding increase in p53, p38, and BAD activation was observed in Western blots (P < 0.001). Lenalidomide 13-25 mitogen-activated protein kinase 14 Homo sapiens 141-144 21968939-7 2012 When PC3 challenged mice were treated with lenalidomide and docetaxel, median survival increased from 48 to 59 days and the rate of tumor growth was significantly reduced (P < 0.05). Lenalidomide 43-55 chromobox 8 Mus musculus 5-8 22474251-7 2012 These findings in the Rhoh(-/-) CLL cells were subsequently demonstrated to closely resemble changes in GTPase activation observed in human CLL samples after in vitro and in vivo treatment with lenalidomide, an agent with known influence on microenvironment protection, and suggest that RhoH plays a critical role in prosurvival CLL cell-cell and cell-microenvironment interactions with this agent. Lenalidomide 194-206 ras homolog family member H Homo sapiens 22-26 22474251-7 2012 These findings in the Rhoh(-/-) CLL cells were subsequently demonstrated to closely resemble changes in GTPase activation observed in human CLL samples after in vitro and in vivo treatment with lenalidomide, an agent with known influence on microenvironment protection, and suggest that RhoH plays a critical role in prosurvival CLL cell-cell and cell-microenvironment interactions with this agent. Lenalidomide 194-206 ras homolog family member H Homo sapiens 287-291 22698399-3 2012 Lenalidomide kills ABC DLBCL cells by augmenting interferon beta (IFNbeta) production, owing to the oncogenic MYD88 mutations in these lymphomas. Lenalidomide 0-12 MYD88 innate immune signal transduction adaptor Homo sapiens 110-115 22698399-4 2012 In a cereblon-dependent fashion, lenalidomide downregulates IRF4 and SPIB, transcription factors that together prevent IFNbeta production by repressing IRF7 and amplify prosurvival NF-kappaB signaling by transactivating CARD11. Lenalidomide 33-45 interferon regulatory factor 4 Homo sapiens 60-64 22698399-4 2012 In a cereblon-dependent fashion, lenalidomide downregulates IRF4 and SPIB, transcription factors that together prevent IFNbeta production by repressing IRF7 and amplify prosurvival NF-kappaB signaling by transactivating CARD11. Lenalidomide 33-45 Spi-B transcription factor Homo sapiens 69-73 22698399-4 2012 In a cereblon-dependent fashion, lenalidomide downregulates IRF4 and SPIB, transcription factors that together prevent IFNbeta production by repressing IRF7 and amplify prosurvival NF-kappaB signaling by transactivating CARD11. Lenalidomide 33-45 interferon beta 1 Homo sapiens 119-126 22698399-4 2012 In a cereblon-dependent fashion, lenalidomide downregulates IRF4 and SPIB, transcription factors that together prevent IFNbeta production by repressing IRF7 and amplify prosurvival NF-kappaB signaling by transactivating CARD11. Lenalidomide 33-45 interferon regulatory factor 7 Homo sapiens 152-156 22698399-4 2012 In a cereblon-dependent fashion, lenalidomide downregulates IRF4 and SPIB, transcription factors that together prevent IFNbeta production by repressing IRF7 and amplify prosurvival NF-kappaB signaling by transactivating CARD11. Lenalidomide 33-45 caspase recruitment domain family member 11 Homo sapiens 220-226 22698399-5 2012 Blockade of B cell receptor signaling using the BTK inhibitor ibrutinib also downregulates IRF4 and consequently synergizes with lenalidomide in killing ABC DLBCLs, suggesting attractive therapeutic strategies. Lenalidomide 129-141 Bruton tyrosine kinase Homo sapiens 48-51 22698399-5 2012 Blockade of B cell receptor signaling using the BTK inhibitor ibrutinib also downregulates IRF4 and consequently synergizes with lenalidomide in killing ABC DLBCLs, suggesting attractive therapeutic strategies. Lenalidomide 129-141 ATP binding cassette subfamily B member 6 (Langereis blood group) Homo sapiens 153-156 22826791-2 2012 The common expression of vascular endothelial growth factor (VEGF) and its receptor in HEH provide a rationale for the reported use of antiangiogenic drugs, including bevacizumab, lenalidomide and thalidomide. Lenalidomide 180-192 vascular endothelial growth factor A Homo sapiens 25-59 22826791-2 2012 The common expression of vascular endothelial growth factor (VEGF) and its receptor in HEH provide a rationale for the reported use of antiangiogenic drugs, including bevacizumab, lenalidomide and thalidomide. Lenalidomide 180-192 vascular endothelial growth factor A Homo sapiens 61-65 22171982-0 2012 Downregulation of BCL2 by AT-101 enhances the antileukaemic effect of lenalidomide both by an immune dependant and independent manner. Lenalidomide 70-82 BCL2 apoptosis regulator Homo sapiens 18-22 22171982-3 2012 Lenalidomide, an immunomodulator, is clinically effective in CLL and can enhance the anti-CLL effects of CD20 targeting monoclonal antibody, rituximab. Lenalidomide 0-12 keratin 20 Homo sapiens 105-109 21080211-2 2012 The in vitro cytotoxicity of perifosine, bortezomib and lenalidomide against 6 cell lines derived from hematological malignancies was investigated using trypan blue staining, flow cytometry-based detection of activated caspases, Annexin V assays, immunohistochemistry studies (KI-67 and caspase-3 staining) and the immature-myeloid-information (IMI) technique. Lenalidomide 56-68 annexin A5 Homo sapiens 229-238 21080211-2 2012 The in vitro cytotoxicity of perifosine, bortezomib and lenalidomide against 6 cell lines derived from hematological malignancies was investigated using trypan blue staining, flow cytometry-based detection of activated caspases, Annexin V assays, immunohistochemistry studies (KI-67 and caspase-3 staining) and the immature-myeloid-information (IMI) technique. Lenalidomide 56-68 caspase 3 Homo sapiens 287-296 21520044-7 2012 In addition, the pretreatment of LLL12 blocked the promotion of the cell proliferation and resistance to lenalidomide by IL-6. Lenalidomide 105-117 interleukin 6 Homo sapiens 121-125 21748755-0 2012 Lenalidomide modulates IL-8 and anti-prostate antibody levels in men with biochemically recurrent prostate cancer. Lenalidomide 0-12 C-X-C motif chemokine ligand 8 Homo sapiens 23-27 21748755-6 2012 RESULTS: Treatment with lenalidomide was associated with global changes in immunoreactivity to a number of prostate-associated antigens, as well as with changes in circulating levels of the T(H) 2 cytokines IL-4, IL-5, IL-10, and IL-13. Lenalidomide 24-36 interleukin 4 Homo sapiens 207-211 21748755-6 2012 RESULTS: Treatment with lenalidomide was associated with global changes in immunoreactivity to a number of prostate-associated antigens, as well as with changes in circulating levels of the T(H) 2 cytokines IL-4, IL-5, IL-10, and IL-13. Lenalidomide 24-36 interleukin 5 Homo sapiens 213-217 21748755-6 2012 RESULTS: Treatment with lenalidomide was associated with global changes in immunoreactivity to a number of prostate-associated antigens, as well as with changes in circulating levels of the T(H) 2 cytokines IL-4, IL-5, IL-10, and IL-13. Lenalidomide 24-36 interleukin 10 Homo sapiens 219-224 21748755-6 2012 RESULTS: Treatment with lenalidomide was associated with global changes in immunoreactivity to a number of prostate-associated antigens, as well as with changes in circulating levels of the T(H) 2 cytokines IL-4, IL-5, IL-10, and IL-13. Lenalidomide 24-36 interleukin 13 Homo sapiens 230-235 23556117-4 2012 Other recent advances include a better understanding of the pathogenesis of disease including haplodeficiency of several candidate genes, and elucidation of the lenalidomide-specific effect on two phosphatases ultimately leading to p53 degradation in the erythroid progenitors and cell cycle arrest in earlier myeloid progenitors. Lenalidomide 161-173 tumor protein p53 Homo sapiens 232-235 22301101-4 2012 In particular, GDF15 conferred resistance to melphalan, bortezomib, and to a lesser extent, lenalidomide in both stroma-dependent and stroma-independent multiple myeloma cells. Lenalidomide 92-104 growth differentiation factor 15 Homo sapiens 15-20 22261445-0 2012 p53 nuclear expression correlates with hemizygous TP53 deletion and predicts an adverse outcome for patients with relapsed/refractory multiple myeloma treated with lenalidomide. Lenalidomide 164-176 tumor protein p53 Homo sapiens 0-3 22261445-1 2012 del(17p13)(TP53) seems to be an independent poor prognostic factor in patients with relapsed/refractory multiple myeloma (MM) receiving lenalidomide. Lenalidomide 136-148 tumor protein p53 Homo sapiens 11-15 22261445-2 2012 However, whether aberrant p53 nuclear expression detected by immunohistochemical analysis can be used as a surrogate marker for del(17p13)(TP53) in prognostic evaluation of lenalidomide-treated relapsed/refractory MM remains unclear. Lenalidomide 173-185 tumor protein p53 Homo sapiens 26-29 22261445-7 2012 Our results suggest that p53 nuclear expression is associated with adverse outcome in patients with relapsed/refractory MM receiving lenalidomide-based therapy and that p53 immunohistochemical analysis may serve as a simple, rapid method to predict del(17p13)(TP53) in this patient subgroup. Lenalidomide 133-145 tumor protein p53 Homo sapiens 25-28 22109882-0 2012 The immunomodulatory drug lenalidomide restores a vitamin D sensitive phenotype to the vitamin D resistant breast cancer cell line MDA-MB-231 through inhibition of BCL-2: potential for breast cancer therapeutics. Lenalidomide 26-38 BCL2 apoptosis regulator Homo sapiens 164-169 22109882-8 2012 Whereas lenalidomide alone had no effect on cell growth, a 50% inhibition of cell growth by 1,25-D3 was achieved with additional 1 muM lenalidomide in resistant cells. Lenalidomide 135-147 latexin Homo sapiens 131-134 22109882-10 2012 An apoptosis protein array showed that the 1,25-D3 and lenalidomide combination increased pro-apoptotic proteins (phosphorylated p53) and decreased BCL-2 expression. Lenalidomide 55-67 tumor protein p53 Homo sapiens 129-132 22109882-10 2012 An apoptosis protein array showed that the 1,25-D3 and lenalidomide combination increased pro-apoptotic proteins (phosphorylated p53) and decreased BCL-2 expression. Lenalidomide 55-67 BCL2 apoptosis regulator Homo sapiens 148-153 22001752-9 2012 In conclusion, salvage mobilization with plerixafor plus G-CSF is successful in the majority of patients with MM previously treated with lenalidomide. Lenalidomide 137-149 colony stimulating factor 3 Homo sapiens 57-62 21860026-0 2011 Cereblon expression is required for the antimyeloma activity of lenalidomide and pomalidomide. Lenalidomide 64-76 cereblon Homo sapiens 0-8 22792112-8 2012 Preliminary data regarding the role of lenalidomide in addition to chemoimmunotherapy (R-CHOP) in first line clinical trials were discussed; data of safety, feasibility and efficacy were promising. Lenalidomide 39-51 DNA damage inducible transcript 3 Homo sapiens 89-93 22919394-5 2012 The molecular mechanisms and targets of lenalidomide remain largely unknown, but recent evidence shows cereblon (CRBN) as a possible mediator of its therapeutical effects. Lenalidomide 40-52 cereblon Homo sapiens 113-117 22997574-3 2012 Lenalidomide has the potential to invoke changes in TNF-alpha with less toxicity than thalidomide. Lenalidomide 0-12 tumor necrosis factor Homo sapiens 52-61 22997574-4 2012 This pilot study evaluated lenalidomide at reduction of TNF-alpha and improvement of behavior and language in children with autism with elevated TNF-alpha. Lenalidomide 27-39 tumor necrosis factor Homo sapiens 56-65 22997574-4 2012 This pilot study evaluated lenalidomide at reduction of TNF-alpha and improvement of behavior and language in children with autism with elevated TNF-alpha. Lenalidomide 27-39 tumor necrosis factor Homo sapiens 145-154 22997574-5 2012 Subjects with elevated TNF-alpha were given 2.5 mgs lenalidomide daily for 12-weeks. Lenalidomide 52-64 tumor necrosis factor Homo sapiens 23-32 22666210-0 2012 PSA Response to Lenalidomide Therapy in a Pre-Treated Patient with Metastatic Prostate Cancer Refractory to Hormones and Chemotherapy: A Case Report. Lenalidomide 16-28 aminopeptidase puromycin sensitive Homo sapiens 0-3 22666210-4 2012 In this paper, we report that lenalidomide showed antitumoral activity in a patient with HRPC and bone metastases pre-treated with chemotherapy, decreased the PSA level and improved the patient"s health status for the first 5 months. Lenalidomide 30-42 aminopeptidase puromycin sensitive Homo sapiens 159-162 22571700-2 2012 For a long period of time, the standard therapy for MDS was hematopoietic stem cell transplantation, however DNA methyltransferase inhibitors (DNMT inhibitors) including decitabine (DAC) and azacitidine (AZA), and lenalidomide, a derivative of thalidomide have been highlighted as new chemotherapeutic agents for MDS. Lenalidomide 214-226 DNA methyltransferase 1 Homo sapiens 143-147 22161851-0 2011 Effect of combined dexamethasone/lenalidomide therapy on NK cell-receptor levels in myeloma patients. Lenalidomide 33-45 killer cell immunoglobulin like receptor, two Ig domains and long cytoplasmic tail 4 Homo sapiens 57-73 21860026-3 2011 CRBN depletion is initially cytotoxic to human myeloma cells, but surviving cells with stable CRBN depletion become highly resistant to both lenalidomide and pomalidomide, but not to the unrelated drugs bortezomib, dexamethasone, and melphalan. Lenalidomide 141-153 cereblon Homo sapiens 0-4 21860026-4 2011 Acquired deletion of CRBN was found to be the primary genetic event differentiating isogenic MM1.S cell lines cultured to be sensitive or resistant to lenalidomide and pomalidomide. Lenalidomide 151-163 cereblon Homo sapiens 21-25 21802139-0 2011 Anti-myelin associated glycoprotein neuropathy responding to lenalidomide. Lenalidomide 61-73 myelin associated glycoprotein Homo sapiens 5-35 21812019-0 2011 Inflammation, TNFalpha and endothelial dysfunction link lenalidomide to venous thrombosis in chronic lymphocytic leukemia. Lenalidomide 56-68 tumor necrosis factor Homo sapiens 14-22 21812019-10 2011 In patients with lenalidomide-related DVTs, TNFalpha, and sVCAM-1 were more strongly upregulated than in all other patients (p < 0.05) and TNFalpha and sVCAM-1 levels were significantly correlated (r = 0.65, p < 0.001). Lenalidomide 17-29 tumor necrosis factor Homo sapiens 44-52 21812019-10 2011 In patients with lenalidomide-related DVTs, TNFalpha, and sVCAM-1 were more strongly upregulated than in all other patients (p < 0.05) and TNFalpha and sVCAM-1 levels were significantly correlated (r = 0.65, p < 0.001). Lenalidomide 17-29 tumor necrosis factor Homo sapiens 142-150 21812019-11 2011 These data link lenalidomide associated DVTs with TNFalpha upregulation and endothelial cell dysfunction and suggest that aspirin may have a role for DVT prophylaxis in these patients. Lenalidomide 16-28 tumor necrosis factor Homo sapiens 50-58 22110196-9 2011 Western blot analysis showed lenalidomide significantly reduced pERK expression in all cell lines (p<0.05). Lenalidomide 29-41 eukaryotic translation initiation factor 2 alpha kinase 3 Homo sapiens 64-68 22010965-5 2011 Lenalidomide was unable to directly induce apoptosis in CLL cells in vitro, however it modulated costimulatory (CD80, CD83, CD86) surface molecules on CLL cells in vitro and in vivo. Lenalidomide 0-12 CD80 molecule Homo sapiens 112-116 22010965-5 2011 Lenalidomide was unable to directly induce apoptosis in CLL cells in vitro, however it modulated costimulatory (CD80, CD83, CD86) surface molecules on CLL cells in vitro and in vivo. Lenalidomide 0-12 CD83 molecule Homo sapiens 118-122 22010965-5 2011 Lenalidomide was unable to directly induce apoptosis in CLL cells in vitro, however it modulated costimulatory (CD80, CD83, CD86) surface molecules on CLL cells in vitro and in vivo. Lenalidomide 0-12 CD86 molecule Homo sapiens 124-128 22010965-7 2011 Cytokine analysis showed increase in levels of TNF-alpha post-lenalidomide treatment, consistent with acute inflammatory reaction. Lenalidomide 62-74 tumor necrosis factor Homo sapiens 47-56 22010965-8 2011 Furthermore, the basal cytokine profile (high IL-8, MIG, IP-10 and IL-4 levels and low IL-5, MIP1a, MIP1b, IL12/p70) was predictive of clinical response to lenalidomide. Lenalidomide 156-168 C-X-C motif chemokine ligand 8 Homo sapiens 46-50 22010965-8 2011 Furthermore, the basal cytokine profile (high IL-8, MIG, IP-10 and IL-4 levels and low IL-5, MIP1a, MIP1b, IL12/p70) was predictive of clinical response to lenalidomide. Lenalidomide 156-168 C-X-C motif chemokine ligand 9 Homo sapiens 52-55 22010965-8 2011 Furthermore, the basal cytokine profile (high IL-8, MIG, IP-10 and IL-4 levels and low IL-5, MIP1a, MIP1b, IL12/p70) was predictive of clinical response to lenalidomide. Lenalidomide 156-168 C-X-C motif chemokine ligand 10 Homo sapiens 57-62 22010965-8 2011 Furthermore, the basal cytokine profile (high IL-8, MIG, IP-10 and IL-4 levels and low IL-5, MIP1a, MIP1b, IL12/p70) was predictive of clinical response to lenalidomide. Lenalidomide 156-168 interleukin 4 Homo sapiens 67-71 22010965-8 2011 Furthermore, the basal cytokine profile (high IL-8, MIG, IP-10 and IL-4 levels and low IL-5, MIP1a, MIP1b, IL12/p70) was predictive of clinical response to lenalidomide. Lenalidomide 156-168 interleukin 5 Homo sapiens 87-91 22010965-8 2011 Furthermore, the basal cytokine profile (high IL-8, MIG, IP-10 and IL-4 levels and low IL-5, MIP1a, MIP1b, IL12/p70) was predictive of clinical response to lenalidomide. Lenalidomide 156-168 C-C motif chemokine ligand 3 Homo sapiens 93-98 22010965-8 2011 Furthermore, the basal cytokine profile (high IL-8, MIG, IP-10 and IL-4 levels and low IL-5, MIP1a, MIP1b, IL12/p70) was predictive of clinical response to lenalidomide. Lenalidomide 156-168 C-C motif chemokine ligand 4 Homo sapiens 100-105 22010965-8 2011 Furthermore, the basal cytokine profile (high IL-8, MIG, IP-10 and IL-4 levels and low IL-5, MIP1a, MIP1b, IL12/p70) was predictive of clinical response to lenalidomide. Lenalidomide 156-168 annexin A6 Homo sapiens 112-115 21860026-5 2011 Gene expression changes induced by lenalidomide were dramatically suppressed in the presence of CRBN depletion, further demonstrating that CRBN is required for lenalidomide activity. Lenalidomide 35-47 cereblon Homo sapiens 96-100 21860026-5 2011 Gene expression changes induced by lenalidomide were dramatically suppressed in the presence of CRBN depletion, further demonstrating that CRBN is required for lenalidomide activity. Lenalidomide 35-47 cereblon Homo sapiens 139-143 21860026-5 2011 Gene expression changes induced by lenalidomide were dramatically suppressed in the presence of CRBN depletion, further demonstrating that CRBN is required for lenalidomide activity. Lenalidomide 160-172 cereblon Homo sapiens 96-100 21860026-5 2011 Gene expression changes induced by lenalidomide were dramatically suppressed in the presence of CRBN depletion, further demonstrating that CRBN is required for lenalidomide activity. Lenalidomide 160-172 cereblon Homo sapiens 139-143 21860026-6 2011 Downstream targets of CRBN include interferon regulatory factor 4 (IRF4) previously reported to also be a target of lenalidomide. Lenalidomide 116-128 cereblon Homo sapiens 22-26 21860026-6 2011 Downstream targets of CRBN include interferon regulatory factor 4 (IRF4) previously reported to also be a target of lenalidomide. Lenalidomide 116-128 interferon regulatory factor 4 Homo sapiens 35-65 21860026-6 2011 Downstream targets of CRBN include interferon regulatory factor 4 (IRF4) previously reported to also be a target of lenalidomide. Lenalidomide 116-128 interferon regulatory factor 4 Homo sapiens 67-71 21860026-7 2011 Patients exposed to, and putatively resistant to, lenalidomide had lower CRBN levels in paired samples before and after therapy. Lenalidomide 50-62 cereblon Homo sapiens 73-77 21523725-10 2011 CONCLUSIONS: TFR is a unique immune-mediated phenomenon noted with lenalidomide treatment only in patients with CLL that correlates with clinical response. Lenalidomide 67-79 transferrin receptor Homo sapiens 13-16 21630308-0 2011 Lenalidomide, cyclophosphamide and dexamethasone (CRd) for newly diagnosed multiple myeloma: results from a phase 2 trial. Lenalidomide 0-12 cone-rod homeobox Homo sapiens 50-53 21707574-0 2011 Lenalidomide downregulates the cell survival factor, interferon regulatory factor-4, providing a potential mechanistic link for predicting response. Lenalidomide 0-12 interferon regulatory factor 4 Homo sapiens 53-83 21707574-4 2011 This study showed that lenalidomide downregulated IRF4 levels in MM cell lines and bone marrow samples within 8 h of drug exposure. Lenalidomide 23-35 interferon regulatory factor 4 Homo sapiens 50-54 21707574-6 2011 In eight MM cell lines, high IRF4 levels correlated with increased lenalidomide sensitivity. Lenalidomide 67-79 interferon regulatory factor 4 Homo sapiens 29-33 21707574-8 2011 Among MM patients with high levels of IRF4 expression, treatment with lenalidomide led to a significantly longer overall survival than other therapies in a retrospective analysis. Lenalidomide 70-82 interferon regulatory factor 4 Homo sapiens 38-42 21707574-9 2011 These data confirm the central role of IRF4 in MM pathogenesis; indicate that this is an important mechanism by which lenalidomide exerts its antitumour effects; and may provide a mechanistic biomarker to predict response to lenalidomide. Lenalidomide 118-130 interferon regulatory factor 4 Homo sapiens 39-43 21707574-9 2011 These data confirm the central role of IRF4 in MM pathogenesis; indicate that this is an important mechanism by which lenalidomide exerts its antitumour effects; and may provide a mechanistic biomarker to predict response to lenalidomide. Lenalidomide 225-237 interferon regulatory factor 4 Homo sapiens 39-43 21677134-0 2011 Lenalidomide enhances antigen-specific activity and decreases CD45RA expression of T cells from patients with multiple myeloma. Lenalidomide 0-12 protein tyrosine phosphatase receptor type C Homo sapiens 62-66 21677134-1 2011 The aim of this study was to investigate whether the specific T cell response against the multiple myeloma Ag HM1.24 is enhanced by the immunomodulatory drug lenalidomide (Revlimid). Lenalidomide 158-170 cholinergic receptor muscarinic 1 Homo sapiens 110-113 21677134-1 2011 The aim of this study was to investigate whether the specific T cell response against the multiple myeloma Ag HM1.24 is enhanced by the immunomodulatory drug lenalidomide (Revlimid). Lenalidomide 172-180 cholinergic receptor muscarinic 1 Homo sapiens 110-113 21677134-5 2011 We found that activation of HM1.24-specific T cells from healthy donors and patients with plasma cell dyscrasias was enhanced significantly by lenalidomide and furthermore that the impact of lenalidomide on T cells depends on the duration of the exposure. Lenalidomide 143-155 cholinergic receptor muscarinic 1 Homo sapiens 28-31 21677134-5 2011 We found that activation of HM1.24-specific T cells from healthy donors and patients with plasma cell dyscrasias was enhanced significantly by lenalidomide and furthermore that the impact of lenalidomide on T cells depends on the duration of the exposure. Lenalidomide 191-203 cholinergic receptor muscarinic 1 Homo sapiens 28-31 21677134-6 2011 Notably, lenalidomide supports the downregulation of CD45RA on T cells upon activation, observed in healthy donors and in patients in vitro and also in patients during lenalidomide therapy in vivo. Lenalidomide 9-21 protein tyrosine phosphatase receptor type C Homo sapiens 53-57 21677134-6 2011 Notably, lenalidomide supports the downregulation of CD45RA on T cells upon activation, observed in healthy donors and in patients in vitro and also in patients during lenalidomide therapy in vivo. Lenalidomide 168-180 protein tyrosine phosphatase receptor type C Homo sapiens 53-57 21677134-7 2011 We showed for the first time, to our knowledge, that lenalidomide enhances the Ag-specific activation of T cells and the subsequent downregulation of CD45RA expression of T cells in vitro and in vivo. Lenalidomide 53-65 protein tyrosine phosphatase receptor type C Homo sapiens 150-154 21856771-9 2011 In mice treated with lenalidomide, BNKL levels of active caspase-3 were significantly augmented, thus showing proapoptotic and cytotoxic effects of this drug in vivo. Lenalidomide 21-33 caspase 3 Mus musculus 57-66 21825263-2 2011 The preclinical combination of lenalidomide with the mTOR inhibitor CCI-779 has displayed synergy in vitro and represents a novel combination in MM. Lenalidomide 31-43 mechanistic target of rapamycin kinase Homo sapiens 53-57 21825263-9 2011 Detailed mechanistic interrogation of this pharmacokinetic interaction demonstrated that lenalidomide was an ABCB1 (P-glycoprotein [P-gp]) substrate. Lenalidomide 89-101 ATP binding cassette subfamily B member 1 Homo sapiens 109-114 21825263-9 2011 Detailed mechanistic interrogation of this pharmacokinetic interaction demonstrated that lenalidomide was an ABCB1 (P-glycoprotein [P-gp]) substrate. Lenalidomide 89-101 ATP binding cassette subfamily B member 1 Homo sapiens 116-130 21825263-9 2011 Detailed mechanistic interrogation of this pharmacokinetic interaction demonstrated that lenalidomide was an ABCB1 (P-glycoprotein [P-gp]) substrate. Lenalidomide 89-101 ATP binding cassette subfamily B member 1 Homo sapiens 132-136 21825263-11 2011 Pharmacokinetic and clinical interactions between lenalidomide and CCI-779 seemed to occur, with in vitro data indicating lenalidomide was an ABCB1 (P-gp) substrate. Lenalidomide 122-134 ATP binding cassette subfamily B member 1 Homo sapiens 142-147 21825263-11 2011 Pharmacokinetic and clinical interactions between lenalidomide and CCI-779 seemed to occur, with in vitro data indicating lenalidomide was an ABCB1 (P-gp) substrate. Lenalidomide 122-134 ATP binding cassette subfamily B member 1 Homo sapiens 149-153 21825263-12 2011 To our knowledge, this is the first report of a clinically significant P-gp-based drug-drug interaction with lenalidomide. Lenalidomide 109-121 ATP binding cassette subfamily B member 1 Homo sapiens 71-75 21658626-7 2011 Other agents inhibit various cellular pathways including those triggered by the B-cell receptor, including spleen tyrosine kinase (Syk) and Bruton"s tyrosine kinase, and other intracellular pathways such as the mammalian target of rapamycin (mTOR), PI3-kinase, and apoptosis, and drugs that target the tumor microenvironment, notably the immunomodulatory agent lenalidomide. Lenalidomide 361-373 mechanistic target of rapamycin kinase Homo sapiens 211-240 21347656-2 2011 MP plus thalidomide (MPT), bortezomib (VMP), or lenalidomide (MPR), as induction plus maintenance, have proved to be superior to MP and are currently the treatment of choice for this population. Lenalidomide 48-60 progesterone receptor membrane component 1 Homo sapiens 62-65 21551245-6 2011 Moreover, E2A knockdown synergizes with the immunomodulatory drug lenalidomide to reduce CLL viability. Lenalidomide 66-78 transcription factor 3 Homo sapiens 10-13 21523725-1 2011 BACKGROUND: In patients with chronic lymphocytic leukemia (CLL), treatment with lenalidomide induces a unique, previously uncharacterized, immune response called tumor flare reaction (TFR). Lenalidomide 80-92 transferrin receptor Homo sapiens 184-187 21523726-0 2011 Chromosomal aberrations +1q21 and del(17p13) predict survival in patients with recurrent multiple myeloma treated with lenalidomide and dexamethasone. Lenalidomide 119-131 H3 histone pseudogene 6 Homo sapiens 40-43 21307145-7 2011 Lenalidomide-treated MMECs showed changes in VEGF/VEGFR2 signaling pathway and several proteins controlling EC motility, cytoskeleton remodeling, and energy metabolism pathways. Lenalidomide 0-12 vascular endothelial growth factor A Homo sapiens 45-49 21389327-6 2011 Down-regulation of IRF4 by lenalidomide was confirmed by longitudinal studies of bone marrow samples from 23 patients obtained before and during lenalidomide treatment using CD138+/IRF4+ double labeling. Lenalidomide 27-39 interferon regulatory factor 4 Homo sapiens 19-23 21389327-6 2011 Down-regulation of IRF4 by lenalidomide was confirmed by longitudinal studies of bone marrow samples from 23 patients obtained before and during lenalidomide treatment using CD138+/IRF4+ double labeling. Lenalidomide 27-39 interferon regulatory factor 4 Homo sapiens 181-185 21389327-6 2011 Down-regulation of IRF4 by lenalidomide was confirmed by longitudinal studies of bone marrow samples from 23 patients obtained before and during lenalidomide treatment using CD138+/IRF4+ double labeling. Lenalidomide 145-157 interferon regulatory factor 4 Homo sapiens 19-23 21321360-6 2011 Importantly, lenalidomide decreased the percentage and clonogenicity of SP cells, and also induced phosphorylation changes in Akt, GSK-3alpha/beta, MEK1, c-Jun, p53, and p70S6K in SP cells. Lenalidomide 13-25 AKT serine/threonine kinase 1 Homo sapiens 126-129 21321360-6 2011 Importantly, lenalidomide decreased the percentage and clonogenicity of SP cells, and also induced phosphorylation changes in Akt, GSK-3alpha/beta, MEK1, c-Jun, p53, and p70S6K in SP cells. Lenalidomide 13-25 glycogen synthase kinase 3 alpha Homo sapiens 131-141 21321360-6 2011 Importantly, lenalidomide decreased the percentage and clonogenicity of SP cells, and also induced phosphorylation changes in Akt, GSK-3alpha/beta, MEK1, c-Jun, p53, and p70S6K in SP cells. Lenalidomide 13-25 mitogen-activated protein kinase kinase 1 Homo sapiens 148-152 21321360-6 2011 Importantly, lenalidomide decreased the percentage and clonogenicity of SP cells, and also induced phosphorylation changes in Akt, GSK-3alpha/beta, MEK1, c-Jun, p53, and p70S6K in SP cells. Lenalidomide 13-25 Jun proto-oncogene, AP-1 transcription factor subunit Homo sapiens 154-159 21321360-6 2011 Importantly, lenalidomide decreased the percentage and clonogenicity of SP cells, and also induced phosphorylation changes in Akt, GSK-3alpha/beta, MEK1, c-Jun, p53, and p70S6K in SP cells. Lenalidomide 13-25 tumor protein p53 Homo sapiens 161-164 21321360-6 2011 Importantly, lenalidomide decreased the percentage and clonogenicity of SP cells, and also induced phosphorylation changes in Akt, GSK-3alpha/beta, MEK1, c-Jun, p53, and p70S6K in SP cells. Lenalidomide 13-25 ribosomal protein S6 kinase B1 Homo sapiens 170-176 21378270-3 2011 Here we demonstrate that lenalidomide activation of CLL cells depends on the phosphatidylinositol 3-kinase p110delta (PI3K-delta) pathway. Lenalidomide 25-37 phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit delta Homo sapiens 77-116 21378270-3 2011 Here we demonstrate that lenalidomide activation of CLL cells depends on the phosphatidylinositol 3-kinase p110delta (PI3K-delta) pathway. Lenalidomide 25-37 phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit delta Homo sapiens 118-128 21378270-4 2011 Inhibition of PI3K-delta signaling by the PI3K-delta-inhibiting drug, CAL-101, or by siRNA knockdown of p110delta, abrogates CLL cell activation, costimulatory molecule expression, and vascular endothelial growth factor and basic fibroblast growth factor gene expression that is induced by lenalidomide. Lenalidomide 290-302 phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit delta Homo sapiens 14-24 21378270-4 2011 Inhibition of PI3K-delta signaling by the PI3K-delta-inhibiting drug, CAL-101, or by siRNA knockdown of p110delta, abrogates CLL cell activation, costimulatory molecule expression, and vascular endothelial growth factor and basic fibroblast growth factor gene expression that is induced by lenalidomide. Lenalidomide 290-302 phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit delta Homo sapiens 42-52 21378270-4 2011 Inhibition of PI3K-delta signaling by the PI3K-delta-inhibiting drug, CAL-101, or by siRNA knockdown of p110delta, abrogates CLL cell activation, costimulatory molecule expression, and vascular endothelial growth factor and basic fibroblast growth factor gene expression that is induced by lenalidomide. Lenalidomide 290-302 phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit delta Homo sapiens 104-113 21378270-6 2011 Collectively, these data demonstrate the importance of PI3K-delta signaling for lenalidomide immune modulation. Lenalidomide 80-92 phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit delta Homo sapiens 55-65 21307145-7 2011 Lenalidomide-treated MMECs showed changes in VEGF/VEGFR2 signaling pathway and several proteins controlling EC motility, cytoskeleton remodeling, and energy metabolism pathways. Lenalidomide 0-12 kinase insert domain receptor Homo sapiens 50-56 21189262-0 2011 Evidence of a role for activation of Wnt/beta-catenin signaling in the resistance of plasma cells to lenalidomide. Lenalidomide 101-113 catenin beta 1 Homo sapiens 41-53 21575928-3 2011 The type of anti-MM therapy represents a key factor, with a substantially elevated VTE risk in patients treated with the immunomodulatory drugs (IMiDs) thalidomide or lenalidomide in combination with dexamethasone and/or chemotherapy; VTE risk with lenalidomide-dexamethasone is further increased with concomitant erythropoietin. Lenalidomide 167-179 erythropoietin Homo sapiens 314-328 21189262-7 2011 Components of the beta-catenin destruction complex were also impacted by lenalidomide, which suppressed casein kinase 1alpha expression while augmenting glycogen synthase kinase 3alpha/beta phosphorylation. Lenalidomide 73-85 catenin beta 1 Homo sapiens 18-30 21189262-8 2011 Stimulation of Wnt/beta-catenin signaling with recombinant Wnt-3a, or by overexpression of beta-catenin, reduced the anti-proliferative activity of lenalidomide. Lenalidomide 148-160 catenin beta 1 Homo sapiens 19-31 21189262-8 2011 Stimulation of Wnt/beta-catenin signaling with recombinant Wnt-3a, or by overexpression of beta-catenin, reduced the anti-proliferative activity of lenalidomide. Lenalidomide 148-160 Wnt family member 3A Homo sapiens 59-65 21189262-8 2011 Stimulation of Wnt/beta-catenin signaling with recombinant Wnt-3a, or by overexpression of beta-catenin, reduced the anti-proliferative activity of lenalidomide. Lenalidomide 148-160 catenin beta 1 Homo sapiens 91-103 21189262-9 2011 Conversely, suppression of beta-catenin with small hairpin RNAs restored plasma cell sensitivity to lenalidomide. Lenalidomide 100-112 catenin beta 1 Homo sapiens 27-39 21189262-10 2011 Together, these findings support the hypothesis that lenalidomide mediates activation of Wnt/beta-catenin signaling in plasma cells as a mechanism of inducible chemoresistance through effects at the transcriptional and post-translational levels. Lenalidomide 53-65 catenin beta 1 Homo sapiens 93-105 20421869-2 2011 Recent reports have suggested decreases in the number of CD34+ cells collected and increases in the failure rate among patients whose initial therapy contained lenalidomide when mobilized with G-CSF alone. Lenalidomide 160-172 colony stimulating factor 3 Homo sapiens 193-198 20479709-8 2011 These data suggest that CD34+ hematopoietic stem cells can be successfully and predictably collected with combination plerixafor plus G-CSF for primary or secondary mobilization in the majority of patients with MM who have been previously treated with lenalidomide. Lenalidomide 252-264 CD34 molecule Homo sapiens 24-28 21159364-6 2011 RESULTS: At THL and LEN concentrations resembling those observed in myeloma patients in vivo and in the presence of tumor necrosis factor-alpha (TNFalpha) we observed significantly increased TF activity in human umbilical vein endothelial cells in vitro. Lenalidomide 20-23 coagulation factor III, tissue factor Homo sapiens 191-193 21159364-10 2011 CONCLUSIONS: Our in vitro data support the hypothesis that THL and LEN induce a hypercoagulable state through increased endothelial TF expression. Lenalidomide 67-70 coagulation factor III, tissue factor Homo sapiens 132-134 20978269-7 2011 Lymphocyte subset depletion experiments revealed that lenalidomide"s enhancement of NK cell-mediated cytotoxicity was mediated by CD4(+) T-cell production of interleukin 2 and that dexamethasone acted by suppressing interleukin-2 production. Lenalidomide 54-66 CD4 molecule Homo sapiens 130-133 20978269-7 2011 Lymphocyte subset depletion experiments revealed that lenalidomide"s enhancement of NK cell-mediated cytotoxicity was mediated by CD4(+) T-cell production of interleukin 2 and that dexamethasone acted by suppressing interleukin-2 production. Lenalidomide 54-66 interleukin 2 Homo sapiens 158-171 20978269-7 2011 Lymphocyte subset depletion experiments revealed that lenalidomide"s enhancement of NK cell-mediated cytotoxicity was mediated by CD4(+) T-cell production of interleukin 2 and that dexamethasone acted by suppressing interleukin-2 production. Lenalidomide 54-66 interleukin 2 Homo sapiens 216-229 20978269-8 2011 Similarly, the reduced ability of NK cells from patients with MM to respond to lenalidomide was also due to impaired CD4 T-cell function. Lenalidomide 79-91 CD4 molecule Homo sapiens 117-120 21215621-1 2011 Several thalidomide analogues were synthesized and compared to thalidomide and its more active analogue, lenalidomide, for their ability to inhibit the production of the pro-inflammatory cytokine tumour necrosis factor (TNF)-alpha and interleukin (IL)-6 by LPS-activated peripheral blood mononuclear cells (PBMCs). Lenalidomide 105-117 tumor necrosis factor Homo sapiens 196-230 21215621-1 2011 Several thalidomide analogues were synthesized and compared to thalidomide and its more active analogue, lenalidomide, for their ability to inhibit the production of the pro-inflammatory cytokine tumour necrosis factor (TNF)-alpha and interleukin (IL)-6 by LPS-activated peripheral blood mononuclear cells (PBMCs). Lenalidomide 105-117 interleukin 6 Homo sapiens 235-253 21130040-2 2011 At 0.03 muM to 1 muM, lenalidomide enhanced generation of IL-2 and IFN-gamma by T cell receptor-stimulated T cells of young subjects up to respective maximum increases of 17-fold and three-fold, but at 0.3 muM and 1 muM suppressed IL-17 generation. Lenalidomide 22-34 latexin Homo sapiens 8-11 21130040-2 2011 At 0.03 muM to 1 muM, lenalidomide enhanced generation of IL-2 and IFN-gamma by T cell receptor-stimulated T cells of young subjects up to respective maximum increases of 17-fold and three-fold, but at 0.3 muM and 1 muM suppressed IL-17 generation. Lenalidomide 22-34 latexin Homo sapiens 17-20 21130040-2 2011 At 0.03 muM to 1 muM, lenalidomide enhanced generation of IL-2 and IFN-gamma by T cell receptor-stimulated T cells of young subjects up to respective maximum increases of 17-fold and three-fold, but at 0.3 muM and 1 muM suppressed IL-17 generation. Lenalidomide 22-34 interleukin 2 Homo sapiens 58-62 21130040-2 2011 At 0.03 muM to 1 muM, lenalidomide enhanced generation of IL-2 and IFN-gamma by T cell receptor-stimulated T cells of young subjects up to respective maximum increases of 17-fold and three-fold, but at 0.3 muM and 1 muM suppressed IL-17 generation. Lenalidomide 22-34 interferon gamma Homo sapiens 67-76 21130040-2 2011 At 0.03 muM to 1 muM, lenalidomide enhanced generation of IL-2 and IFN-gamma by T cell receptor-stimulated T cells of young subjects up to respective maximum increases of 17-fold and three-fold, but at 0.3 muM and 1 muM suppressed IL-17 generation. Lenalidomide 22-34 latexin Homo sapiens 17-20 21130040-2 2011 At 0.03 muM to 1 muM, lenalidomide enhanced generation of IL-2 and IFN-gamma by T cell receptor-stimulated T cells of young subjects up to respective maximum increases of 17-fold and three-fold, but at 0.3 muM and 1 muM suppressed IL-17 generation. Lenalidomide 22-34 latexin Homo sapiens 17-20 21130040-2 2011 At 0.03 muM to 1 muM, lenalidomide enhanced generation of IL-2 and IFN-gamma by T cell receptor-stimulated T cells of young subjects up to respective maximum increases of 17-fold and three-fold, but at 0.3 muM and 1 muM suppressed IL-17 generation. Lenalidomide 22-34 interleukin 17A Homo sapiens 231-236 21130040-3 2011 The same concentrations of lenalidomide enhanced IL-2 and IFN-gamma generation by stimulated T cells of old subjects more, with greater respective maximal increases of up to 120-fold and six-fold, without suppressing IL-17 generation. Lenalidomide 27-39 interleukin 2 Homo sapiens 49-53 21130040-3 2011 The same concentrations of lenalidomide enhanced IL-2 and IFN-gamma generation by stimulated T cells of old subjects more, with greater respective maximal increases of up to 120-fold and six-fold, without suppressing IL-17 generation. Lenalidomide 27-39 interferon gamma Homo sapiens 58-67 21130040-4 2011 Lenalidomide enhanced proliferation and suppressed apoptosis of stimulated T cells from old subjects, by IL-2-dependent mechanisms, and restored diminished T cell chemotactic responses to CCL21 and sphingosine 1-phosphate. Lenalidomide 0-12 interleukin 2 Homo sapiens 105-109 21130040-4 2011 Lenalidomide enhanced proliferation and suppressed apoptosis of stimulated T cells from old subjects, by IL-2-dependent mechanisms, and restored diminished T cell chemotactic responses to CCL21 and sphingosine 1-phosphate. Lenalidomide 0-12 C-C motif chemokine ligand 21 Homo sapiens 188-193 21273172-9 2011 When controlling for the number of previous antimyeloma therapies, beta2-microglobulin levels, and Durie-Salmon stage (which adversely affected survival in these patients), continued lenalidomide treatment (HR, 0.137; 95% CI, 0.045-0.417; P=.0005) or each additional cycle of lenalidomide (HR, 0.921; 95% CI, 0.886-0.957; P<.0001) were both associated with longer survival. Lenalidomide 183-195 beta-2-microglobulin Homo sapiens 67-86 21322779-6 2011 Disease-specific sensitivity to lenalidomide results from the drug"s inhibitory effect on two haplodeficient phosphatases, PP2Acalpha and CDC25c, which are coregulators of the G(2)/M checkpoint. Lenalidomide 32-44 cell division cycle 25C Homo sapiens 138-144 23556075-9 2011 Correlative studies and/or retrospective analysis of lenalidomide-treated patients had identified several biomarkers associated with clinical endpoints in CLL (i.e. changes in tumor necrosis factor alpha [TNF-alpha] or vascular endothelial growth factor [VEGF] levels) or DLBCL (non-GCB phenotype) patients, but need to be validated. Lenalidomide 53-65 tumor necrosis factor Homo sapiens 176-203 23556075-9 2011 Correlative studies and/or retrospective analysis of lenalidomide-treated patients had identified several biomarkers associated with clinical endpoints in CLL (i.e. changes in tumor necrosis factor alpha [TNF-alpha] or vascular endothelial growth factor [VEGF] levels) or DLBCL (non-GCB phenotype) patients, but need to be validated. Lenalidomide 53-65 tumor necrosis factor Homo sapiens 205-214 20848094-8 2011 The extent of ADCC and its enhancement by lenalidomide correlated with NK cell expression of NKG2D and DNAM-1, and tumor cell expression of PVR and MIC-A. Lenalidomide 42-54 CD226 molecule Homo sapiens 103-109 20848094-11 2011 Lenalidomide enhances the ability of IgG1-isotype antibodies to mediate ADCC of solid tumor cells, the extent of which is largely dependent on NKG2D-NKG2D ligand interactions, but appears to be independent of MIC-A/B. Lenalidomide 0-12 killer cell lectin like receptor K1 Homo sapiens 143-148 20848094-11 2011 Lenalidomide enhances the ability of IgG1-isotype antibodies to mediate ADCC of solid tumor cells, the extent of which is largely dependent on NKG2D-NKG2D ligand interactions, but appears to be independent of MIC-A/B. Lenalidomide 0-12 killer cell lectin like receptor K1 Homo sapiens 149-154 22003441-7 2011 Intriguingly, a new role for CRBN has been identified, i.e, the binding of immunomodulatory drugs (IMiDs), e.g. thalidomide, to CRBN has now been associated with teratogenicity and also the cytotoxicity of IMiDs, including lenalidomide, which are widely used to treat multiple myeloma patients. Lenalidomide 223-235 cereblon Homo sapiens 29-33 22003441-7 2011 Intriguingly, a new role for CRBN has been identified, i.e, the binding of immunomodulatory drugs (IMiDs), e.g. thalidomide, to CRBN has now been associated with teratogenicity and also the cytotoxicity of IMiDs, including lenalidomide, which are widely used to treat multiple myeloma patients. Lenalidomide 223-235 cereblon Homo sapiens 128-132 21127185-0 2010 Increases in B-type natriuretic peptide (BNP) during treatment with lenalidomide in AL amyloidosis. Lenalidomide 68-80 natriuretic peptide B Homo sapiens 13-39 20848094-8 2011 The extent of ADCC and its enhancement by lenalidomide correlated with NK cell expression of NKG2D and DNAM-1, and tumor cell expression of PVR and MIC-A. Lenalidomide 42-54 killer cell lectin like receptor K1 Homo sapiens 93-98 21127185-0 2010 Increases in B-type natriuretic peptide (BNP) during treatment with lenalidomide in AL amyloidosis. Lenalidomide 68-80 natriuretic peptide B Homo sapiens 41-44 19965642-0 2010 Lenalidomide treatment promotes CD154 expression on CLL cells and enhances production of antibodies by normal B cells through a PI3-kinase-dependent pathway. Lenalidomide 0-12 CD40 ligand Homo sapiens 32-37 20689431-6 2010 The chemokine receptor 4 (CXCR4) inhibitor, AMD-3100, is recommended for patients who have received lenalidomide and failed to mobilize stem cells after G-SCF and cyclophosphamide. Lenalidomide 100-112 C-X-C motif chemokine receptor 4 Homo sapiens 26-31 20491881-0 2010 Changes in RPS14 expression levels during lenalidomide treatment in Low- and Intermediate-1-risk myelodysplastic syndromes with chromosome 5q deletion. Lenalidomide 42-54 ribosomal protein S14 Homo sapiens 11-16 20594187-6 2010 Several drugs effective for the treatment of MM, including proteasome inhibitors, thalidomide, lenalidomide and arsenic trioxide, block NF-kappaB activation. Lenalidomide 95-107 nuclear factor kappa B subunit 1 Homo sapiens 136-145 20846100-0 2010 Response to lenalidomide in a patient with myelodysplastic syndrome with isolated del(5q) and JAK2 V617F mutation. Lenalidomide 12-24 Janus kinase 2 Homo sapiens 94-98 20460501-6 2010 We show that lenalidomide down-regulates PD-L1 on primary MM cells and may augment CT-011"s enhancement of NK-cell function against MM. Lenalidomide 13-25 CD274 molecule Homo sapiens 41-46 20194893-0 2010 Efficacy of single-agent lenalidomide in patients with JAK2 (V617F) mutated refractory anemia with ring sideroblasts and thrombocytosis. Lenalidomide 25-37 Janus kinase 2 Homo sapiens 55-59 19965642-4 2010 Here, we demonstrate that lenalidomide induces expression of functional CD154 antigen on CLL cells both in vitro and in vivo. Lenalidomide 26-38 CD40 ligand Homo sapiens 72-77 19965642-7 2010 In CLL patients receiving lenalidomide, similar evidence of CD154 activation is observed including BID, DR5, and p73 induction and also development of anti-ROR1 tumor-directed antibodies. Lenalidomide 26-38 CD40 ligand Homo sapiens 60-65 19965642-7 2010 In CLL patients receiving lenalidomide, similar evidence of CD154 activation is observed including BID, DR5, and p73 induction and also development of anti-ROR1 tumor-directed antibodies. Lenalidomide 26-38 BH3 interacting domain death agonist Homo sapiens 99-102 19965642-7 2010 In CLL patients receiving lenalidomide, similar evidence of CD154 activation is observed including BID, DR5, and p73 induction and also development of anti-ROR1 tumor-directed antibodies. Lenalidomide 26-38 TNF receptor superfamily member 10b Homo sapiens 104-107 19965642-7 2010 In CLL patients receiving lenalidomide, similar evidence of CD154 activation is observed including BID, DR5, and p73 induction and also development of anti-ROR1 tumor-directed antibodies. Lenalidomide 26-38 tumor protein p73 Homo sapiens 113-116 19965642-7 2010 In CLL patients receiving lenalidomide, similar evidence of CD154 activation is observed including BID, DR5, and p73 induction and also development of anti-ROR1 tumor-directed antibodies. Lenalidomide 26-38 receptor tyrosine kinase like orphan receptor 1 Homo sapiens 156-160 19965642-8 2010 Our data demonstrate that lenalidomide promotes CD154 expression on CLL cells with subsequent activation phenotype, and may therefore reverse the humoral immune defect observed in this disease. Lenalidomide 26-38 CD40 ligand Homo sapiens 48-53 19965623-2 2010 We show that IMiDs down-regulate PU.1, a key transcription factor involved in granulocyte differentiation in vitro and in patients treated with lenalidomide. Lenalidomide 144-156 Spi-1 proto-oncogene Homo sapiens 33-37 20359632-5 2010 In del(5q) patients, lenalidomide suppresses the clone by inhibiting the nuclear sequestration of the haplodeficient cell cycle regulatory protein cdc25c, thereby promoting selective G2 arrest and apoptosis. Lenalidomide 21-33 cell division cycle 25C Homo sapiens 147-153 20088798-2 2010 The effect of dexamethasone on the immunomodulatory activities of lenalidomide such as T cell and natural killer (NK) cell activation was measured via interleukin [IL]-2 production, and interferon-gamma and granzyme B production respectively. Lenalidomide 66-78 granzyme B Homo sapiens 207-217 20088798-5 2010 Mechanistically, lenalidomide plus dexamethasone synergistically induced expression of the tumor suppressor genes Egr1, Egr2, Egr3, p15, p21, and p27 in MM cell lines and MM patient cells. Lenalidomide 17-29 early growth response 1 Homo sapiens 114-118 20088798-5 2010 Mechanistically, lenalidomide plus dexamethasone synergistically induced expression of the tumor suppressor genes Egr1, Egr2, Egr3, p15, p21, and p27 in MM cell lines and MM patient cells. Lenalidomide 17-29 early growth response 2 Homo sapiens 120-124 20088798-5 2010 Mechanistically, lenalidomide plus dexamethasone synergistically induced expression of the tumor suppressor genes Egr1, Egr2, Egr3, p15, p21, and p27 in MM cell lines and MM patient cells. Lenalidomide 17-29 early growth response 3 Homo sapiens 126-130 20088798-5 2010 Mechanistically, lenalidomide plus dexamethasone synergistically induced expression of the tumor suppressor genes Egr1, Egr2, Egr3, p15, p21, and p27 in MM cell lines and MM patient cells. Lenalidomide 17-29 cyclin dependent kinase inhibitor 2B Homo sapiens 132-135 20088798-5 2010 Mechanistically, lenalidomide plus dexamethasone synergistically induced expression of the tumor suppressor genes Egr1, Egr2, Egr3, p15, p21, and p27 in MM cell lines and MM patient cells. Lenalidomide 17-29 H3 histone pseudogene 16 Homo sapiens 137-140 20088798-5 2010 Mechanistically, lenalidomide plus dexamethasone synergistically induced expression of the tumor suppressor genes Egr1, Egr2, Egr3, p15, p21, and p27 in MM cell lines and MM patient cells. Lenalidomide 17-29 interferon alpha inducible protein 27 Homo sapiens 146-149 20088798-7 2010 Lenalidomide alone increased T cell production of IL-2, and NK cell production of interferon-gamma and granzyme B. Lenalidomide 0-12 interleukin 2 Homo sapiens 50-54 20088798-7 2010 Lenalidomide alone increased T cell production of IL-2, and NK cell production of interferon-gamma and granzyme B. Lenalidomide 0-12 interferon gamma Homo sapiens 82-98 20088798-7 2010 Lenalidomide alone increased T cell production of IL-2, and NK cell production of interferon-gamma and granzyme B. Lenalidomide 0-12 granzyme B Homo sapiens 103-113 19773257-4 2010 RESULTS: Compared to baseline, lenalidomide treatment significantly decreased the proportion of bone marrow CD34+ cells, increased the proportion of CD36(+)/GlycoA(+) and CD36(-)/GlycoA(+) erythroid cells and the percentage of apoptotic cells within these cell compartments. Lenalidomide 31-43 CD34 molecule Homo sapiens 108-112 19773257-8 2010 CONCLUSIONS: The beneficial effect of lenalidomide in patients with lower risk myelodysplastic syndrome with del(5q) is associated with significant increases in the proportion of bone marrow erythroid precursor cells and in the frequency of clonogenic progenitor cells, a substantial improvement in the hematopoiesis-supporting potential of bone marrow stroma and significant alterations in the adhesion profile of bone marrow CD34(+) cells. Lenalidomide 38-50 CD34 molecule Homo sapiens 427-431 19584825-0 2010 Lenalidomide as salvage therapy after allo-SCT for multiple myeloma is effective and leads to an increase of activated NK (NKp44(+)) and T (HLA-DR(+)) cells. Lenalidomide 0-12 natural cytotoxicity triggering receptor 2 Homo sapiens 123-128 19584825-9 2010 Immunomonitoring after lenalidomide showed significant increase of activated NK (NKp44(+)) and T (HLA-DR(+)) cells, as well as regulatory T cells (CD4(+), CD25(+), CD127(lo)), supporting an immunomodulating anti-myeloma effect of lenalidomide. Lenalidomide 23-35 natural cytotoxicity triggering receptor 2 Homo sapiens 81-86 19965674-5 2010 Furthermore, studies using biochemical inhibitors of caspase-8 versus caspase-9 demonstrate that NPI-0052 plus lenalidomide-triggered apoptosis is primarily dependent on caspase-8 signaling. Lenalidomide 111-123 caspase 8 Homo sapiens 53-62 19965674-5 2010 Furthermore, studies using biochemical inhibitors of caspase-8 versus caspase-9 demonstrate that NPI-0052 plus lenalidomide-triggered apoptosis is primarily dependent on caspase-8 signaling. Lenalidomide 111-123 caspase 9 Homo sapiens 70-79 19965674-5 2010 Furthermore, studies using biochemical inhibitors of caspase-8 versus caspase-9 demonstrate that NPI-0052 plus lenalidomide-triggered apoptosis is primarily dependent on caspase-8 signaling. Lenalidomide 111-123 caspase 8 Homo sapiens 170-179 20621290-0 2010 Lenalidomide down regulates the production of interferon-gamma and the expression of inhibitory cytotoxic receptors of human Natural Killer cells. Lenalidomide 0-12 interferon gamma Homo sapiens 46-62 20621290-2 2010 The effect of Lenalidomide towards Peripheral Blood Mononuclear Cells (PBMC) has been studied and direct effects towards T cells have been described, such as an increase of interferon-gamma (IFN-gamma) and interleukin (IL)-2 production. Lenalidomide 14-26 interferon gamma Homo sapiens 173-189 20621290-5 2010 Here we show that Lenalidomide can inhibit the production of IFN-gamma by NK cells from healthy donors. Lenalidomide 18-30 interferon gamma Homo sapiens 61-70 19738071-0 2009 Pomalidomide and lenalidomide induce p21 WAF-1 expression in both lymphoma and multiple myeloma through a LSD1-mediated epigenetic mechanism. Lenalidomide 17-29 cyclin dependent kinase inhibitor 1A Homo sapiens 37-40 19733563-6 2009 We previously reported the neuroprotective effects of lenalidomide, when treatment was started 2 months prior to onset of disease in the G93A SOD1 transgenic mouse model of ALS. Lenalidomide 54-66 superoxide dismutase 1, soluble Mus musculus 142-146 19733563-7 2009 Since in ALS patients, treatment can only begin after the appearance of symptoms, we sought to determine the efficacy of lenalidomide administration starting at symptom onset in the G93A SOD1 mice. Lenalidomide 121-133 superoxide dismutase 1 Homo sapiens 187-191 19733563-10 2009 Qualitative histological analysis showed that lenalidomide treatment modestly reduced the expression of the proinflammatory cytokines Fas Ligand, IL-1beta, TNF-alpha and CD40 ligand. Lenalidomide 46-58 Fas ligand (TNF superfamily, member 6) Mus musculus 134-144 19733563-10 2009 Qualitative histological analysis showed that lenalidomide treatment modestly reduced the expression of the proinflammatory cytokines Fas Ligand, IL-1beta, TNF-alpha and CD40 ligand. Lenalidomide 46-58 interleukin 1 alpha Mus musculus 146-154 19733563-10 2009 Qualitative histological analysis showed that lenalidomide treatment modestly reduced the expression of the proinflammatory cytokines Fas Ligand, IL-1beta, TNF-alpha and CD40 ligand. Lenalidomide 46-58 tumor necrosis factor Mus musculus 156-165 19797731-0 2009 Clonal heterogeneity in the 5q- syndrome: p53 expressing progenitors prevail during lenalidomide treatment and expand at disease progression. Lenalidomide 84-96 tumor protein p53 Homo sapiens 42-45 19728370-3 2009 The authors reported the results of a phase 2/3 study comparing the safety and efficacy of 2 doses of lenalidomide in patients with relapsed metastatic melanoma disease refractory to previous treatment with dacarbazine, temozolomide, interleukin-2, or interferon-alpha. Lenalidomide 102-114 interleukin 2 Homo sapiens 234-247 19738071-8 2009 Based on our findings, we propose a model in which pomalidomide and lenalidomide modify the chromatin structure of the p21(WAF-1) promoter through demethylation and acetylation of H3K9. Lenalidomide 68-80 cyclin dependent kinase inhibitor 1A Homo sapiens 119-122 19738071-8 2009 Based on our findings, we propose a model in which pomalidomide and lenalidomide modify the chromatin structure of the p21(WAF-1) promoter through demethylation and acetylation of H3K9. Lenalidomide 68-80 cyclin dependent kinase inhibitor 1A Homo sapiens 123-128 19738071-0 2009 Pomalidomide and lenalidomide induce p21 WAF-1 expression in both lymphoma and multiple myeloma through a LSD1-mediated epigenetic mechanism. Lenalidomide 17-29 cyclin dependent kinase inhibitor 1A Homo sapiens 41-46 19738071-0 2009 Pomalidomide and lenalidomide induce p21 WAF-1 expression in both lymphoma and multiple myeloma through a LSD1-mediated epigenetic mechanism. Lenalidomide 17-29 lysine demethylase 1A Homo sapiens 106-110 19738071-7 2009 Interestingly, lysine-specific demethylase-1 (LSD1) silencing reduced both pomalidomide and lenalidomide up-regulation of p21(WAF-1), suggesting that this histone demethylase is involved in the priming of the p21(WAF-1) promoter. Lenalidomide 92-104 lysine demethylase 1A Homo sapiens 15-44 19738071-7 2009 Interestingly, lysine-specific demethylase-1 (LSD1) silencing reduced both pomalidomide and lenalidomide up-regulation of p21(WAF-1), suggesting that this histone demethylase is involved in the priming of the p21(WAF-1) promoter. Lenalidomide 92-104 lysine demethylase 1A Homo sapiens 46-50 19738071-7 2009 Interestingly, lysine-specific demethylase-1 (LSD1) silencing reduced both pomalidomide and lenalidomide up-regulation of p21(WAF-1), suggesting that this histone demethylase is involved in the priming of the p21(WAF-1) promoter. Lenalidomide 92-104 cyclin dependent kinase inhibitor 1A Homo sapiens 122-125 19738071-7 2009 Interestingly, lysine-specific demethylase-1 (LSD1) silencing reduced both pomalidomide and lenalidomide up-regulation of p21(WAF-1), suggesting that this histone demethylase is involved in the priming of the p21(WAF-1) promoter. Lenalidomide 92-104 cyclin dependent kinase inhibitor 1A Homo sapiens 126-131 19734418-0 2009 Lenalidomide-induced upregulation of CD80 on tumor cells correlates with T-cell activation, the rapid onset of a cytokine release syndrome and leukemic cell clearance in chronic lymphocytic leukemia. Lenalidomide 0-12 CD80 molecule Homo sapiens 37-41 19734418-5 2009 RESULTS: Lenalidomide upregulated the co-stimulatory molecule CD80 on chronic lymphocytic leukemia and mantle cell lymphoma cells but not on normal peripheral blood B cells in vitro. Lenalidomide 9-21 CD80 molecule Homo sapiens 62-66 19734418-7 2009 Strong CD80 upregulation and T-cell activation predicted more severe side effects, manifesting in 83% of patients as a cytokine release syndrome within 8-72 h after the first dose of lenalidomide. Lenalidomide 183-195 CD80 molecule Homo sapiens 7-11 19734418-11 2009 CONCLUSIONS: Upregulation of CD80 on tumor cells and T-cell activation correlate with unique toxicities of lenalidomide in chronic lymphocytic leukemia. Lenalidomide 107-119 CD80 molecule Homo sapiens 29-33 19470455-2 2009 Although haploinsufficiency for the RPS14 gene and others encoded within the common deleted region (CDR) have been implicated in the pathogenesis of the del(5q) phenotype, the molecular basis of the karyotype specificity of lenalidomide remains unexplained. Lenalidomide 224-236 ribosomal protein S14 Homo sapiens 36-41 19470455-4 2009 We show that the dual specificity phosphatases, Cdc25C and PP2Acalpha, which are coregulators of the G(2)-M checkpoint, are inhibited by lenalidomide. Lenalidomide 137-149 cell division cycle 25C Homo sapiens 48-54 19470455-6 2009 Lenalidomide inhibited phosphatase activity either directly (Cdc25C) or indirectly (PP2A) with corresponding retention of inhibitory phospho-tyrosine residues. Lenalidomide 0-12 cell division cycle 25C Homo sapiens 61-67 19470455-6 2009 Lenalidomide inhibited phosphatase activity either directly (Cdc25C) or indirectly (PP2A) with corresponding retention of inhibitory phospho-tyrosine residues. Lenalidomide 0-12 protein phosphatase 2 phosphatase activator Homo sapiens 84-88 19470455-8 2009 Small interfering RNA (shRNA) suppression of Cdc25C and PP2Acalpha gene expression recapitulated del(5q) susceptibility to lenalidomide with induction of G(2) arrest and apoptosis in both U937 and primary nondel(5q) MDS cells. Lenalidomide 123-135 cell division cycle 25C Homo sapiens 45-51 19470455-9 2009 These data establish a role for allelic haplodeficiency of the lenalidomide inhibitable Cdc25C and PP2Acalpha phosphatases in the selective drug sensitivity of del(5q) MDS. Lenalidomide 63-75 cell division cycle 25C Homo sapiens 88-94 19183192-0 2009 The humanized CD40 antibody SGN-40 demonstrates pre-clinical activity that is enhanced by lenalidomide in chronic lymphocytic leukaemia. Lenalidomide 90-102 CD40 molecule Homo sapiens 14-18 19717386-2 2009 We describe a case of an elderly patient who presented with extramedullary, IgA-lambda-secreting multiple myeloma with plasmablastic features who demonstrated impressive clinical response to single-agent lenalidomide. Lenalidomide 204-216 CD79a molecule Homo sapiens 76-79 19525184-3 2009 Here, we describe 6 patients with International Prognostic Scoring System low- or intermediate-1-risk MDS who began lenalidomide therapy between April 2002 and June 2003 as part of the MDS-001 study and who have maintained long-term therapy. Lenalidomide 116-128 BBX high mobility group box domain containing Homo sapiens 185-192 19302559-2 2009 Multivariate analysis showed that fewer prior therapies, along with beta(2)-microglobulin (< or = 2.5 mg/L), predicted a better time to progression (TTP; study end-point) with lenalidomide plus dexamethasone treatment. Lenalidomide 179-191 beta-2-microglobulin Homo sapiens 68-89 19030860-0 2009 Lenalidomide: in vitro evaluation of the metabolism and assessment of cytochrome P450 inhibition and induction. Lenalidomide 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-85 19030860-1 2009 PURPOSE: To assess the potential for drug-drug interactions between lenalidomide and substrates and inhibitors of cytochrome P450 (CYP) isozymes. Lenalidomide 68-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-134 19030860-3 2009 The inhibitory and inductive effects of lenalidomide on the CYP activities were evaluated in human liver microsomes and cultured human hepatocytes, respectively. Lenalidomide 40-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-63 19406725-1 2009 BACKGROUND: Initial analysis of the combination melphalan, prednisone, plus lenalidomide (MPR) showed significant antimyeloma activity in patients with untreated multiple myeloma, with neutropenia and thrombocytopenia as the most frequent side effects. Lenalidomide 76-88 progesterone receptor membrane component 1 Homo sapiens 90-93 19509231-3 2009 A drug screen for anti-MM agents that decrease Hif-1alpha and c-Myc levels identified a variety of compounds, including bortezomib, lenalidomide, enzastaurin, and adaphostin. Lenalidomide 132-144 MYC proto-oncogene, bHLH transcription factor a Danio rerio 62-67 19183192-7 2009 Previous studies by our group and others have demonstrated that lenalidomide upregulates CD40 expression on primary B CLL cells and activates NK-cells. Lenalidomide 64-76 CD40 molecule Homo sapiens 89-93 19778857-2 2009 The MDS-003 multicenter registration trial in deletion of chromosome 5q (del[5q]) showed that lenalidomide suppresses the del(5q) clone in patients who achieve transfusion independence and is a prerequisite for sustained restoration of effective erythropoiesis. Lenalidomide 94-106 proteasome assembly chaperone 2 Homo sapiens 4-11 19181778-0 2009 The response to lenalidomide of myelodysplastic syndrome patients with deletion del(5q) can be sequentially monitored in CD34+ progenitor cells. Lenalidomide 16-28 CD34 molecule Homo sapiens 121-125 18784738-0 2009 Lenalidomide alone or in combination with dexamethasone is highly effective in patients with relapsed multiple myeloma following allogeneic stem cell transplantation and increases the frequency of CD4+Foxp3+ T cells. Lenalidomide 0-12 CD4 molecule Homo sapiens 197-200 18784738-0 2009 Lenalidomide alone or in combination with dexamethasone is highly effective in patients with relapsed multiple myeloma following allogeneic stem cell transplantation and increases the frequency of CD4+Foxp3+ T cells. Lenalidomide 0-12 forkhead box P3 Homo sapiens 201-206 18805433-5 2009 We also found an inhibitory effect of lenalidomide on the associations between cadherin 5, beta-catenin and CD31, adherens junction proteins whose interaction is critical for endothelial cell cord formation. Lenalidomide 38-50 cadherin 5 Homo sapiens 79-89 18805433-5 2009 We also found an inhibitory effect of lenalidomide on the associations between cadherin 5, beta-catenin and CD31, adherens junction proteins whose interaction is critical for endothelial cell cord formation. Lenalidomide 38-50 catenin beta 1 Homo sapiens 91-103 18805433-5 2009 We also found an inhibitory effect of lenalidomide on the associations between cadherin 5, beta-catenin and CD31, adherens junction proteins whose interaction is critical for endothelial cell cord formation. Lenalidomide 38-50 platelet and endothelial cell adhesion molecule 1 Homo sapiens 108-112 18805433-6 2009 Furthermore, lenalidomide inhibited VEGF-induced PI3K-Akt pathway signaling, which is known to regulate adherens junction formation. Lenalidomide 13-25 vascular endothelial growth factor A Homo sapiens 36-40 18805433-7 2009 We also found a strong inhibitory effect of lenalidomide on hypoxia-induced endothelial cell formation of cords and HIF-1 alpha expression, the main mediator of hypoxia-mediated effects and a key driver of angiogenesis and metastasis. Lenalidomide 44-56 hypoxia inducible factor 1 subunit alpha Homo sapiens 116-127 19707399-3 2009 In the near future melphalan-prednisone-lenalidomide (Revlimid((R)), MPR) will also provide a third therapeutic option (MPT, MPV, and MPR), in elderly multiple myeloma, eventually. Lenalidomide 40-52 progesterone receptor membrane component 1 Homo sapiens 134-137 19707399-3 2009 In the near future melphalan-prednisone-lenalidomide (Revlimid((R)), MPR) will also provide a third therapeutic option (MPT, MPV, and MPR), in elderly multiple myeloma, eventually. Lenalidomide 54-62 progesterone receptor membrane component 1 Homo sapiens 134-137 18950461-3 2008 In a study of the immunomodulatory combination BiRD, (lenalidomide and dexamethasone with clarithromycin), we frequently detected the emergence of mono- and oligo-clonal immunoglobulins unrelated to the baseline diagnostic M-protein. Lenalidomide 54-66 myomesin 2 Homo sapiens 223-232 19067746-3 2009 In this study, the effect of different treatment regimens for MM on serum DKK-1 was evaluated and correlated with the response to treatment in 101 myeloma patients receiving bortezomib, thalidomide, lenalidomide, adriamycin and dexamethasone (AD) or high-dose chemotherapy (HDCT) followed by autologous stem cell transplantation (ASCT). Lenalidomide 199-211 dickkopf WNT signaling pathway inhibitor 1 Homo sapiens 74-79 19067746-6 2009 A significant decrease of DKK-1 after therapy was seen in the following groups: Bortezomib (4059 pg/mL vs. 1862 pg/mL, P = 0.016), lenalidomide (11837 pg/mL vs. 4374 pg/mL, P = 0.039), AD (1668 pg/mL vs. 1241 pg/mL, P = 0.016), and AD + HDCT + ASCT (2446 pg/mL vs. 1082 pg/mL, P = 0.001). Lenalidomide 131-143 dickkopf WNT signaling pathway inhibitor 1 Homo sapiens 26-31 18772452-0 2008 Lenalidomide down-regulates the CD20 antigen and antagonizes direct and antibody-dependent cellular cytotoxicity of rituximab on primary chronic lymphocytic leukemia cells. Lenalidomide 0-12 keratin 20 Homo sapiens 32-36 18772452-2 2008 The anti-CD20 antibody rituximab is active in CLL and represents a rational agent to combine with lenalidomide. Lenalidomide 98-110 keratin 20 Homo sapiens 9-13 18772452-4 2008 In contrast to previous reports using CD20-positive lymphoma cell lines, lenalidomide down-regulated CD20 surface antigen expression in CLL patient cells via enhanced internalization, without influencing transcription. Lenalidomide 73-85 keratin 20 Homo sapiens 101-105 18772452-6 2008 In addition, CD20 surface antigen down-modulation by lenalidomide in CLL was accompanied by diminished rituximab-mediated apoptosis and ADCC. Lenalidomide 53-65 keratin 20 Homo sapiens 13-17 18772452-8 2008 In addition, they suggest that lenalidomide therapy might be useful to enhance targeted delivery of RNAi-based therapies using CD20 immunoliposomes in B-cell malignancies. Lenalidomide 31-43 keratin 20 Homo sapiens 127-131 18799726-4 2008 Lenalidomide + dexamethasone led to higher overall response rate (ORR), longer time to progression (TTP), and progression-free survival (PFS) versus placebo + dexamethasone despite prior thalidomide exposure. Lenalidomide 0-12 ZFP36 ring finger protein Homo sapiens 100-103 18799726-5 2008 Among lenalidomide + dexamethasone-treated patients, ORR was higher in thalidomide-naive versus thalidomide-exposed patients (P = .04), with longer median TTP (P = .04) and PFS (P = .02). Lenalidomide 6-18 ZFP36 ring finger protein Homo sapiens 155-158 18980990-9 2008 Importantly, lenalidomide further augmented the Th1 polarization by iNKT cell lines via increased Th1 cytokine production and reduced Th2 cytokine production. Lenalidomide 13-25 negative elongation factor complex member C/D Homo sapiens 48-51 18980990-9 2008 Importantly, lenalidomide further augmented the Th1 polarization by iNKT cell lines via increased Th1 cytokine production and reduced Th2 cytokine production. Lenalidomide 13-25 negative elongation factor complex member C/D Homo sapiens 98-101 18427150-10 2008 In vitro studies demonstrated lenalidomide-induced B-cell activation (upregulation of CD40 and CD86) corresponding to degree of tumor flare, possibly explaining the tumor flare observation. Lenalidomide 30-42 CD40 molecule Homo sapiens 86-90 18713253-2 2008 TE risk may be elevated with combination regimens, notably thalidomide/lenalidomide plus high-dose dexamethasone; concomitant erythropoietin appears to further increase the risk with lenalidomide-dexamethasone. Lenalidomide 183-195 erythropoietin Homo sapiens 126-140 18628480-0 2008 lenalidomide enhances natural killer cell and monocyte-mediated antibody-dependent cellular cytotoxicity of rituximab-treated CD20+ tumor cells. Lenalidomide 0-12 keratin 20 Homo sapiens 126-130 18628480-5 2008 RESULTS: Lenalidomide directly enhanced IFN-gamma production via Fc-gamma receptor-mediated signaling in response to IgG. Lenalidomide 9-21 interferon gamma Homo sapiens 40-49 18628480-5 2008 RESULTS: Lenalidomide directly enhanced IFN-gamma production via Fc-gamma receptor-mediated signaling in response to IgG. Lenalidomide 9-21 Fc gamma receptor Ia Homo sapiens 65-82 18536579-5 2008 Moreover, several drugs that are effective against multiple myeloma, including bortezomib, thalidomide, lenalidomide and arsenic trioxide, have been found to block activation of NF-kappaB. Lenalidomide 104-116 nuclear factor kappa B subunit 1 Homo sapiens 178-187 18596740-6 2008 We identified PU.1 and pERK as major targets of lenalidomide, and nuclear factor of activated T cells of bortezomib, resulting in inhibition of osteoclastogenesis. Lenalidomide 48-60 eukaryotic translation initiation factor 2 alpha kinase 3 Homo sapiens 23-27 18596740-9 2008 Importantly, in serum from MM patients treated with lenalidomide, the essential bone-remodeling factor RANKL, as well as the RANKL/OPG ratio, were significantly reduced, whereas osteoprotegerin (OPG) was increased. Lenalidomide 52-64 TNF superfamily member 11 Homo sapiens 103-108 18596740-9 2008 Importantly, in serum from MM patients treated with lenalidomide, the essential bone-remodeling factor RANKL, as well as the RANKL/OPG ratio, were significantly reduced, whereas osteoprotegerin (OPG) was increased. Lenalidomide 52-64 TNF superfamily member 11 Homo sapiens 125-130 18596740-9 2008 Importantly, in serum from MM patients treated with lenalidomide, the essential bone-remodeling factor RANKL, as well as the RANKL/OPG ratio, were significantly reduced, whereas osteoprotegerin (OPG) was increased. Lenalidomide 52-64 TNF receptor superfamily member 11b Homo sapiens 131-134 18596740-9 2008 Importantly, in serum from MM patients treated with lenalidomide, the essential bone-remodeling factor RANKL, as well as the RANKL/OPG ratio, were significantly reduced, whereas osteoprotegerin (OPG) was increased. Lenalidomide 52-64 TNF receptor superfamily member 11b Homo sapiens 178-193 18596740-9 2008 Importantly, in serum from MM patients treated with lenalidomide, the essential bone-remodeling factor RANKL, as well as the RANKL/OPG ratio, were significantly reduced, whereas osteoprotegerin (OPG) was increased. Lenalidomide 52-64 TNF receptor superfamily member 11b Homo sapiens 195-198 18427150-10 2008 In vitro studies demonstrated lenalidomide-induced B-cell activation (upregulation of CD40 and CD86) corresponding to degree of tumor flare, possibly explaining the tumor flare observation. Lenalidomide 30-42 CD86 molecule Homo sapiens 95-99 17906467-5 2007 Results from the MDS-003 multicenter deletion 5q registration study show that lenalidomide suppresses the deletion 5q clone, and in higher risk myelodysplastic syndrome patients, may also alter the natural history of disease. Lenalidomide 78-90 proteasome assembly chaperone 2 Homo sapiens 17-24 18067473-0 2008 Should prophylactic granulocyte-colony stimulating factor be used in multiple myeloma patients developing neutropenia under lenalidomide-based therapy? Lenalidomide 124-136 colony stimulating factor 3 Homo sapiens 20-57 18064299-0 2008 Pomalidomide and lenalidomide regulate erythropoiesis and fetal hemoglobin production in human CD34+ cells. Lenalidomide 17-29 CD34 molecule Homo sapiens 95-99 17576924-0 2007 Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients. Lenalidomide 0-12 secreted protein acidic and cysteine rich Homo sapiens 63-68 17724360-2 2007 SUMMARY: Lenalidomide is an analogue of thalidomide and has been shown to be more potent than thalidomide in the stimulation of T-cell, interleukin-2, and interferon-gamma production. Lenalidomide 9-21 interferon gamma Homo sapiens 155-171 17686058-5 2007 Caution should be exercised when lenalidomide therapy is commenced and CrCl should be incorporated as a determinant of the initial dosing of lenalidomide in MM patients. Lenalidomide 141-153 CRCL Homo sapiens 71-75 17581613-4 2007 Among those mobilized with granulocyte-colony stimulating factor (G-CSF) alone, there was a significant decrease in total CD34(+) cells collected (P<0.001), average daily collection (P<0.001), day 1 collection (P<0.001) and increased number of aphereses (P=0.004) in patients treated with lenalidomide compared to those receiving dexamethasone, thalidomide-dexamethasone or VAD. Lenalidomide 298-310 colony stimulating factor 3 Homo sapiens 27-64 17581613-4 2007 Among those mobilized with granulocyte-colony stimulating factor (G-CSF) alone, there was a significant decrease in total CD34(+) cells collected (P<0.001), average daily collection (P<0.001), day 1 collection (P<0.001) and increased number of aphereses (P=0.004) in patients treated with lenalidomide compared to those receiving dexamethasone, thalidomide-dexamethasone or VAD. Lenalidomide 298-310 colony stimulating factor 3 Homo sapiens 66-71 17724360-2 2007 SUMMARY: Lenalidomide is an analogue of thalidomide and has been shown to be more potent than thalidomide in the stimulation of T-cell, interleukin-2, and interferon-gamma production. Lenalidomide 9-21 interleukin 2 Homo sapiens 136-149 17576924-6 2007 Moreover, lenalidomide significantly influenced the pattern of gene expression in del(5q) intermediate erythroblasts, with the VSIG4, PPIC, TPBG, activin A, and SPARC genes up-regulated by >2-fold in all samples and many genes involved in erythropoiesis, including HBA2, GYPA, and KLF1, down-regulated in most samples. Lenalidomide 10-22 V-set and immunoglobulin domain containing 4 Homo sapiens 127-132 17576924-6 2007 Moreover, lenalidomide significantly influenced the pattern of gene expression in del(5q) intermediate erythroblasts, with the VSIG4, PPIC, TPBG, activin A, and SPARC genes up-regulated by >2-fold in all samples and many genes involved in erythropoiesis, including HBA2, GYPA, and KLF1, down-regulated in most samples. Lenalidomide 10-22 peptidylprolyl isomerase C Homo sapiens 134-138 17576924-6 2007 Moreover, lenalidomide significantly influenced the pattern of gene expression in del(5q) intermediate erythroblasts, with the VSIG4, PPIC, TPBG, activin A, and SPARC genes up-regulated by >2-fold in all samples and many genes involved in erythropoiesis, including HBA2, GYPA, and KLF1, down-regulated in most samples. Lenalidomide 10-22 trophoblast glycoprotein Homo sapiens 140-144 17576924-6 2007 Moreover, lenalidomide significantly influenced the pattern of gene expression in del(5q) intermediate erythroblasts, with the VSIG4, PPIC, TPBG, activin A, and SPARC genes up-regulated by >2-fold in all samples and many genes involved in erythropoiesis, including HBA2, GYPA, and KLF1, down-regulated in most samples. Lenalidomide 10-22 secreted protein acidic and cysteine rich Homo sapiens 161-166 17576924-6 2007 Moreover, lenalidomide significantly influenced the pattern of gene expression in del(5q) intermediate erythroblasts, with the VSIG4, PPIC, TPBG, activin A, and SPARC genes up-regulated by >2-fold in all samples and many genes involved in erythropoiesis, including HBA2, GYPA, and KLF1, down-regulated in most samples. Lenalidomide 10-22 hemoglobin subunit alpha 2 Homo sapiens 268-272 17576924-6 2007 Moreover, lenalidomide significantly influenced the pattern of gene expression in del(5q) intermediate erythroblasts, with the VSIG4, PPIC, TPBG, activin A, and SPARC genes up-regulated by >2-fold in all samples and many genes involved in erythropoiesis, including HBA2, GYPA, and KLF1, down-regulated in most samples. Lenalidomide 10-22 glycophorin A (MNS blood group) Homo sapiens 274-278 17576924-6 2007 Moreover, lenalidomide significantly influenced the pattern of gene expression in del(5q) intermediate erythroblasts, with the VSIG4, PPIC, TPBG, activin A, and SPARC genes up-regulated by >2-fold in all samples and many genes involved in erythropoiesis, including HBA2, GYPA, and KLF1, down-regulated in most samples. Lenalidomide 10-22 Kruppel like factor 1 Homo sapiens 284-288 17576924-9 2007 We conclude that lenalidomide inhibits growth of del(5q) erythroid progenitors and that the up-regulation of SPARC and activin A may underlie the potent effects of lenalidomide in MDS with del(5)(q31). Lenalidomide 164-176 secreted protein acidic and cysteine rich Homo sapiens 109-114 17255354-5 2007 Despite its role as a tumor suppressor and mediator of apoptosis in other cell types including osteoblasts, our data suggest that DKK1, a stress-responsive gene in MM, does not mediate apoptotic signaling, is not activated by TP53, and its forced overexpression could not inhibit cell growth or sensitize MM cells to apoptosis following treatment with thalidomide or lenalidomide. Lenalidomide 367-379 dickkopf WNT signaling pathway inhibitor 1 Homo sapiens 130-134 17255789-5 2007 Furthermore, lenalidomide induces a direct cytotoxic effect against 5q- clones in leukemia cell lines and enhances ligand-induced erythropoietin receptor signaling in erythroid progenitors. Lenalidomide 13-25 erythropoietin Homo sapiens 130-144 17234786-0 2007 Lenalidomide and CC-4047 inhibit the proliferation of malignant B cells while expanding normal CD34+ progenitor cells. Lenalidomide 0-12 CD34 molecule Homo sapiens 95-99 17234786-6 2007 Finally, CC-4047 and lenalidomide had synergistic effects with valproic acid [a histone deacetylase (HDAC) inhibitor] by increasing the apoptosis of Namalwa cells and enhancing CD34+ cell expansion. Lenalidomide 21-33 CD34 molecule Homo sapiens 177-181 17369076-5 2007 The thalidomide analogs, lenalidomide (CC-5013; Revlimid) and CC-4047 (Actimid), have enhanced potency as inhibitors of TNF-alpha and other inflammatory cytokines, as well as greater capacity to promote T-cell activation and suppress angiogenesis. Lenalidomide 25-37 tumor necrosis factor Homo sapiens 120-129 17168657-4 2006 It seems that lenalidomide and CC-4047 are more powerful in inhibiting TNFalpha and have, except for myelosuppression, less side effects than Thal. Lenalidomide 14-26 tumor necrosis factor Homo sapiens 71-79 17242661-6 2006 These studies identified lenalidomide as a highly active erythropoietic- and cytogenetic-remitting agent in lower-risk MDS patients who otherwise would not be expected to benefit from recombinant erythropoietin therapy. Lenalidomide 25-37 erythropoietin Homo sapiens 196-210 17242663-7 2006 The costs of lenalidomide therapy were substantially offset by reduced blood transfusion and EPO costs. Lenalidomide 13-25 erythropoietin Homo sapiens 93-96 16494942-0 2006 Lenalidomide inhibits proliferation of Namalwa CSN.70 cells and interferes with Gab1 phosphorylation and adaptor protein complex assembly. Lenalidomide 0-12 GRB2 associated binding protein 1 Homo sapiens 80-84 16990762-0 2006 Hematologic and cytogenetic response to lenalidomide monotherapy in acute myeloid leukemia arising from JAK2(V617F) positive, del(5)(q13q33) myelodysplastic syndrome. Lenalidomide 40-52 Janus kinase 2 Homo sapiens 104-114 17076650-3 2006 Lenalidomide (CC-5013; Revlimid), a second generation IMiD, has shown significant erythropoietic activity in patients with lower risk MDS that have failed or are not candidates for recombinant erythropoietin treatment. Lenalidomide 0-12 erythropoietin Homo sapiens 193-207 17076650-3 2006 Lenalidomide (CC-5013; Revlimid), a second generation IMiD, has shown significant erythropoietic activity in patients with lower risk MDS that have failed or are not candidates for recombinant erythropoietin treatment. Lenalidomide 14-21 erythropoietin Homo sapiens 193-207 17076650-3 2006 Lenalidomide (CC-5013; Revlimid), a second generation IMiD, has shown significant erythropoietic activity in patients with lower risk MDS that have failed or are not candidates for recombinant erythropoietin treatment. Lenalidomide 23-31 erythropoietin Homo sapiens 193-207 17076650-4 2006 Unlike cytokine therapy, lenalidomide suppresses select MDS clones and enhances erythropoietin receptor signaling to restore erythropoiesis. Lenalidomide 25-37 erythropoietin Homo sapiens 80-94 17076650-10 2006 A phase III Intergroup trial (ECOG 2905) will test the capacity to potentiate erythropoietin response by comparing response to lenalidomide monotherapy to the combination of darbepoetin and lenalidomide in non-deletion 5q MDS patients. Lenalidomide 127-139 erythropoietin Homo sapiens 78-92 17076650-10 2006 A phase III Intergroup trial (ECOG 2905) will test the capacity to potentiate erythropoietin response by comparing response to lenalidomide monotherapy to the combination of darbepoetin and lenalidomide in non-deletion 5q MDS patients. Lenalidomide 190-202 erythropoietin Homo sapiens 78-92 16569772-7 2006 Together these data demonstrate that LEN and its analogues enhance CD1d-mediated presentation of glycolipid antigens and support combining these agents with NKT targeted approaches for protection against tumors. Lenalidomide 37-40 CD1d molecule Homo sapiens 67-71 16494942-5 2006 In the most lenalidomide-sensitive chromosome 5 deleted cell line, Namalwa CSN.70, the compound induced G0/G1 cell cycle arrest, inhibited Akt and Gab1 phosphorylation, and inhibited the ability of Gab1 to associate with a receptor tyrosine kinase. Lenalidomide 12-24 AKT serine/threonine kinase 1 Homo sapiens 139-142 16494942-5 2006 In the most lenalidomide-sensitive chromosome 5 deleted cell line, Namalwa CSN.70, the compound induced G0/G1 cell cycle arrest, inhibited Akt and Gab1 phosphorylation, and inhibited the ability of Gab1 to associate with a receptor tyrosine kinase. Lenalidomide 12-24 GRB2 associated binding protein 1 Homo sapiens 147-151 16494942-5 2006 In the most lenalidomide-sensitive chromosome 5 deleted cell line, Namalwa CSN.70, the compound induced G0/G1 cell cycle arrest, inhibited Akt and Gab1 phosphorylation, and inhibited the ability of Gab1 to associate with a receptor tyrosine kinase. Lenalidomide 12-24 GRB2 associated binding protein 1 Homo sapiens 198-202 16357183-0 2005 Immunomodulatory drug lenalidomide (CC-5013, IMiD3) augments anti-CD40 SGN-40-induced cytotoxicity in human multiple myeloma: clinical implications. Lenalidomide 22-34 CD40 molecule Homo sapiens 66-70 16735711-4 2006 Lenalidomide is particularly active in treating the anemia of del(5q) MDS, which is especially relevant given the low response rate to erythropoietin in this group of patients. Lenalidomide 0-12 erythropoietin Homo sapiens 135-149 16510725-5 2006 We tested the neuroprotective effect of thalidomide and its analog lenalidomide, pharmacological agents that inhibit the expression of TNF-alpha and other cytokines by destabilizing their mRNA. Lenalidomide 67-79 tumor necrosis factor Mus musculus 135-144 16357183-7 2005 Lenalidomide also up-regulated CD40L on CD56(+)CD3(-) NK cells, facilitating IL-2-mediated activation of NK cells. Lenalidomide 0-12 CD40 ligand Homo sapiens 31-36 16357183-7 2005 Lenalidomide also up-regulated CD40L on CD56(+)CD3(-) NK cells, facilitating IL-2-mediated activation of NK cells. Lenalidomide 0-12 neural cell adhesion molecule 1 Homo sapiens 40-44 16357183-7 2005 Lenalidomide also up-regulated CD40L on CD56(+)CD3(-) NK cells, facilitating IL-2-mediated activation of NK cells. Lenalidomide 0-12 interleukin 2 Homo sapiens 77-81 16357183-8 2005 In addition, lenalidomide induced the CD56(dim) NK subset, which are more potent mediators of ADCC against target MM cells than the CD56(bright) NK subset. Lenalidomide 13-25 neural cell adhesion molecule 1 Homo sapiens 38-42 16357183-9 2005 Finally, pretreatment of both effector and target MM cells with lenalidomide markedly enhanced SGN-40-mediated ADCC against CD40-expressing MM cells. Lenalidomide 64-76 CD40 molecule Homo sapiens 124-128 17124060-5 2006 Lenalidomide can lead to RBC transfusion independence in at least two thirds of cases of the 5q- syndrome, two thirds of those responses persisting after 2 years of treatment. Lenalidomide 0-12 RNA, 7SL, cytoplasmic 263, pseudogene Homo sapiens 25-28 16357183-2 2005 Here, we studied the clinical significance of an immunomodulatory drug lenalidomide on SGN-40-induced cytotoxicity against CD138(+)CD40(+) MM lines and patient MM cells. Lenalidomide 71-83 CD40 molecule Homo sapiens 131-135 16357183-4 2005 Combined lenalidomide and SGN-40 significantly induced MM apoptosis, evidenced by enhanced cleavage of caspase-3/8/poly(ADP-ribose)polymerase and increased sub-G(0) cells, compared with either single agent at the same doses. Lenalidomide 9-21 poly(ADP-ribose) polymerase 1 Homo sapiens 103-141 16357183-5 2005 Pretreatment of effector cells with lenalidomide augmented SGN-40-induced MM cell lysis, associated with an increased number of CD56(+)CD3(-) natural killer (NK) cells expressing CD16 and LFA-1. Lenalidomide 36-48 neural cell adhesion molecule 1 Homo sapiens 128-132 16357183-5 2005 Pretreatment of effector cells with lenalidomide augmented SGN-40-induced MM cell lysis, associated with an increased number of CD56(+)CD3(-) natural killer (NK) cells expressing CD16 and LFA-1. Lenalidomide 36-48 Fc gamma receptor IIIa Homo sapiens 179-183 16357183-5 2005 Pretreatment of effector cells with lenalidomide augmented SGN-40-induced MM cell lysis, associated with an increased number of CD56(+)CD3(-) natural killer (NK) cells expressing CD16 and LFA-1. Lenalidomide 36-48 integrin subunit alpha L Homo sapiens 188-193 15703420-13 2005 CONCLUSIONS: Lenalidomide has hematologic activity in patients with low-risk myelodysplastic syndromes who have no response to erythropoietin or who are unlikely to benefit from conventional therapy. Lenalidomide 13-25 erythropoietin Homo sapiens 127-141 16085015-8 2005 In nonclinical studies, the effects of lenalidomide include potentiation of clonogenic response to erythropoietin, activation of integrin-mediated adhesion, cell cycle arrest, sensitization to apoptotic signals, and abrogation of cellular response to receptor-initiated trophic signals. Lenalidomide 39-51 erythropoietin Homo sapiens 99-113 15797261-0 2005 Orally administered lenalidomide (CC-5013) is anti-angiogenic in vivo and inhibits endothelial cell migration and Akt phosphorylation in vitro. Lenalidomide 20-32 AKT serine/threonine kinase 1 Rattus norvegicus 114-117 15797261-0 2005 Orally administered lenalidomide (CC-5013) is anti-angiogenic in vivo and inhibits endothelial cell migration and Akt phosphorylation in vitro. Lenalidomide 34-41 AKT serine/threonine kinase 1 Rattus norvegicus 114-117 15797261-4 2005 We now show that oral administration of lenalidomide attenuates growth factor-induced angiogenesis in vivo; the rat mesenteric window assay was utilized to show that lenalidomide significantly inhibits vascularization in a dose-dependent manner. Lenalidomide 40-52 myotrophin Rattus norvegicus 64-77 15797261-4 2005 We now show that oral administration of lenalidomide attenuates growth factor-induced angiogenesis in vivo; the rat mesenteric window assay was utilized to show that lenalidomide significantly inhibits vascularization in a dose-dependent manner. Lenalidomide 166-178 myotrophin Rattus norvegicus 64-77 15797261-5 2005 We also found that lenalidomide significantly inhibits growth factor-induced endothelial cell migration. Lenalidomide 19-31 myotrophin Rattus norvegicus 55-68 15797261-6 2005 This correlates with the inhibitory effect of lenalidomide on growth factor-induced Akt phosphorylation, thereby providing a potential mechanism for its anti-migratory and subsequent anti-angiogenic effects. Lenalidomide 46-58 myotrophin Rattus norvegicus 62-75 15797261-6 2005 This correlates with the inhibitory effect of lenalidomide on growth factor-induced Akt phosphorylation, thereby providing a potential mechanism for its anti-migratory and subsequent anti-angiogenic effects. Lenalidomide 46-58 AKT serine/threonine kinase 1 Rattus norvegicus 84-87 34952465-0 2022 Lenalidomide attenuates post-inflammation pulmonary fibrosis through blocking NF-kappaB signaling pathway. Lenalidomide 0-12 nuclear factor kappa B subunit 1 Homo sapiens 78-87 33770460-7 2021 Combination of lenalidomide and rituximab has been a safe and effective immune modality for patients with refractory indolent lymphomas; it is currently being used as a backbone to bring other targeted agents such as tazemetostat (EZH2 inhibitor) into earlier lines of treatment. Lenalidomide 15-27 enhancer of zeste 2 polycomb repressive complex 2 subunit Homo sapiens 231-235 32859220-5 2020 ATLL cell lines were also sensitive to lenalidomide, which repressed IRF4 expression. Lenalidomide 39-51 interferon regulatory factor 4 Homo sapiens 69-73 32859220-8 2020 CONCLUSIONS: These data support the pursuit of IRF4 as a therapeutic target in ATLL with the use of either ASOs or lenalidomide. Lenalidomide 115-127 interferon regulatory factor 4 Homo sapiens 47-51 34952465-8 2022 In vitro study revealed that lenalidomide inhibited NF-kappaB signaling in LPS-induced macrophage, and further attenuated macrophage-induced myofibroblast activation. Lenalidomide 29-41 nuclear factor kappa B subunit 1 Homo sapiens 52-61 34952465-9 2022 Meanwhile, lenalidomide could inhibit TGF-beta1-induced myofibroblast activation through suppressing TGF-beta1 downstream MAPK signaling. Lenalidomide 11-23 transforming growth factor beta 1 Homo sapiens 38-47 34952465-9 2022 Meanwhile, lenalidomide could inhibit TGF-beta1-induced myofibroblast activation through suppressing TGF-beta1 downstream MAPK signaling. Lenalidomide 11-23 transforming growth factor beta 1 Homo sapiens 101-110 34952465-10 2022 In vivo study showed that lenalidomide inhibited pro-inflammatory cytokines TNF-alpha and IL-6 while enhanced anti-fibrotic cytokines IFN-gamma and IL-10 in bleomycin-induced inflammation model, and attenuated pulmonary fibrosis and collagen deposition in the following fibrosis stage. Lenalidomide 26-38 tumor necrosis factor Homo sapiens 76-85 34952465-10 2022 In vivo study showed that lenalidomide inhibited pro-inflammatory cytokines TNF-alpha and IL-6 while enhanced anti-fibrotic cytokines IFN-gamma and IL-10 in bleomycin-induced inflammation model, and attenuated pulmonary fibrosis and collagen deposition in the following fibrosis stage. Lenalidomide 26-38 interleukin 6 Homo sapiens 90-94 34952465-10 2022 In vivo study showed that lenalidomide inhibited pro-inflammatory cytokines TNF-alpha and IL-6 while enhanced anti-fibrotic cytokines IFN-gamma and IL-10 in bleomycin-induced inflammation model, and attenuated pulmonary fibrosis and collagen deposition in the following fibrosis stage. Lenalidomide 26-38 interferon gamma Homo sapiens 134-143 34952465-10 2022 In vivo study showed that lenalidomide inhibited pro-inflammatory cytokines TNF-alpha and IL-6 while enhanced anti-fibrotic cytokines IFN-gamma and IL-10 in bleomycin-induced inflammation model, and attenuated pulmonary fibrosis and collagen deposition in the following fibrosis stage. Lenalidomide 26-38 interleukin 10 Homo sapiens 148-153 34952465-11 2022 In conclusion, our results demonstrate that lenalidomide possesses potential anti-fibrotic effects through suppressing NF-kappaB signaling. Lenalidomide 44-56 nuclear factor kappa B subunit 1 Homo sapiens 119-128 34936696-1 2022 Immunomodulatory (IMiD) agents like lenalidomide and pomalidomide induce the recruitment of IKZF1 and other targets to the CRL4CRBN E3 ubiquitin ligase, resulting in their ubiquitination and degradation. Lenalidomide 36-48 IKAROS family zinc finger 1 Homo sapiens 92-97 34936696-9 2022 Moreover, lenalidomide-sensitive TCLs can acquire stable resistance via ZFP91 rewiring, which involves casein kinase 2 (CK2) mediated c-Jun inactivation. Lenalidomide 10-22 ZFP91 zinc finger protein, atypical E3 ubiquitin ligase Homo sapiens 72-77 34936696-9 2022 Moreover, lenalidomide-sensitive TCLs can acquire stable resistance via ZFP91 rewiring, which involves casein kinase 2 (CK2) mediated c-Jun inactivation. Lenalidomide 10-22 Jun proto-oncogene, AP-1 transcription factor subunit Homo sapiens 134-139 34588172-0 2021 Caspase-8 regulates the anti-myeloma activity of bortezomib and lenalidomide. Lenalidomide 64-76 caspase 8 Homo sapiens 0-9 34667029-0 2021 An allogeneic multiple myeloma GM-CSF-secreting vaccine with lenalidomide induces long-term immunity and durable clinical responses in patients in near complete remission. Lenalidomide 61-73 colony stimulating factor 2 Homo sapiens 31-37 34674611-0 2021 Lenalidomide improves the antitumor activity of CAR-T cells directed toward the intracellular Wilms Tumor 1 antigen. Lenalidomide 0-12 nuclear receptor subfamily 1, group I, member 3 Mus musculus 48-51 34674611-9 2021 Our data demonstrated that LEN improved the anti-tumor activity of CAR-T cells in vivo by increasing the infiltration of tumors with CD3+ and CD8+ T cells. Lenalidomide 27-30 nuclear receptor subfamily 1, group I, member 3 Mus musculus 67-70 34674611-11 2021 CONCLUSION: These results demonstrate that lenalidomide potentiates WT1 CAR-T activity and paves the way to evaluate the combination of LEN with CAR-T for a planned clinical trial. Lenalidomide 43-55 nuclear receptor subfamily 1, group I, member 3 Mus musculus 72-75 34588172-5 2021 Either inhibition or genetic depletion of CASP-8 decreased the CRBN cleavage upon Btz treatment, which could potentiate the anti-myeloma activity of IMiD lenalidomide (Len). Lenalidomide 168-171 caspase 8 Homo sapiens 42-48 34588172-5 2021 Either inhibition or genetic depletion of CASP-8 decreased the CRBN cleavage upon Btz treatment, which could potentiate the anti-myeloma activity of IMiD lenalidomide (Len). Lenalidomide 168-171 cereblon Homo sapiens 63-67 34588172-6 2021 This work suggests that administration of CASP-8 inhibitors might enhance the overall effectiveness of Btz/Len-based therapeutic treatment for myeloma patients. Lenalidomide 107-110 caspase 8 Homo sapiens 42-48 34588172-7 2021 Significance Statement Caspase-8 activation upon bortezomib treatment results in the cleavage of cereblon, a substrate receptor of the cullin 4-RING E3 ligase, which is responsible for the degradation of two transcription factors IKZF1 and IKZF3 in the presence of immunomodulatory drugs including lenalidomide. Lenalidomide 298-310 caspase 8 Homo sapiens 23-32 34588172-7 2021 Significance Statement Caspase-8 activation upon bortezomib treatment results in the cleavage of cereblon, a substrate receptor of the cullin 4-RING E3 ligase, which is responsible for the degradation of two transcription factors IKZF1 and IKZF3 in the presence of immunomodulatory drugs including lenalidomide. Lenalidomide 298-310 cereblon Homo sapiens 97-105 34588172-7 2021 Significance Statement Caspase-8 activation upon bortezomib treatment results in the cleavage of cereblon, a substrate receptor of the cullin 4-RING E3 ligase, which is responsible for the degradation of two transcription factors IKZF1 and IKZF3 in the presence of immunomodulatory drugs including lenalidomide. Lenalidomide 298-310 IKAROS family zinc finger 1 Homo sapiens 230-235 34588172-7 2021 Significance Statement Caspase-8 activation upon bortezomib treatment results in the cleavage of cereblon, a substrate receptor of the cullin 4-RING E3 ligase, which is responsible for the degradation of two transcription factors IKZF1 and IKZF3 in the presence of immunomodulatory drugs including lenalidomide. Lenalidomide 298-310 IKAROS family zinc finger 3 Homo sapiens 240-245 34588172-8 2021 The administration of caspase-8 inhibitor may enhance the anti-myeloma activity of the combination therapy with bortezomib and lenalidomide to multiple myeloma. Lenalidomide 127-139 caspase 8 Homo sapiens 22-31 34151705-0 2021 Efficacy of lenalidomide in a patient with systemic mastocytosis associated with SF3B1-mutant myelodysplastic syndrome. Lenalidomide 12-24 splicing factor 3b subunit 1 Homo sapiens 81-86 34885822-3 2021 In this study, a series of novel CRBN-recruiting HDAC PROTACs were designed and synthesized by linking hydroxamic acid and benzamide with lenalidomide, pomalidomide, and CC-220 through linkers of different lengths and types. Lenalidomide 138-150 cereblon Homo sapiens 33-37 34885822-3 2021 In this study, a series of novel CRBN-recruiting HDAC PROTACs were designed and synthesized by linking hydroxamic acid and benzamide with lenalidomide, pomalidomide, and CC-220 through linkers of different lengths and types. Lenalidomide 138-150 histone deacetylase 9 Homo sapiens 49-53 34704527-9 2022 EXPERT OPINION: The chemical space of CRBN ligands which is related to the classical IMiDs (thalidomide/lenalidomide/pomalidomide) is comprehensively covered by the current patent literature. Lenalidomide 104-116 cereblon Homo sapiens 38-42 34780623-2 2022 The CD30-targeted antibody-drug conjugate brentuximab vedotin (BV) and the immunomodulator lenalidomide (Len) have demonstrated promising activity as single agents in this population. Lenalidomide 91-103 TNF receptor superfamily member 8 Homo sapiens 4-8 34780623-2 2022 The CD30-targeted antibody-drug conjugate brentuximab vedotin (BV) and the immunomodulator lenalidomide (Len) have demonstrated promising activity as single agents in this population. Lenalidomide 105-108 TNF receptor superfamily member 8 Homo sapiens 4-8 34392531-6 2021 Moreover, CRBN depletion significantly impairs the ability of NK cells to migrate and reduces the enhancing effect of Lenalidomide on NK cell migration. Lenalidomide 118-130 cereblon Homo sapiens 10-14 34392531-9 2021 Altogether our data identify a critical role for CRBN in regulating NK cell functions and suggest that this protein may mediate the stimulatory effect of Lenalidomide on NK cells. Lenalidomide 154-166 cereblon Homo sapiens 49-53 34503111-8 2021 Notwithstanding, the mechanisms by which PIM kinases modulate the immune microenvironment and synergize with the immunomodulatory agents such as lenalidomide have not been deliberately depicted. Lenalidomide 145-157 Pim-1 proto-oncogene, serine/threonine kinase Homo sapiens 41-44 34341502-8 2021 Furthermore, treating the EOL-1 human eosinophilic cell line with lenalidomide yielded a dose-dependent decrease in Aiolos. Lenalidomide 66-78 IKAROS family zinc finger 3 Homo sapiens 116-122 34455369-0 2021 Involvement of EZH2 inhibition in lenalidomide and pomalidomide-mediated growth suppression in HTLV-1-infected cells. Lenalidomide 34-46 enhancer of zeste 2 polycomb repressive complex 2 subunit Homo sapiens 15-19 34455369-4 2021 In three of four tested HTLV-1-infected cell lines, the cells treated with lenalidomide or pomalidomide exhibited mild growth suppression without apoptosis, which was associated with decreased IRF4, c-Myc, and phosphorylated STAT3 levels as well as enhanced SOCS3 expression. Lenalidomide 75-87 interferon regulatory factor 4 Homo sapiens 193-197 34455369-4 2021 In three of four tested HTLV-1-infected cell lines, the cells treated with lenalidomide or pomalidomide exhibited mild growth suppression without apoptosis, which was associated with decreased IRF4, c-Myc, and phosphorylated STAT3 levels as well as enhanced SOCS3 expression. Lenalidomide 75-87 MYC proto-oncogene, bHLH transcription factor Homo sapiens 199-204 34455369-4 2021 In three of four tested HTLV-1-infected cell lines, the cells treated with lenalidomide or pomalidomide exhibited mild growth suppression without apoptosis, which was associated with decreased IRF4, c-Myc, and phosphorylated STAT3 levels as well as enhanced SOCS3 expression. Lenalidomide 75-87 signal transducer and activator of transcription 3 Homo sapiens 225-230 34455369-4 2021 In three of four tested HTLV-1-infected cell lines, the cells treated with lenalidomide or pomalidomide exhibited mild growth suppression without apoptosis, which was associated with decreased IRF4, c-Myc, and phosphorylated STAT3 levels as well as enhanced SOCS3 expression. Lenalidomide 75-87 suppressor of cytokine signaling 3 Homo sapiens 258-263 34680233-6 2021 We have found that lenalidomide-mediated degradation of IKZF1 leads to activation of the G protein-coupled receptor 68 (GPR68)/calcium/calpain pro-apoptotic pathway and inhibition of the regulator of calcineurin 1 (RCAN1)/calcineurin pro-survival pathway in MDS and acute myeloid leukemia (AML). Lenalidomide 19-31 IKAROS family zinc finger 1 Homo sapiens 56-61 34680233-6 2021 We have found that lenalidomide-mediated degradation of IKZF1 leads to activation of the G protein-coupled receptor 68 (GPR68)/calcium/calpain pro-apoptotic pathway and inhibition of the regulator of calcineurin 1 (RCAN1)/calcineurin pro-survival pathway in MDS and acute myeloid leukemia (AML). Lenalidomide 19-31 G protein-coupled receptor 68 Homo sapiens 89-118 34680233-6 2021 We have found that lenalidomide-mediated degradation of IKZF1 leads to activation of the G protein-coupled receptor 68 (GPR68)/calcium/calpain pro-apoptotic pathway and inhibition of the regulator of calcineurin 1 (RCAN1)/calcineurin pro-survival pathway in MDS and acute myeloid leukemia (AML). Lenalidomide 19-31 G protein-coupled receptor 68 Homo sapiens 120-125 34680233-6 2021 We have found that lenalidomide-mediated degradation of IKZF1 leads to activation of the G protein-coupled receptor 68 (GPR68)/calcium/calpain pro-apoptotic pathway and inhibition of the regulator of calcineurin 1 (RCAN1)/calcineurin pro-survival pathway in MDS and acute myeloid leukemia (AML). Lenalidomide 19-31 regulator of calcineurin 1 Homo sapiens 187-213 34680233-6 2021 We have found that lenalidomide-mediated degradation of IKZF1 leads to activation of the G protein-coupled receptor 68 (GPR68)/calcium/calpain pro-apoptotic pathway and inhibition of the regulator of calcineurin 1 (RCAN1)/calcineurin pro-survival pathway in MDS and acute myeloid leukemia (AML). Lenalidomide 19-31 regulator of calcineurin 1 Homo sapiens 215-220 34583995-9 2021 Notably, USP15 is highly expressed in IMiD-resistant cells, and depletion of USP15 sensitizes these cells to lenalidomide. Lenalidomide 109-121 ubiquitin specific peptidase 15 Homo sapiens 9-14 34583995-9 2021 Notably, USP15 is highly expressed in IMiD-resistant cells, and depletion of USP15 sensitizes these cells to lenalidomide. Lenalidomide 109-121 ubiquitin specific peptidase 15 Homo sapiens 77-82 34503175-0 2021 miR-22 Modulates Lenalidomide Activity by Counteracting MYC Addiction in Multiple Myeloma. Lenalidomide 17-29 microRNA 22 Homo sapiens 0-6 34503175-8 2021 Interestingly, we found that IMiD lenalidomide increases miR-22 expression by reducing MYC repression and, most importantly, that the combination of lenalidomide with miR-22 mimics results in a synergistic direct and NK-mediated cytotoxic activity. Lenalidomide 34-46 microRNA 22 Homo sapiens 57-63 34503175-8 2021 Interestingly, we found that IMiD lenalidomide increases miR-22 expression by reducing MYC repression and, most importantly, that the combination of lenalidomide with miR-22 mimics results in a synergistic direct and NK-mediated cytotoxic activity. Lenalidomide 34-46 MYC proto-oncogene, bHLH transcription factor Homo sapiens 87-90 34503175-8 2021 Interestingly, we found that IMiD lenalidomide increases miR-22 expression by reducing MYC repression and, most importantly, that the combination of lenalidomide with miR-22 mimics results in a synergistic direct and NK-mediated cytotoxic activity. Lenalidomide 149-161 microRNA 22 Homo sapiens 57-63 34503175-9 2021 Conclusions: Taken together, our findings indicate that: (1) low miR-22 expression could represent a potential predictive biomarker of poor lenalidomide response in MM patients; and (2) miR-22 reduces MYC oncogenic activity, thus triggering a novel synthetic lethality loop, which sensitizes MM cells to lenalidomide. Lenalidomide 140-152 microRNA 22 Homo sapiens 65-71 34503175-9 2021 Conclusions: Taken together, our findings indicate that: (1) low miR-22 expression could represent a potential predictive biomarker of poor lenalidomide response in MM patients; and (2) miR-22 reduces MYC oncogenic activity, thus triggering a novel synthetic lethality loop, which sensitizes MM cells to lenalidomide. Lenalidomide 140-152 microRNA 22 Homo sapiens 186-192 34503175-9 2021 Conclusions: Taken together, our findings indicate that: (1) low miR-22 expression could represent a potential predictive biomarker of poor lenalidomide response in MM patients; and (2) miR-22 reduces MYC oncogenic activity, thus triggering a novel synthetic lethality loop, which sensitizes MM cells to lenalidomide. Lenalidomide 140-152 MYC proto-oncogene, bHLH transcription factor Homo sapiens 201-204 34503175-9 2021 Conclusions: Taken together, our findings indicate that: (1) low miR-22 expression could represent a potential predictive biomarker of poor lenalidomide response in MM patients; and (2) miR-22 reduces MYC oncogenic activity, thus triggering a novel synthetic lethality loop, which sensitizes MM cells to lenalidomide. Lenalidomide 304-316 microRNA 22 Homo sapiens 65-71 34503175-9 2021 Conclusions: Taken together, our findings indicate that: (1) low miR-22 expression could represent a potential predictive biomarker of poor lenalidomide response in MM patients; and (2) miR-22 reduces MYC oncogenic activity, thus triggering a novel synthetic lethality loop, which sensitizes MM cells to lenalidomide. Lenalidomide 304-316 microRNA 22 Homo sapiens 186-192 34503175-9 2021 Conclusions: Taken together, our findings indicate that: (1) low miR-22 expression could represent a potential predictive biomarker of poor lenalidomide response in MM patients; and (2) miR-22 reduces MYC oncogenic activity, thus triggering a novel synthetic lethality loop, which sensitizes MM cells to lenalidomide. Lenalidomide 304-316 MYC proto-oncogene, bHLH transcription factor Homo sapiens 201-204 34733611-4 2021 These peculiar mechanisms of action make TCL an attractive target for lenalidomide. Lenalidomide 70-82 ras homolog family member J Homo sapiens 41-44 34733611-5 2021 We have identified five clinical trials in which lenalidomide monotherapy was investigated to treat TCL, including cutaneous TCL (CTCL) and adult T-cell lymphoma/leukemia (ATLL). Lenalidomide 49-61 ras homolog family member J Homo sapiens 100-103 34733611-5 2021 We have identified five clinical trials in which lenalidomide monotherapy was investigated to treat TCL, including cutaneous TCL (CTCL) and adult T-cell lymphoma/leukemia (ATLL). Lenalidomide 49-61 ras homolog family member J Homo sapiens 125-128 34733611-11 2021 In conclusion, lenalidomide could represent a suitable treatment option for relapsed/refractory TCL, especially for neoplasms with a T-follicular helper origin, such as angioimmunoblastic TCL. Lenalidomide 15-27 ras homolog family member J Homo sapiens 96-99 34680570-0 2021 Lenalidomide and Pomalidomide Improve Function and Induce FcgammaRI/CD64 in Multiple Myeloma Neutrophils. Lenalidomide 0-12 Fc gamma receptor Ia Homo sapiens 58-67 34680570-0 2021 Lenalidomide and Pomalidomide Improve Function and Induce FcgammaRI/CD64 in Multiple Myeloma Neutrophils. Lenalidomide 0-12 Fc gamma receptor Ia Homo sapiens 68-72 34680570-10 2021 Similarly, in NDMM patients, lenalidomide but not bortezomib upregulated FcgammaRI/CD64 expression, improving phagocytic activity and oxidative bursta as tested in vitro. Lenalidomide 29-41 Fc gamma receptor Ia Homo sapiens 73-82 34680570-10 2021 Similarly, in NDMM patients, lenalidomide but not bortezomib upregulated FcgammaRI/CD64 expression, improving phagocytic activity and oxidative bursta as tested in vitro. Lenalidomide 29-41 Fc gamma receptor Ia Homo sapiens 83-87 34390739-6 2021 Treatment with thalidomide, lenalidomide and pomalidomide reduced peripheral nerve injury-induced proinflammatory cytokines (TNFalpha, IL-1beta and IL-6) in the ipsilateral spinal cords of neuropathic rats and LPS-treated primary microglial cells. Lenalidomide 28-40 tumor necrosis factor Rattus norvegicus 125-133 34390739-6 2021 Treatment with thalidomide, lenalidomide and pomalidomide reduced peripheral nerve injury-induced proinflammatory cytokines (TNFalpha, IL-1beta and IL-6) in the ipsilateral spinal cords of neuropathic rats and LPS-treated primary microglial cells. Lenalidomide 28-40 interleukin 1 alpha Rattus norvegicus 135-143 34390739-6 2021 Treatment with thalidomide, lenalidomide and pomalidomide reduced peripheral nerve injury-induced proinflammatory cytokines (TNFalpha, IL-1beta and IL-6) in the ipsilateral spinal cords of neuropathic rats and LPS-treated primary microglial cells. Lenalidomide 28-40 interleukin 6 Rattus norvegicus 148-152 34552051-6 2021 Lenalidomide refractoriness at first relapse was associated with inferior PFS2 (8.1 (95% CI: 6.4, 9.9) months vs. 19.9 (95% CI: 9.7, 30.2; p = 0.002) months in nonrefractory patients). Lenalidomide 0-12 GINS complex subunit 2 Homo sapiens 74-78 34262292-14 2021 Chidamide enhanced the effect of lenalidomide-induced degradation of IKZF1 and IKZF3 by elevating H2O2. Lenalidomide 33-45 IKAROS family zinc finger 1 Mus musculus 69-74 34316334-2 2021 Cereblon (CRBN)-targeting immunomodulatory agents (IMiDs ) lenalidomide (LEN) and pomalidomide (POM) play a central role in combination regimens due to their pleiotropic antitumor/immunomodulatory mechanisms that synergize with many anti-myeloma approved or developmental agents. Lenalidomide 59-71 cereblon Homo sapiens 10-14 34316334-2 2021 Cereblon (CRBN)-targeting immunomodulatory agents (IMiDs ) lenalidomide (LEN) and pomalidomide (POM) play a central role in combination regimens due to their pleiotropic antitumor/immunomodulatory mechanisms that synergize with many anti-myeloma approved or developmental agents. Lenalidomide 73-76 cereblon Homo sapiens 10-14 34405005-10 2021 Our results also suggested that lenalidomide might relieve relapsed lymphoma with mutations in NFKBIA 202C>T (p.Q68*) and NFKBIE 433A>T (p.K145*) by targeting NF-Kappa B signaling. Lenalidomide 32-44 NFKB inhibitor epsilon Homo sapiens 122-128 34405005-10 2021 Our results also suggested that lenalidomide might relieve relapsed lymphoma with mutations in NFKBIA 202C>T (p.Q68*) and NFKBIE 433A>T (p.K145*) by targeting NF-Kappa B signaling. Lenalidomide 32-44 nuclear factor kappa B subunit 1 Homo sapiens 159-169 34262292-14 2021 Chidamide enhanced the effect of lenalidomide-induced degradation of IKZF1 and IKZF3 by elevating H2O2. Lenalidomide 33-45 IKAROS family zinc finger 3 Mus musculus 79-84 34395218-16 2021 However, the NMDAR antagonist MK-801 (0.01 mg/kg, i.p) significantly reversed the anticonvulsive effects of lenalidomide (10 mg/kg). Lenalidomide 108-120 glutamate receptor, ionotropic, NMDA1 (zeta 1) Mus musculus 13-18 34395218-17 2021 Conclusions: Our study demonstrated a role for the NMDAR/NO pathway in the anticonvulsive effects of lenalidomide on the PTZ-induced clonic seizures in mice. Lenalidomide 101-113 glutamate receptor, ionotropic, NMDA1 (zeta 1) Mus musculus 51-56 34168051-4 2021 The sensitizing effect of IKZF1 loss-of-function was then validated and leveraged for combination treatment with lenalidomide. Lenalidomide 113-125 IKAROS family zinc finger 1 Homo sapiens 26-31 34168051-7 2021 Treating cells with tazemetostat and lenalidomide, an immunomodulatory drug that selectively degrades Ikaros and Aiolos, phenocopied the effects of the CRISPR/Cas9 screen. Lenalidomide 37-49 IKAROS family zinc finger 1 Homo sapiens 102-108 34168051-7 2021 Treating cells with tazemetostat and lenalidomide, an immunomodulatory drug that selectively degrades Ikaros and Aiolos, phenocopied the effects of the CRISPR/Cas9 screen. Lenalidomide 37-49 IKAROS family zinc finger 3 Homo sapiens 113-119 34168051-11 2021 Gene expression of key immune response factors such as IRF7 and DDX58 were induced in cells treated with lenalidomide and tazemetostat, with a concomitant increase of H3K27 acetylation at their promoters. Lenalidomide 105-117 interferon regulatory factor 7 Homo sapiens 55-59 34168051-11 2021 Gene expression of key immune response factors such as IRF7 and DDX58 were induced in cells treated with lenalidomide and tazemetostat, with a concomitant increase of H3K27 acetylation at their promoters. Lenalidomide 105-117 DExD/H-box helicase 58 Homo sapiens 64-69 34207079-4 2021 Here, we evaluated the expression of CRL4-CRBN complex proteins and their downstream targets with immunohistochemistry (IHC) staining in 130 bone marrow samples from MM patients treated with thalidomide or lenalidomide-based regimens. Lenalidomide 206-218 interleukin 17 receptor B Homo sapiens 37-41 34207079-4 2021 Here, we evaluated the expression of CRL4-CRBN complex proteins and their downstream targets with immunohistochemistry (IHC) staining in 130 bone marrow samples from MM patients treated with thalidomide or lenalidomide-based regimens. Lenalidomide 206-218 cereblon Homo sapiens 42-46 34117218-2 2021 We found that lenalidomide (LEN), a representative of immunomodulatory drugs (IMiDs), which have been long used for the treatment of multiple myeloma, specifically induced accumulation of Ik6 with the disappearance of functional isoforms within 24 h (i.e., abrupt and complete shut-down of the IKZF1 activity) in Ik6-positive Ph+ALL cells in a neddylation-dependent manner. Lenalidomide 14-26 IKAROS family zinc finger 1 Homo sapiens 294-299 34117218-2 2021 We found that lenalidomide (LEN), a representative of immunomodulatory drugs (IMiDs), which have been long used for the treatment of multiple myeloma, specifically induced accumulation of Ik6 with the disappearance of functional isoforms within 24 h (i.e., abrupt and complete shut-down of the IKZF1 activity) in Ik6-positive Ph+ALL cells in a neddylation-dependent manner. Lenalidomide 28-31 IKAROS family zinc finger 1 Homo sapiens 294-299 34088671-4 2021 RNA sequencing of MM patient samples shows nearly universal ERK pathway activation at relapse on lenalidomide maintenance therapy, confirming its clinical relevance. Lenalidomide 97-109 mitogen-activated protein kinase 1 Homo sapiens 60-63 35359004-0 2022 SIRPalpha+ macrophages are increased in patients with FL who progress or relapse after frontline lenalidomide and rituximab. Lenalidomide 97-109 signal regulatory protein alpha Homo sapiens 0-9 35583604-1 2022 PURPOSE: Cereblon (CRBN), a substrate receptor of the E3 ubiquitin ligase complex CRL4CRBN, is the target of the small molecules lenalidomide (Len) and avadomide (Ava). Lenalidomide 129-141 cereblon Homo sapiens 19-23 35467582-0 2022 Combination Therapy of an Antibody Specific for Transferrin Receptor 1 (ch128.1/IgG1) With Bortezomib or Lenalidomide Results in Increased Survival in an In Vivo Model of Human Multiple Myeloma: A Brief Communication. Lenalidomide 105-117 transferrin receptor Homo sapiens 48-70 35467582-0 2022 Combination Therapy of an Antibody Specific for Transferrin Receptor 1 (ch128.1/IgG1) With Bortezomib or Lenalidomide Results in Increased Survival in an In Vivo Model of Human Multiple Myeloma: A Brief Communication. Lenalidomide 105-117 LOC105243590 Mus musculus 80-84 35583604-1 2022 PURPOSE: Cereblon (CRBN), a substrate receptor of the E3 ubiquitin ligase complex CRL4CRBN, is the target of the small molecules lenalidomide (Len) and avadomide (Ava). Lenalidomide 129-141 interleukin 17 receptor B Homo sapiens 82-90 35583604-1 2022 PURPOSE: Cereblon (CRBN), a substrate receptor of the E3 ubiquitin ligase complex CRL4CRBN, is the target of the small molecules lenalidomide (Len) and avadomide (Ava). Lenalidomide 143-146 cereblon Homo sapiens 19-23 35583604-1 2022 PURPOSE: Cereblon (CRBN), a substrate receptor of the E3 ubiquitin ligase complex CRL4CRBN, is the target of the small molecules lenalidomide (Len) and avadomide (Ava). Lenalidomide 143-146 interleukin 17 receptor B Homo sapiens 82-90 35595613-6 2022 On the other hand, the efficiency of Thalidomide derivates in myelodysplastic syndromes (MDS), such as Lenalidomide, acted as the starting point for the development of targeted leukemia-associated protein degradation molecules. Lenalidomide 103-115 ribosomal protein L34 Homo sapiens 177-204 35586751-7 2022 Moreover, we speculate that lenalidomide improves cognitive function by regulating intracranial inflammation via multiple pathways, not only by TNF-alpha blocking. Lenalidomide 28-40 tumor necrosis factor Homo sapiens 144-153 35512188-0 2022 Lenalidomide promotes the development of TP53-mutated therapy-related myeloid neoplasms. Lenalidomide 0-12 tumor protein p53 Homo sapiens 41-45 35512188-3 2022 In a systematic analysis of 416 patients with t-MN and detailed prior exposure history, we found that TP53 mutations were significantly associated with prior treatment with thalidomide analogs, specifically lenalidomide. Lenalidomide 207-219 tumor protein p53 Homo sapiens 102-106 35512188-4 2022 We demonstrated experimentally that lenalidomide treatment provides a selective advantage to Trp53-mutant hematopoietic stem and progenitor cells (HSPCs) in vitro and in vivo, the effect of which was specific to Trp53-mutant HSPCs and was not observed in HSPCs with other CH mutations. Lenalidomide 36-48 tumor protein p53 Homo sapiens 93-98 35512188-4 2022 We demonstrated experimentally that lenalidomide treatment provides a selective advantage to Trp53-mutant hematopoietic stem and progenitor cells (HSPCs) in vitro and in vivo, the effect of which was specific to Trp53-mutant HSPCs and was not observed in HSPCs with other CH mutations. Lenalidomide 36-48 tumor protein p53 Homo sapiens 212-217 35512188-6 2022 These findings highlight the role of lenalidomide treatment in promoting TP53-mutated t-MNs and offer a potential alternative strategy to mitigate the risk of t-MN development. Lenalidomide 37-49 tumor protein p53 Homo sapiens 73-77 35064823-1 2022 PURPOSE: This study sought to compare the efficacy of prophylactic long-acting and standard granulocyte colony-stimulating factor (G-CSF) on febrile neutropenia, early infections, and treatment delay in patients with newly diagnosed multiple myeloma (MM) receiving the therapeutic regimen of bortezomib, lenalidomide, and dexamethasone (VRd). Lenalidomide 304-316 colony stimulating factor 3 Homo sapiens 131-136 34979567-0 2022 SORT1/LAMP2-mediated Extracellular Vesicle Secretion and Cell Adhesion Are Linked to Lenalidomide Resistance in Multiple Myeloma. Lenalidomide 85-97 sortilin 1 Homo sapiens 0-5 34979567-0 2022 SORT1/LAMP2-mediated Extracellular Vesicle Secretion and Cell Adhesion Are Linked to Lenalidomide Resistance in Multiple Myeloma. Lenalidomide 85-97 lysosomal associated membrane protein 2 Homo sapiens 6-11 34979567-8 2022 Together, our findings reveal a novel mechanism of lenalidomide resistance in MM mediated by EV secretion and cell adhesion via SORT1 and LAMP2. Lenalidomide 51-63 sortilin 1 Homo sapiens 128-133 34979567-8 2022 Together, our findings reveal a novel mechanism of lenalidomide resistance in MM mediated by EV secretion and cell adhesion via SORT1 and LAMP2. Lenalidomide 51-63 lysosomal associated membrane protein 2 Homo sapiens 138-143 35380709-8 2022 RSK2 or CREB1 inhibition increased CRBN levels and were synergic with lenalidomide in inducing cell death, especially in CD56-expressing MM cells. Lenalidomide 70-82 ribosomal protein S6 kinase A3 Homo sapiens 0-4 35380709-8 2022 RSK2 or CREB1 inhibition increased CRBN levels and were synergic with lenalidomide in inducing cell death, especially in CD56-expressing MM cells. Lenalidomide 70-82 cAMP responsive element binding protein 1 Homo sapiens 8-13 35380709-8 2022 RSK2 or CREB1 inhibition increased CRBN levels and were synergic with lenalidomide in inducing cell death, especially in CD56-expressing MM cells. Lenalidomide 70-82 cereblon Homo sapiens 35-39 35380709-8 2022 RSK2 or CREB1 inhibition increased CRBN levels and were synergic with lenalidomide in inducing cell death, especially in CD56-expressing MM cells. Lenalidomide 70-82 neural cell adhesion molecule 1 Homo sapiens 121-125 35433994-7 2022 In order to understand the possible mechanisms of lenalidomide in CAR-T therapy, CAR-T copies of peripheral blood were regularly detected by quantitative real-time polymerase chain reaction, and an in vitro test was also conducted. Lenalidomide 50-62 CXADR pseudogene 1 Homo sapiens 66-69 35089607-1 2022 In the phase 3 APOLLO trial, daratumumab in combination with pomalidomide and dexamethasone (D-Pd) significantly reduced the rate of disease progression or death by 37% relative to Pd alone in patients with relapsed/refractory multiple myeloma (RRMM) who had received >=1 prior line of therapy including lenalidomide and a proteasome inhibitor. Lenalidomide 304-316 dihydropyrimidine dehydrogenase Homo sapiens 93-97 35194739-0 2022 Integrin beta5 and beta7 expression in lenalidomide-resistant multiple myeloma cells. Lenalidomide 39-51 integrin subunit beta 5 Homo sapiens 0-14 35194739-0 2022 Integrin beta5 and beta7 expression in lenalidomide-resistant multiple myeloma cells. Lenalidomide 39-51 immunoglobulin kappa variable 2D-24 (non-functional) Homo sapiens 19-24 35338347-6 2022 Overexpression of IRF4 and c-Myc is a major mechanism of Len resistance. Lenalidomide 57-60 interferon regulatory factor 4 Homo sapiens 18-22 35338347-6 2022 Overexpression of IRF4 and c-Myc is a major mechanism of Len resistance. Lenalidomide 57-60 MYC proto-oncogene, bHLH transcription factor Homo sapiens 27-32 35321428-3 2022 Bortezomib and lenalidomide activate caspase-8 and promote the apoptosis of myeloma cells. Lenalidomide 15-27 caspase 8 Homo sapiens 37-46 35321428-4 2022 However, caspase-8 inhibition potentiated the antiproliferative effect of lenalidomide and bortezomib in myeloma cells, suggesting that caspase-8 could regulate proliferation and apoptosis in the opposite pathway. Lenalidomide 74-86 caspase 8 Homo sapiens 136-145 35433994-7 2022 In order to understand the possible mechanisms of lenalidomide in CAR-T therapy, CAR-T copies of peripheral blood were regularly detected by quantitative real-time polymerase chain reaction, and an in vitro test was also conducted. Lenalidomide 50-62 CXADR pseudogene 1 Homo sapiens 81-84 35433994-9 2022 Furthermore, one case responding to CAR-T therapy initially but suffering a relapse shortly achieved complete remission again after lenalidomide exposure, with an increase in the number of CAR-T copies detected. Lenalidomide 132-144 CXADR pseudogene 1 Homo sapiens 36-39 35433994-10 2022 The in vitro test showed that lenalidomide could delay the exhaustion of CAR-T cells. Lenalidomide 30-42 CXADR pseudogene 1 Homo sapiens 73-76 35433994-12 2022 We confirmed that lenalidomide maintenance can improve patients" OS, and the delayed exhaustion of CAR-T cells may contribute to this OS benefit. Lenalidomide 18-30 CXADR pseudogene 1 Homo sapiens 99-102 35197447-0 2022 Proteomic profiling reveals CDK6 upregulation as a targetable resistance mechanism for lenalidomide in multiple myeloma. Lenalidomide 87-99 cyclin dependent kinase 6 Homo sapiens 28-32 35092513-6 2022 CDK9 inhibitors were able to induce dose-dependent cytotoxicity in Doxorubicin-resistant, Lenalidomide-resistant and Bortezomib-resistant cell lines. Lenalidomide 90-102 cyclin dependent kinase 9 Homo sapiens 0-4 35092513-9 2022 Synergistic effects between CDK9 inhibitors and either Venetoclax, Bortezomib, Lenalidomide or Erlotinib have been proven and are awaiting verification in clinical trials. Lenalidomide 79-91 cyclin dependent kinase 9 Homo sapiens 28-32 34768284-0 2022 MCT1 is a Predictive Marker for Lenalidomide Maintenance Therapy in Multiple Myeloma. Lenalidomide 32-44 solute carrier family 16 member 1 Homo sapiens 0-4 34768284-4 2022 Patients with high gene expression levels of MCT1 showed significantly reduced PFS (31.9 vs. 48.2 months in MCT1high vs. MCT1low, P=.03) and OS (75.9 months vs. not reached (NR) months in MCT1high vs. MCT1low; P=.001) in case of lenalidomide maintenance, whereas MCT1 expression had no significant impact on PFS or OS in patients with bortezomib maintenance. Lenalidomide 229-241 solute carrier family 16 member 1 Homo sapiens 45-49 34768284-6 2022 Functional validation showed that MCT1 overexpression in human MM cell lines significantly reduced efficacy of lenalidomide, while no change was observed upon bortezomib treatment, both in vitro and in an MM xenograft model. Lenalidomide 111-123 solute carrier family 16 member 1 Homo sapiens 34-38 34768284-7 2022 Together, we establish MCT1-expression as a predictive marker for response to lenalidomide-based maintenance treatment. Lenalidomide 78-90 solute carrier family 16 member 1 Homo sapiens 23-27 34978798-1 2022 The thalidomide analogue lenalidomide (Len) is a clinical therapeutic that alters the substrate engagement of cereblon (CRBN), a substrate receptor for the CRL4 E3 ubiquitin ligase. Lenalidomide 25-37 cereblon Homo sapiens 120-124 34978798-1 2022 The thalidomide analogue lenalidomide (Len) is a clinical therapeutic that alters the substrate engagement of cereblon (CRBN), a substrate receptor for the CRL4 E3 ubiquitin ligase. Lenalidomide 25-37 interleukin 17 receptor B Homo sapiens 156-160 34978798-1 2022 The thalidomide analogue lenalidomide (Len) is a clinical therapeutic that alters the substrate engagement of cereblon (CRBN), a substrate receptor for the CRL4 E3 ubiquitin ligase. Lenalidomide 39-42 cereblon Homo sapiens 120-124 34978798-1 2022 The thalidomide analogue lenalidomide (Len) is a clinical therapeutic that alters the substrate engagement of cereblon (CRBN), a substrate receptor for the CRL4 E3 ubiquitin ligase. Lenalidomide 39-42 interleukin 17 receptor B Homo sapiens 156-160 34978798-5 2022 eIF3i is directly labeled by pLen and forms a ternary complex with CRBN in the presence of Len across several epithelial cell lines but is itself not ubiquitylated or degraded. Lenalidomide 91-94 eukaryotic translation initiation factor 3 subunit I Homo sapiens 0-5 34978798-5 2022 eIF3i is directly labeled by pLen and forms a ternary complex with CRBN in the presence of Len across several epithelial cell lines but is itself not ubiquitylated or degraded. Lenalidomide 91-94 cereblon Homo sapiens 67-71