PMID-sentid Pub_year Sent_text compound_name comp_offset prot_official_name organism prot_offset 32737681-5 2021 From the screening of approved drug library, three antiviral agents ritonavir, nelfinavir and saquinavir were predicted to be the most potent Mpro inhibitors. Ritonavir 68-77 NEWENTRY Severe acute respiratory syndrome-related coronavirus 142-146 33551833-13 2020 Two antiviral drugs (Elbasvir and Ritonavir) were included; Elbasvir and Ritonavir formed van der Waals (VDW) interactions with surrounding residues to bind with CSF3, and Elbasvir and Ritonavir significantly inhibited CSF3 protein expression. Ritonavir 73-82 colony stimulating factor 3 Homo sapiens 162-166 33309661-5 2021 A model of the SARS-CoV-2-nsp10-nsp14 complex bound to substrate RNA showed that the ritonavir binding site overlaps with that of the 3" nucleotide of substrate RNA. Ritonavir 85-94 ORF1a polyprotein;ORF1ab polyprotein Severe acute respiratory syndrome coronavirus 2 26-31 33536797-2 2021 Oral docetaxel is co-administered with the cytochrome P450 3A4 and P-glycoprotein inhibitor ritonavir to increase oral bioavailability. Ritonavir 92-101 ATP binding cassette subfamily B member 1 Homo sapiens 67-81 33513992-6 2021 The addition of RTV can inhibit the P-gp and CYP3A4-mediated metabolism of PTX, thus aiding in reversing MDR and sensitizing the cells toward PTX. Ritonavir 16-19 phosphoglycolate phosphatase Homo sapiens 36-40 33513992-6 2021 The addition of RTV can inhibit the P-gp and CYP3A4-mediated metabolism of PTX, thus aiding in reversing MDR and sensitizing the cells toward PTX. Ritonavir 16-19 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 45-51 33551833-13 2020 Two antiviral drugs (Elbasvir and Ritonavir) were included; Elbasvir and Ritonavir formed van der Waals (VDW) interactions with surrounding residues to bind with CSF3, and Elbasvir and Ritonavir significantly inhibited CSF3 protein expression. Ritonavir 73-82 colony stimulating factor 3 Homo sapiens 219-223 33551833-13 2020 Two antiviral drugs (Elbasvir and Ritonavir) were included; Elbasvir and Ritonavir formed van der Waals (VDW) interactions with surrounding residues to bind with CSF3, and Elbasvir and Ritonavir significantly inhibited CSF3 protein expression. Ritonavir 73-82 colony stimulating factor 3 Homo sapiens 162-166 33551833-13 2020 Two antiviral drugs (Elbasvir and Ritonavir) were included; Elbasvir and Ritonavir formed van der Waals (VDW) interactions with surrounding residues to bind with CSF3, and Elbasvir and Ritonavir significantly inhibited CSF3 protein expression. Ritonavir 73-82 colony stimulating factor 3 Homo sapiens 219-223 33003981-5 2020 Moreover, ritonavir increased vascular NADPH oxidase 1, aortic H2O2 levels as well as interleukin-1beta, GATA3 (GATA binding protein 3), the macrophage marker (F4/80), and C-C chemokine receptor type 5 (CCR5) expression. Ritonavir 10-19 GATA binding protein 3 Mus musculus 105-110 33467005-0 2021 Rational Design of CYP3A4 Inhibitors: A One-Atom Linker Elongation in Ritonavir-Like Compounds Leads to a Marked Improvement in the Binding Strength. Ritonavir 70-79 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 19-25 33467005-3 2021 We utilized a rational inhibitor design to investigate the structure-activity relationships in the analogues of ritonavir, the most potent CYP3A4 inhibitor in clinical use. Ritonavir 112-121 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 139-145 33022569-2 2020 Under this scenario, the combination of two HIV-1 protease inhibitors, lopinavir and ritonavir, has attracted attention since they have been previously employed against the SARS-CoV main proteinase (Mpro) and exhibited some signs of effectiveness. Ritonavir 85-94 NEWENTRY Severe acute respiratory syndrome-related coronavirus 199-203 33022569-6 2020 Our study provides the structural and energetic basis of the inhibitory properties of lopinavir and ritonavir on SARS-CoV Mpro and SARS-CoV2 Mpro, allowing us to identify two FDA-approved drugs that can be used against SARS-CoV2 Mpro. Ritonavir 100-109 NEWENTRY Severe acute respiratory syndrome-related coronavirus 122-126 33022569-6 2020 Our study provides the structural and energetic basis of the inhibitory properties of lopinavir and ritonavir on SARS-CoV Mpro and SARS-CoV2 Mpro, allowing us to identify two FDA-approved drugs that can be used against SARS-CoV2 Mpro. Ritonavir 100-109 NEWENTRY Severe acute respiratory syndrome-related coronavirus 141-145 33022569-6 2020 Our study provides the structural and energetic basis of the inhibitory properties of lopinavir and ritonavir on SARS-CoV Mpro and SARS-CoV2 Mpro, allowing us to identify two FDA-approved drugs that can be used against SARS-CoV2 Mpro. Ritonavir 100-109 NEWENTRY Severe acute respiratory syndrome-related coronavirus 141-145 33003981-9 2020 Conversely, selective increases in endothelial leptin signaling with protein tyrosine phosphatase deletion blunted ritonavir-induced endothelial dysfunction. Ritonavir 115-124 leptin Mus musculus 47-53 33387249-7 2021 MM-GBSA calculations demonstrated promising binding affinities of TMC-310911 and ritonavir towards SARS-CoV-2 Mpro, with binding energy values of - 52.8 and - 49.4 kcal/mol, respectively. Ritonavir 81-90 NEWENTRY Severe acute respiratory syndrome-related coronavirus 110-114 33387249-8 2021 Surpass potentialities of TMC-310911 and ritonavir are returned to their capabilities of forming multiple hydrogen bonds with the proximal amino acids inside Mpro"s binding site. Ritonavir 41-50 NEWENTRY Severe acute respiratory syndrome-related coronavirus 158-162 33387249-9 2021 Structural and energetic analyses involving root-mean-square deviation, binding energy per-frame, center-of-mass distance, and hydrogen bond length demonstrated the stability of TMC-310911 and ritonavir inside the Mpro"s active site over the 50 ns MD simulation. Ritonavir 193-202 NEWENTRY Severe acute respiratory syndrome-related coronavirus 214-218 33269470-3 2021 Since ritonavir is a strong inhibitor of CYP3A and nifedipine is mainly metabolized via CYP3A, the combination of ritonavir and nifedipine can potentially cause drug-drug interactions. Ritonavir 6-15 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 41-46 33269470-3 2021 Since ritonavir is a strong inhibitor of CYP3A and nifedipine is mainly metabolized via CYP3A, the combination of ritonavir and nifedipine can potentially cause drug-drug interactions. Ritonavir 114-123 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 41-46 33269470-3 2021 Since ritonavir is a strong inhibitor of CYP3A and nifedipine is mainly metabolized via CYP3A, the combination of ritonavir and nifedipine can potentially cause drug-drug interactions. Ritonavir 114-123 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 88-93 33269470-10 2021 Model simulations showed that the inhibitory effect of ritonavir on CYP3A4 increased the Cmax of nifedipine 17.92-48.85-fold and the AUC 63.30-84.01-fold at steady state and decreased the SBP by more than 40 mmHg. Ritonavir 55-64 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 68-74 33003981-0 2020 HIV Protease Inhibitor Ritonavir Impairs Endothelial Function Via Reduction in Adipose Mass and Endothelial Leptin Receptor-Dependent Increases in NADPH Oxidase 1 (Nox1), C-C Chemokine Receptor Type 5 (CCR5), and Inflammation. Ritonavir 23-32 NADPH oxidase 1 Mus musculus 147-162 33003981-0 2020 HIV Protease Inhibitor Ritonavir Impairs Endothelial Function Via Reduction in Adipose Mass and Endothelial Leptin Receptor-Dependent Increases in NADPH Oxidase 1 (Nox1), C-C Chemokine Receptor Type 5 (CCR5), and Inflammation. Ritonavir 23-32 NADPH oxidase 1 Mus musculus 164-168 33003981-0 2020 HIV Protease Inhibitor Ritonavir Impairs Endothelial Function Via Reduction in Adipose Mass and Endothelial Leptin Receptor-Dependent Increases in NADPH Oxidase 1 (Nox1), C-C Chemokine Receptor Type 5 (CCR5), and Inflammation. Ritonavir 23-32 chemokine (C-C motif) receptor 5 Mus musculus 171-200 33003981-5 2020 Moreover, ritonavir increased vascular NADPH oxidase 1, aortic H2O2 levels as well as interleukin-1beta, GATA3 (GATA binding protein 3), the macrophage marker (F4/80), and C-C chemokine receptor type 5 (CCR5) expression. Ritonavir 10-19 GATA binding protein 3 Mus musculus 112-134 33003981-0 2020 HIV Protease Inhibitor Ritonavir Impairs Endothelial Function Via Reduction in Adipose Mass and Endothelial Leptin Receptor-Dependent Increases in NADPH Oxidase 1 (Nox1), C-C Chemokine Receptor Type 5 (CCR5), and Inflammation. Ritonavir 23-32 chemokine (C-C motif) receptor 5 Mus musculus 202-206 33003981-3 2020 Herein, we tested the hypothesis that ritonavir-mediated lipoatrophy causes endothelial dysfunction via reducing endothelial leptin signaling. Ritonavir 38-47 leptin Homo sapiens 125-131 33003981-4 2020 Methods and Results Long-term (4 weeks) but not short-term (3 days) treatment with ritonavir reduced body weight, fat mass, and leptin levels and induced endothelial dysfunction in mice. Ritonavir 83-92 leptin Mus musculus 128-134 33003981-5 2020 Moreover, ritonavir increased vascular NADPH oxidase 1, aortic H2O2 levels as well as interleukin-1beta, GATA3 (GATA binding protein 3), the macrophage marker (F4/80), and C-C chemokine receptor type 5 (CCR5) expression. Ritonavir 10-19 NADPH oxidase 1 Mus musculus 39-54 33003981-5 2020 Moreover, ritonavir increased vascular NADPH oxidase 1, aortic H2O2 levels as well as interleukin-1beta, GATA3 (GATA binding protein 3), the macrophage marker (F4/80), and C-C chemokine receptor type 5 (CCR5) expression. Ritonavir 10-19 adhesion G protein-coupled receptor E1 Mus musculus 160-165 33003981-5 2020 Moreover, ritonavir increased vascular NADPH oxidase 1, aortic H2O2 levels as well as interleukin-1beta, GATA3 (GATA binding protein 3), the macrophage marker (F4/80), and C-C chemokine receptor type 5 (CCR5) expression. Ritonavir 10-19 chemokine (C-C motif) receptor 5 Mus musculus 172-201 33003981-5 2020 Moreover, ritonavir increased vascular NADPH oxidase 1, aortic H2O2 levels as well as interleukin-1beta, GATA3 (GATA binding protein 3), the macrophage marker (F4/80), and C-C chemokine receptor type 5 (CCR5) expression. Ritonavir 10-19 chemokine (C-C motif) receptor 5 Mus musculus 203-207 33003981-5 2020 Moreover, ritonavir increased vascular NADPH oxidase 1, aortic H2O2 levels as well as interleukin-1beta, GATA3 (GATA binding protein 3), the macrophage marker (F4/80), and C-C chemokine receptor type 5 (CCR5) expression. Ritonavir 10-19 interleukin 1 beta Mus musculus 86-103 33003981-6 2020 Reactive oxygen species scavenging with tempol restored endothelial function, and both NADPH oxidase 1 and CCR5 deletion in mice protected from ritonavir-mediated endothelial dysfunction and vascular inflammation. Ritonavir 144-153 NADPH oxidase 1 Mus musculus 87-102 33003981-6 2020 Reactive oxygen species scavenging with tempol restored endothelial function, and both NADPH oxidase 1 and CCR5 deletion in mice protected from ritonavir-mediated endothelial dysfunction and vascular inflammation. Ritonavir 144-153 chemokine (C-C motif) receptor 5 Mus musculus 107-111 33003981-7 2020 Remarkably, leptin infusion markedly improved endothelial function and significantly reduced vascular NADPH oxidase 1, interleukin-1beta, GATA3, F4/80, and CCR5 levels in ritonavir-treated animals. Ritonavir 171-180 leptin Mus musculus 12-18 33003981-7 2020 Remarkably, leptin infusion markedly improved endothelial function and significantly reduced vascular NADPH oxidase 1, interleukin-1beta, GATA3, F4/80, and CCR5 levels in ritonavir-treated animals. Ritonavir 171-180 NADPH oxidase 1 Mus musculus 102-117 33003981-7 2020 Remarkably, leptin infusion markedly improved endothelial function and significantly reduced vascular NADPH oxidase 1, interleukin-1beta, GATA3, F4/80, and CCR5 levels in ritonavir-treated animals. Ritonavir 171-180 interleukin 1 beta Mus musculus 119-136 33003981-7 2020 Remarkably, leptin infusion markedly improved endothelial function and significantly reduced vascular NADPH oxidase 1, interleukin-1beta, GATA3, F4/80, and CCR5 levels in ritonavir-treated animals. Ritonavir 171-180 chemokine (C-C motif) receptor 5 Mus musculus 156-160 33003981-10 2020 Conclusions All together, these data indicate that ritonavir-associated endothelial dysfunction is a direct consequence of a reduction in adiposity and leptin secretion, which decreases endothelial leptin signaling and leads to a NADPH oxidase 1-induced, CCR5-mediated reduction in NO bioavailability. Ritonavir 51-60 leptin Mus musculus 152-158 33003981-10 2020 Conclusions All together, these data indicate that ritonavir-associated endothelial dysfunction is a direct consequence of a reduction in adiposity and leptin secretion, which decreases endothelial leptin signaling and leads to a NADPH oxidase 1-induced, CCR5-mediated reduction in NO bioavailability. Ritonavir 51-60 leptin Mus musculus 198-204 33003981-10 2020 Conclusions All together, these data indicate that ritonavir-associated endothelial dysfunction is a direct consequence of a reduction in adiposity and leptin secretion, which decreases endothelial leptin signaling and leads to a NADPH oxidase 1-induced, CCR5-mediated reduction in NO bioavailability. Ritonavir 51-60 NADPH oxidase 1 Mus musculus 230-245 33003981-10 2020 Conclusions All together, these data indicate that ritonavir-associated endothelial dysfunction is a direct consequence of a reduction in adiposity and leptin secretion, which decreases endothelial leptin signaling and leads to a NADPH oxidase 1-induced, CCR5-mediated reduction in NO bioavailability. Ritonavir 51-60 chemokine (C-C motif) receptor 5 Mus musculus 255-259 33063648-4 2022 The comparative analysis of the MM/GBSA results revealed that the binding affinity (DeltaGbind) of EGCG and K7G for CYP3A4 and ABCB1 are higher than LUT and EGA and fall between the DeltaGbind of the inhibitors of CYP3A4 and ABCB1 (Ritonavir (strong inhibitor) and Lopinavir (moderate inhibitor)). Ritonavir 232-241 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 116-122 33063648-4 2022 The comparative analysis of the MM/GBSA results revealed that the binding affinity (DeltaGbind) of EGCG and K7G for CYP3A4 and ABCB1 are higher than LUT and EGA and fall between the DeltaGbind of the inhibitors of CYP3A4 and ABCB1 (Ritonavir (strong inhibitor) and Lopinavir (moderate inhibitor)). Ritonavir 232-241 ATP binding cassette subfamily B member 1 Homo sapiens 127-132 33030105-0 2022 Molecular dynamics analysis predicts ritonavir and naloxegol strongly block the SARS-CoV-2 spike protein-hACE2 binding. Ritonavir 37-46 surface glycoprotein Severe acute respiratory syndrome coronavirus 2 91-96 33030105-0 2022 Molecular dynamics analysis predicts ritonavir and naloxegol strongly block the SARS-CoV-2 spike protein-hACE2 binding. Ritonavir 37-46 angiotensin converting enzyme 2 Homo sapiens 105-110 33030105-9 2022 Finally, a long MD analysis revealed two select candidate ligands, including ritonavir and naloxegol, highly stabilizing those key residues engaged in RBD-hACE2 interaction. Ritonavir 77-86 angiotensin converting enzyme 2 Homo sapiens 155-160 32759267-8 2020 ATV/RTV also impaired virus-induced enhancement of IL-6 and TNF-alpha levels. Ritonavir 4-7 interleukin 6 Homo sapiens 51-55 33021083-9 2021 In this study, 37 patients were treated with up to twice-daily 30-mg paclitaxel combined with twice-daily 100-mg ritonavir (MP5/r 30-30/100-100) in 9 dose levels. Ritonavir 113-122 progesterone receptor membrane component 1 Homo sapiens 124-129 33312066-8 2020 Based on high alternative allele frequencies in population and the functional effect of the variants, ABCB1 rs1045642 and rs2032582 could be relevant for reduced clearance of azithromycin, lopinavir and ritonavir drugs and UGT1A7 rs17868323 for hyperbilirubinemia in ritonavir treated COVID-19 patients in Serbian population. Ritonavir 203-212 ATP binding cassette subfamily B member 1 Homo sapiens 102-107 33312066-8 2020 Based on high alternative allele frequencies in population and the functional effect of the variants, ABCB1 rs1045642 and rs2032582 could be relevant for reduced clearance of azithromycin, lopinavir and ritonavir drugs and UGT1A7 rs17868323 for hyperbilirubinemia in ritonavir treated COVID-19 patients in Serbian population. Ritonavir 203-212 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 223-229 33312066-8 2020 Based on high alternative allele frequencies in population and the functional effect of the variants, ABCB1 rs1045642 and rs2032582 could be relevant for reduced clearance of azithromycin, lopinavir and ritonavir drugs and UGT1A7 rs17868323 for hyperbilirubinemia in ritonavir treated COVID-19 patients in Serbian population. Ritonavir 267-276 ATP binding cassette subfamily B member 1 Homo sapiens 102-107 33312066-8 2020 Based on high alternative allele frequencies in population and the functional effect of the variants, ABCB1 rs1045642 and rs2032582 could be relevant for reduced clearance of azithromycin, lopinavir and ritonavir drugs and UGT1A7 rs17868323 for hyperbilirubinemia in ritonavir treated COVID-19 patients in Serbian population. Ritonavir 267-276 UDP glucuronosyltransferase family 1 member A7 Homo sapiens 223-229 33244500-4 2020 Results: A 41-year-old man with human immunodeficiency virus infection on stable antiretroviral therapy that included ritonavir, a cytochrome P450 3A4 inhibitor, presented with new onset diabetes and development of overt cushingoid features over a 4-week period. Ritonavir 118-127 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 131-150 32759267-8 2020 ATV/RTV also impaired virus-induced enhancement of IL-6 and TNF-alpha levels. Ritonavir 4-7 tumor necrosis factor Homo sapiens 60-69 32557601-3 2020 Ritonavir (a strong CYP3A inhibitor) increased ribociclib 400 mg single-dose AUC by 3.2-fold, whereas rifampin (a strong CYP3A inducer) decreased ribociclib AUC by 89% in healthy volunteers (HV). Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 20-25 32583097-10 2020 In addition, we have identified novel FXR antagonistic effects of well-studied hepatotoxic drugs, including bosentan, tolcapone and ritonavir. Ritonavir 132-141 nuclear receptor subfamily 1 group H member 4 Homo sapiens 38-41 32924827-8 2022 The results indicated that, Ritonavir has the highest potency to block SARS-CoV-2 main protease and human TMPRSS2, a host cell factor that aids viral infection. Ritonavir 28-37 transmembrane serine protease 2 Homo sapiens 106-113 32427480-1 2020 Ritonavir is a well-known CYP3A4 and CYP2D6 enzyme inhibitor, frequently used to assess the drug-drug interaction (DDI) liability of susceptible drugs. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 26-32 32725720-1 2020 Oral administration of docetaxel in combination with the CYP3A4 inhibitor ritonavir is used in clinical trials to improve oral bioavailability of docetaxel. Ritonavir 74-83 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 57-63 32427480-1 2020 Ritonavir is a well-known CYP3A4 and CYP2D6 enzyme inhibitor, frequently used to assess the drug-drug interaction (DDI) liability of susceptible drugs. Ritonavir 0-9 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 37-43 32167970-4 2020 We induced this in mice exposed to the PI ritonavir (RTV), and intervened with low-dose inhaled carbon monoxide (CO), activating erythroid 2-related factor (Nrf2)-associated anti-oxidant pathways. Ritonavir 42-51 nuclear factor, erythroid derived 2, like 2 Mus musculus 157-161 32803017-8 2020 In this case, an extensive macular atrophy (with complete loss of photoreceptor, RPE and choriocapillaris layers) and subsequent cone-rod dysfunction appeared after 18 years of ritonavir exposure. Ritonavir 177-186 ribulose-5-phosphate-3-epimerase Homo sapiens 81-84 32597315-7 2021 Phenformin, quercetin, and ritonavir all demonstrated prospective binding affinities for COVID-19 PLpro over 50 ns MD simulations, with binding energy values of -56.6, -40.9, and -37.6 kcal/mol, respectively. Ritonavir 27-36 ORF1a polyprotein;ORF1ab polyprotein Severe acute respiratory syndrome coronavirus 2 98-103 32167970-4 2020 We induced this in mice exposed to the PI ritonavir (RTV), and intervened with low-dose inhaled carbon monoxide (CO), activating erythroid 2-related factor (Nrf2)-associated anti-oxidant pathways. Ritonavir 53-56 nuclear factor, erythroid derived 2, like 2 Mus musculus 157-161 32167970-10 2020 RESULTS: RTV induced glomerular and tubular injury, elevating urinary KIM-1 (p = 0.004). Ritonavir 9-12 hepatitis A virus cellular receptor 1 Mus musculus 70-75 32167970-15 2020 RTV-treated HIV+ women had further increases in cystatin C (n = 20; p = 0.05), with parallel elevation of HO-1. Ritonavir 0-3 cystatin C Homo sapiens 48-58 32167970-15 2020 RTV-treated HIV+ women had further increases in cystatin C (n = 20; p = 0.05), with parallel elevation of HO-1. Ritonavir 0-3 heme oxygenase 1 Homo sapiens 106-110 31735989-1 2020 BACKGROUND: The potential for clinically significant drug interactions (CSDIs) for patients taking ritonavir and cobicistat is high because of their powerful pharmacokinetic effect on the cytochrome P450 (CYP) enzyme system, most notably their inhibitory effect on CYP3A4. Ritonavir 99-108 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 265-271 32044230-0 2020 An increase in side-group hydrophobicity largely improves the potency of ritonavir-like inhibitors of CYP3A4. Ritonavir 73-82 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 102-108 32490327-0 2020 Ritonavir and Lopinavir Suppress RCE1 and CAAX Rab Proteins Sensitizing the Liver to Organelle Stress and Injury. Ritonavir 0-9 Ras converting CAAX endopeptidase 1 Homo sapiens 33-37 32490327-2 2020 Here, we found through total RNA sequencing that the transcription of a host protease Ras converting CAAX endopeptidase 1 (RCE1) was altered in HepG2 cells treated with anti-HIV protease inhibitors, ritonavir and lopinavir. Ritonavir 199-208 Ras converting CAAX endopeptidase 1 Homo sapiens 86-121 32490327-2 2020 Here, we found through total RNA sequencing that the transcription of a host protease Ras converting CAAX endopeptidase 1 (RCE1) was altered in HepG2 cells treated with anti-HIV protease inhibitors, ritonavir and lopinavir. Ritonavir 199-208 Ras converting CAAX endopeptidase 1 Homo sapiens 123-127 32490327-5 2020 Knocking down Rce1 with RNA interference increased ritonavir and lopinavir-induced cell death as well as expression of Golgi stress response markers, TFE3, HSP47 and GCP60, in both primary mouse hepatocytes and mouse liver, and deteriorated alcohol-induced alanine aminotransferase (ALT) and fatty liver injury in mice. Ritonavir 51-60 Ras converting CAAX endopeptidase 1 Mus musculus 14-18 32140912-11 2020 CYP3A4 inhibitors such as ritonavir or cobicistat may increase the chance of this adverse effect. Ritonavir 26-35 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-6 31923087-8 2020 Similarly, ritonavir AUC0-12 was lower during the third trimester [GMR 0.65 (IQR 0.52-0.82); P = 0.007] compared with postpartum, whereas its apparent clearance was higher during the third trimester [GMR 1.53 (IQR 1.22-1.92); P = 0.008] compared with postpartum. Ritonavir 11-20 colony stimulating factor 2 receptor subunit alpha Homo sapiens 67-70 31923087-8 2020 Similarly, ritonavir AUC0-12 was lower during the third trimester [GMR 0.65 (IQR 0.52-0.82); P = 0.007] compared with postpartum, whereas its apparent clearance was higher during the third trimester [GMR 1.53 (IQR 1.22-1.92); P = 0.008] compared with postpartum. Ritonavir 11-20 colony stimulating factor 2 receptor subunit alpha Homo sapiens 200-203 32044230-2 2020 We utilize a rational inhibitor design to decipher structure-activity relationships in analogues of ritonavir, a highly potent CYP3A4 inhibitor marketed as pharmacoenhancer. Ritonavir 100-109 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 127-133 32044230-3 2020 Analysis of compounds with the R1 side-group as phenyl or naphthalene and R2 as indole or naphthalene in different stereo configuration showed that (i) analogues with the R2-naphthalene tend to bind tighter and inhibit CYP3A4 more potently than the R2-phenyl/indole containing counterparts; (ii) stereochemistry becomes a more important contributing factor, as the bulky side-groups limit the ability to optimize protein-ligand interactions; (iii) the relationship between the R1/R2 configuration and preferential binding to CYP3A4 is complex and depends on the side-group functionality/interplay and backbone spacing; and (iv) three inhibitors, 5a-b and 7d, were superior to ritonavir (IC50 of 0.055-0.085 muM vs. 0.130 muM, respectively). Ritonavir 676-685 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 219-225 31531807-6 2019 Ritonavir-boosted paritaprevir/ombitasvir was prescribed to five recipients with Crcl < 30 mL/min/1.73 m2. Ritonavir 0-9 CD59 molecule (CD59 blood group) Homo sapiens 97-102 31971958-0 2020 HIV antiretroviral drugs, dolutegravir, maraviroc and ritonavir-boosted atazanavir use different pathways to affect inflammation, senescence and insulin sensitivity in human coronary endothelial cells. Ritonavir 54-63 insulin Homo sapiens 145-152 31481446-4 2019 Lopinavir, ritonavir, saquinavir, atazanavir, maraviroc, ledipasvir, and daclatasvir inhibited the efflux of a model ABCB1 substrate, rhodamine 123 (RHD123), in Caco-2 cells and rat-derived PCIS. Ritonavir 11-20 ATP binding cassette subfamily B member 1 Homo sapiens 117-122 31696528-9 2019 ARV therapy with ritonavir confers an enantioselective interaction between the enantiomers of bupivacaine and placental P-gp, producing greater inhibition of efflux transport of the R-bupivacaine enantiomer. Ritonavir 17-26 ATP binding cassette subfamily B member 1 Homo sapiens 120-124 31410542-3 2019 Therefore, we investigated the long-term prognosis after thyroidectomy and (adjuvant) initial radioactive iodine therapy (RIT) of OPTC compared to PTC. Ritonavir 122-125 opticin Homo sapiens 130-134 31410542-16 2019 Patients suffering from OPTC present with the same clinical long-term outcome indifferent to PTC after (adjuvant) initial RIT after matching. Ritonavir 122-125 opticin Homo sapiens 24-28 31481446-5 2019 Lopinavir, ritonavir, saquinavir, and atazanavir also significantly inhibited RHD123 efflux in human-derived PCIS, while possible interindividual variability was observed in the inhibition of intestinal ABCB1 by maraviroc, ledipasvir, and daclatasvir. Ritonavir 11-20 ATP binding cassette subfamily B member 1 Homo sapiens 203-208 31456404-5 2019 These methods were applied to a soluble N-terminally truncated CYP3A4 form, and the results show that there are few changes in the average structure upon binding ketoconazole, ritonavir, or midazolam. Ritonavir 176-185 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 63-69 31548507-4 2019 Indeed, molecular modeling of PLCgamma1 with SLP76 peptide and with previously reported inhibitors (ritonavir, anethole, daunorubicin, diflunisal, and rosiglitazone) facilitated the identification of the common critical residues (Gln805, Arg806, Asp808, Glu809, Asp825, Gly827, and Trp828) as well as the quantification of their interaction through binding energies calculations. Ritonavir 100-109 phospholipase C gamma 1 Homo sapiens 30-39 31517073-4 2019 We here present a 70-year-old man who was anti-NT5C1A antibody-positive in association with IBM and chronic hepatitis C. The initial treatment of ombitasvir/paritaprevir/ritonavir for his chronic hepatitis C was successful; however, his symptoms of IBM did not improve. Ritonavir 170-179 5'-nucleotidase, cytosolic IA Homo sapiens 47-53 31034908-7 2019 Ritonavir and cobicistat are MATE1 and MATE2K inhibitors with IC50 values of 3.1 and 90 muM (ritonavir), and 4.4 and 3.2 muM (cobicistat), respectively. Ritonavir 0-9 solute carrier family 47 member 1 Homo sapiens 29-34 31034908-7 2019 Ritonavir and cobicistat are MATE1 and MATE2K inhibitors with IC50 values of 3.1 and 90 muM (ritonavir), and 4.4 and 3.2 muM (cobicistat), respectively. Ritonavir 0-9 solute carrier family 47 member 2 Homo sapiens 39-45 31034908-7 2019 Ritonavir and cobicistat are MATE1 and MATE2K inhibitors with IC50 values of 3.1 and 90 muM (ritonavir), and 4.4 and 3.2 muM (cobicistat), respectively. Ritonavir 0-9 latexin Homo sapiens 88-91 31034908-7 2019 Ritonavir and cobicistat are MATE1 and MATE2K inhibitors with IC50 values of 3.1 and 90 muM (ritonavir), and 4.4 and 3.2 muM (cobicistat), respectively. Ritonavir 0-9 latexin Homo sapiens 121-124 31034908-8 2019 However, the unbound cytosolic concentrations (Cu,cytosol) of ritonavir and cobicistat in human renal proximal tubule epithelial cells, 0.065 and 0.10 muM, respectively, after incubation with the clinical maximum total plasma concentrations at pharmacoenhancer doses does not support inhibition in vivo; Cu,cytosol >30 fold lower than IC50s. Ritonavir 62-71 latexin Homo sapiens 151-154 31345777-9 2019 In a nude mouse model, MSLN-targeted RIT treatment of SW48 CRC tumors resulted in a significant decrease in tumor volume. Ritonavir 37-40 mesothelin Mus musculus 23-27 31266750-7 2019 This transplacental transport was saturable and significantly diminished after the addition of the ABCB1/Abcb1 inhibitors elacridar, zosuquidar, and ritonavir. Ritonavir 149-158 ATP binding cassette subfamily B member 1A Rattus norvegicus 99-104 31266750-7 2019 This transplacental transport was saturable and significantly diminished after the addition of the ABCB1/Abcb1 inhibitors elacridar, zosuquidar, and ritonavir. Ritonavir 149-158 ATP binding cassette subfamily B member 1A Rattus norvegicus 105-110 30997718-5 2019 The expression of p53, as one of the key regulators of senescence, was upregulated after 48 hours of Ritonavir-NO treatment only in metastatic B16F10 cells, ranking it as a late-response event. Ritonavir 101-110 transformation related protein 53, pseudogene Mus musculus 18-21 30985556-7 2019 In participants on ritonavir-boosted protease inhibitor at entry, mean reductions in fasting LDL-C and non-HDL-C at week 24 were significantly greater for DOR/3TC/TDF vs Baseline Regimen (P < 0.0001). Ritonavir 19-28 sex determining region Y Homo sapiens 155-166 30951643-1 2019 INTRODUCTION: Drugs used in HIV treatment; all protease inhibitors, some non-nucleoside reverse transcriptase inhibitors, and pharmacoenhancers ritonavir and cobicistat can inhibit cytochrome P450 (CYP) enzymes. Ritonavir 144-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 181-196 31059280-7 2019 Treatments of these foam cells with ritonavir, nelfinavir, and saquinavir at least doubled cholestryl ester accumulation ( P=0.02, 0.0009, and 0.02, respectively), whereas RNA silencing of the AR-RGN top key driver, PQBP1 (polyglutamine binding protein 1), significantly curbed cholestryl ester accumulation following treatment with any of these ART drugs by >37% ( P<0.05). Ritonavir 36-45 polyglutamine binding protein 1 Homo sapiens 216-221 31059280-7 2019 Treatments of these foam cells with ritonavir, nelfinavir, and saquinavir at least doubled cholestryl ester accumulation ( P=0.02, 0.0009, and 0.02, respectively), whereas RNA silencing of the AR-RGN top key driver, PQBP1 (polyglutamine binding protein 1), significantly curbed cholestryl ester accumulation following treatment with any of these ART drugs by >37% ( P<0.05). Ritonavir 36-45 polyglutamine binding protein 1 Homo sapiens 223-254 30951643-1 2019 INTRODUCTION: Drugs used in HIV treatment; all protease inhibitors, some non-nucleoside reverse transcriptase inhibitors, and pharmacoenhancers ritonavir and cobicistat can inhibit cytochrome P450 (CYP) enzymes. Ritonavir 144-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 198-201 31133852-0 2019 miRNA-218 Targets Lipin-1 and Glucose Transporter Type 4 Genes in 3T3-L1 Cells Treated With Lopinavir/Ritonavir. Ritonavir 102-111 lipin 1 Mus musculus 18-25 31124411-4 2019 Results: Higher concentrations of ATV and RTV were significantly associated with CYP3A5 6986 GG and SLCO1B1 521 TC or CC. Ritonavir 42-45 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 81-87 31124411-4 2019 Results: Higher concentrations of ATV and RTV were significantly associated with CYP3A5 6986 GG and SLCO1B1 521 TC or CC. Ritonavir 42-45 solute carrier organic anion transporter family member 1B1 Homo sapiens 100-107 31133852-0 2019 miRNA-218 Targets Lipin-1 and Glucose Transporter Type 4 Genes in 3T3-L1 Cells Treated With Lopinavir/Ritonavir. Ritonavir 102-111 solute carrier family 2 (facilitated glucose transporter), member 4 Mus musculus 30-56 31133852-6 2019 Objectives: To determine whether lopinavir/ritonavir (LPV/RTV) can modulate lipogenesis in adipocytes affecting miRNA-218 and lipin-1 mRNA expression, and to investigate the functional link between miRNA-218 and GLUT-4 mRNA expression. Ritonavir 43-52 lipin 1 Mus musculus 126-133 31133852-10 2019 The anti-miR-218 transfection of 3T3-L1 cells significantly ameliorated the adipogenic dysfunction and restored mRNA levels of lipin-1 and GLUT-4 consequent to LPV/RTV treatment. Ritonavir 164-167 microRNA 218 Mus musculus 9-16 31133852-10 2019 The anti-miR-218 transfection of 3T3-L1 cells significantly ameliorated the adipogenic dysfunction and restored mRNA levels of lipin-1 and GLUT-4 consequent to LPV/RTV treatment. Ritonavir 164-167 lipin 1 Mus musculus 127-134 31133852-10 2019 The anti-miR-218 transfection of 3T3-L1 cells significantly ameliorated the adipogenic dysfunction and restored mRNA levels of lipin-1 and GLUT-4 consequent to LPV/RTV treatment. Ritonavir 164-167 solute carrier family 2 (facilitated glucose transporter), member 4 Mus musculus 139-145 30783000-4 2019 Coadministration of doravirine with CYP3A inhibitors (ritonavir or ketoconazole) increased doravirine exposure approximately 3-fold. Ritonavir 54-63 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 36-41 30642080-0 2019 Ginsenoside Rb1 Blocks Ritonavir-Induced Oxidative Stress and eNOS Downregulation through Activation of Estrogen Receptor-Beta and Upregulation of SOD in Human Endothelial Cells. Ritonavir 23-32 RB transcriptional corepressor 1 Homo sapiens 12-15 30912932-0 2019 Structure-Activity Relationships of Rationally Designed Ritonavir Analogues: Impact of Side-Group Stereochemistry, Headgroup Spacing, and Backbone Composition on the Interaction with CYP3A4. Ritonavir 56-65 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 183-189 30912932-1 2019 In a continuing effort to identify structural attributes required for strong binding and potent inhibition of human drug-metabolizing CYP3A4, we designed ten ritonavir-like analogues differing in the side-group stereochemistry, backbone atomic composition, and headgroup spacing. Ritonavir 158-167 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 134-140 30959935-3 2019 Norvir oral powder, 100 mg (NOP) was developed to replace OS. Ritonavir 0-6 prepronociceptin Homo sapiens 29-32 30660696-0 2019 Oral coadministration of elacridar and ritonavir enhances brain accumulation and oral availability of the novel ALK/ROS1 inhibitor lorlatinib. Ritonavir 39-48 anaplastic lymphoma kinase Mus musculus 112-115 30660696-0 2019 Oral coadministration of elacridar and ritonavir enhances brain accumulation and oral availability of the novel ALK/ROS1 inhibitor lorlatinib. Ritonavir 39-48 Ros1 proto-oncogene Mus musculus 116-120 30541118-0 2019 Improvement in insulin sensitivity and serum leptin concentration after the switch from a ritonavir-boosted PI to raltegravir or dolutegravir in non-diabetic HIV-infected patients. Ritonavir 90-99 insulin Homo sapiens 15-22 30541118-0 2019 Improvement in insulin sensitivity and serum leptin concentration after the switch from a ritonavir-boosted PI to raltegravir or dolutegravir in non-diabetic HIV-infected patients. Ritonavir 90-99 leptin Homo sapiens 45-51 30926609-0 2019 Active-site differences between substrate-free and ritonavir-bound cytochrome P450 (CYP) 3A5 reveal plasticity differences between CYP3A5 and CYP3A4. Ritonavir 51-60 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 67-92 30926609-0 2019 Active-site differences between substrate-free and ritonavir-bound cytochrome P450 (CYP) 3A5 reveal plasticity differences between CYP3A5 and CYP3A4. Ritonavir 51-60 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 131-137 30926609-0 2019 Active-site differences between substrate-free and ritonavir-bound cytochrome P450 (CYP) 3A5 reveal plasticity differences between CYP3A5 and CYP3A4. Ritonavir 51-60 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 142-148 30926609-6 2019 We have previously shown that the structure of the CYP3A5-ritonavir complex differs substantially from that of the CYP3A4-ritonavir complex. Ritonavir 58-67 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 51-57 30926609-6 2019 We have previously shown that the structure of the CYP3A5-ritonavir complex differs substantially from that of the CYP3A4-ritonavir complex. Ritonavir 58-67 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 115-121 30926609-6 2019 We have previously shown that the structure of the CYP3A5-ritonavir complex differs substantially from that of the CYP3A4-ritonavir complex. Ritonavir 122-131 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 51-57 30926609-6 2019 We have previously shown that the structure of the CYP3A5-ritonavir complex differs substantially from that of the CYP3A4-ritonavir complex. Ritonavir 122-131 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 115-121 30926609-10 2019 Comparison with the CYP3A5-ritonavir complex confirmed conserved CYP3A5 structural features and indicated differences in plasticity between CYP3A4 and CYP3A5 that favor alternative ritonavir conformations. Ritonavir 27-36 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 20-26 30926609-10 2019 Comparison with the CYP3A5-ritonavir complex confirmed conserved CYP3A5 structural features and indicated differences in plasticity between CYP3A4 and CYP3A5 that favor alternative ritonavir conformations. Ritonavir 27-36 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 65-71 30926609-10 2019 Comparison with the CYP3A5-ritonavir complex confirmed conserved CYP3A5 structural features and indicated differences in plasticity between CYP3A4 and CYP3A5 that favor alternative ritonavir conformations. Ritonavir 27-36 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 140-146 30926609-10 2019 Comparison with the CYP3A5-ritonavir complex confirmed conserved CYP3A5 structural features and indicated differences in plasticity between CYP3A4 and CYP3A5 that favor alternative ritonavir conformations. Ritonavir 27-36 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 65-71 30926609-10 2019 Comparison with the CYP3A5-ritonavir complex confirmed conserved CYP3A5 structural features and indicated differences in plasticity between CYP3A4 and CYP3A5 that favor alternative ritonavir conformations. Ritonavir 181-190 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 20-26 30926609-10 2019 Comparison with the CYP3A5-ritonavir complex confirmed conserved CYP3A5 structural features and indicated differences in plasticity between CYP3A4 and CYP3A5 that favor alternative ritonavir conformations. Ritonavir 181-190 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 140-146 30782819-12 2019 Treatment with an 1.5 mg/kg LMB-75 QODx5 anti-BCMA RIT beginning on day 4 caused the complete disappearance of tumors for 80 days. Ritonavir 51-54 tumor necrosis factor receptor superfamily, member 17 Mus musculus 46-50 30660696-5 2019 Oral coadministration of the CYP3A inhibitor ritonavir caused reversion to the AUC0-8h levels seen in wild-type and Cyp3a-/- mice, without altering the relative tissue distribution of lorlatinib. Ritonavir 45-54 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 29-34 30660696-5 2019 Oral coadministration of the CYP3A inhibitor ritonavir caused reversion to the AUC0-8h levels seen in wild-type and Cyp3a-/- mice, without altering the relative tissue distribution of lorlatinib. Ritonavir 45-54 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 116-121 30660696-6 2019 Moreover, simultaneous pharmacological inhibition of P-glycoprotein and CYP3A4 with oral elacridar and ritonavir in CYP3A4-humanized mice profoundly increased lorlatinib brain concentrations, but not its oral availability or other relative tissue distribution. Ritonavir 103-112 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 116-122 30450781-2 2019 In this phase 3b open-label study, we assessed changes in IFN-stimulated genes (ISGs) in non-cirrhotic treatment-naive or pegIFN/RBV-experienced HCV-GT1a-infected patients receiving paritaprevir/ritonavir/ombitasvir + dasabuvir + ribavirin (PrOD + R) for 12 weeks. Ritonavir 195-204 interferon alpha 1 Homo sapiens 58-61 30642080-0 2019 Ginsenoside Rb1 Blocks Ritonavir-Induced Oxidative Stress and eNOS Downregulation through Activation of Estrogen Receptor-Beta and Upregulation of SOD in Human Endothelial Cells. Ritonavir 23-32 estrogen receptor 2 Homo sapiens 104-126 30642080-0 2019 Ginsenoside Rb1 Blocks Ritonavir-Induced Oxidative Stress and eNOS Downregulation through Activation of Estrogen Receptor-Beta and Upregulation of SOD in Human Endothelial Cells. Ritonavir 23-32 superoxide dismutase 1 Homo sapiens 147-150 29696643-2 2019 Ritonavir and clopidogrel are inhibitors of CYP3A4 and CYP2C8, respectively. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 44-50 29696643-2 2019 Ritonavir and clopidogrel are inhibitors of CYP3A4 and CYP2C8, respectively. Ritonavir 0-9 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 55-61 31143861-8 2019 The mean increase in CD4 count (57.16%) after 6 months was statistically significant (P < 0.05) with tenofovir + lamivudine + atazanavir/ritonavir regimen in forty patients. Ritonavir 140-149 CD4 molecule Homo sapiens 21-24 30546876-8 2018 In addition, ritonavir inhibited the expression of Runx2, a key regulator of osteoblast differentiation, in the early period of osteoblast differentiation. Ritonavir 13-22 runt related transcription factor 2 Mus musculus 51-56 30322872-4 2018 Frequencies of CD4+ cells were significantly affected by ritonavir (CD69+ P=3E-05; CD134 P=4E-06; CD25+ P=E-07; central memory P=0.02; effector P=6E-03; effector memory P=6E-05). Ritonavir 57-66 CD4 antigen Mus musculus 15-18 30322872-4 2018 Frequencies of CD4+ cells were significantly affected by ritonavir (CD69+ P=3E-05; CD134 P=4E-06; CD25+ P=E-07; central memory P=0.02; effector P=6E-03; effector memory P=6E-05). Ritonavir 57-66 CD69 antigen Mus musculus 68-72 30322872-4 2018 Frequencies of CD4+ cells were significantly affected by ritonavir (CD69+ P=3E-05; CD134 P=4E-06; CD25+ P=E-07; central memory P=0.02; effector P=6E-03; effector memory P=6E-05). Ritonavir 57-66 interleukin 2 receptor, alpha chain Mus musculus 98-102 30322872-10 2018 Mice benefited from treatment with ritonavir as indicated by significantly decreased colon (P=7E-04) and histological (P=1E-04) scores, frequencies of M2 monocytes (CD14+ CD163; P=0.02), and Glu levels (P=2E-05). Ritonavir 35-44 CD14 antigen Mus musculus 165-169 30326221-0 2018 Formononetin and biochanin A protects against ritonavir induced hepatotoxicity via modulation of NfkappaB/pAkt signaling molecules. Ritonavir 46-55 B cell linker Homo sapiens 17-28 30326221-0 2018 Formononetin and biochanin A protects against ritonavir induced hepatotoxicity via modulation of NfkappaB/pAkt signaling molecules. Ritonavir 46-55 nuclear factor kappa B subunit 1 Homo sapiens 97-105 30326221-5 2018 Hence, the present study was inquested to ascertain the effect of Formononetin (FMN) and Biochanin A (BCA) on RIT induced hepatotoxicity. Ritonavir 110-113 B cell linker Homo sapiens 89-100 30326221-5 2018 Hence, the present study was inquested to ascertain the effect of Formononetin (FMN) and Biochanin A (BCA) on RIT induced hepatotoxicity. Ritonavir 110-113 B cell linker Homo sapiens 102-105 30326221-9 2018 KEY FINDINGS: FMN and BCA ameliorated the increased levels of biochemical markers of liver, attenuated the RIT induced Bax, caspase-3, NFkappaB and eNOS activation and persuaded the Bcl2 and pAkt level. Ritonavir 107-110 B cell linker Homo sapiens 22-25 30326221-9 2018 KEY FINDINGS: FMN and BCA ameliorated the increased levels of biochemical markers of liver, attenuated the RIT induced Bax, caspase-3, NFkappaB and eNOS activation and persuaded the Bcl2 and pAkt level. Ritonavir 107-110 BCL2 associated X, apoptosis regulator Homo sapiens 119-122 30326221-9 2018 KEY FINDINGS: FMN and BCA ameliorated the increased levels of biochemical markers of liver, attenuated the RIT induced Bax, caspase-3, NFkappaB and eNOS activation and persuaded the Bcl2 and pAkt level. Ritonavir 107-110 caspase 3 Homo sapiens 124-133 30326221-9 2018 KEY FINDINGS: FMN and BCA ameliorated the increased levels of biochemical markers of liver, attenuated the RIT induced Bax, caspase-3, NFkappaB and eNOS activation and persuaded the Bcl2 and pAkt level. Ritonavir 107-110 nuclear factor kappa B subunit 1 Homo sapiens 135-143 30044899-0 2018 Amenamevir: Studies of Potential CYP3A-Mediated Pharmacokinetic Interactions With Midazolam, Cyclosporine, and Ritonavir in Healthy Volunteers. Ritonavir 111-120 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 33-38 30044899-5 2018 (3) Ritonavir (inhibitor of CYP3A): When given with single doses of ritonavir 600 mg, geometric mean Cmax of amenamevir after 400-mg and 1200-mg single doses was, respectively, about 1.4 and 1.6 times higher, and geometric mean AUC0- about 2.6 and 3.3 times higher, than after amenamevir alone. Ritonavir 4-13 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 28-33 30044899-5 2018 (3) Ritonavir (inhibitor of CYP3A): When given with single doses of ritonavir 600 mg, geometric mean Cmax of amenamevir after 400-mg and 1200-mg single doses was, respectively, about 1.4 and 1.6 times higher, and geometric mean AUC0- about 2.6 and 3.3 times higher, than after amenamevir alone. Ritonavir 68-77 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 28-33 30158154-7 2018 Similarly, treatment with the human immunodeficiency virus protease inhibitors lopinavir and ritonavir reduced the accumulation of Lamin-A at nuclear membranes and preserved nuclear membrane compliance after mechanical ventilation, mimicking the protective phenotype of Zmpste24-/- animals. Ritonavir 93-102 lamin A/C Homo sapiens 131-138 30429604-4 2018 A large-scale screen revealed that the HIV protease inhibitor ritonavir is a potent disruptor of the CD95-PLCgamma1 interaction. Ritonavir 62-71 Fas cell surface death receptor Homo sapiens 101-105 30429604-4 2018 A large-scale screen revealed that the HIV protease inhibitor ritonavir is a potent disruptor of the CD95-PLCgamma1 interaction. Ritonavir 62-71 phospholipase C gamma 1 Homo sapiens 106-115 30429604-5 2018 A structure-activity relationship approach highlighted that ritonavir is a peptidomimetic that shares structural characteristics with CID with respect to docking to PLCgamma1. Ritonavir 60-69 phospholipase C gamma 1 Homo sapiens 165-174 29943426-6 2018 This P450 was identified by screening of actinobacterial strains for amodiaquine and ritonavir metabolizing activities, followed by genome sequencing and expression of the annotated S. platensis P450s in Escherichia coli. Ritonavir 85-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 5-9 30098863-9 2018 Inhibition of MDR1, MRP4 and/or HIF-1alpha with ritonavir (RTV) reconstitutes AhR function in Th17-cells, enhancing therapeutic effects of UCB in dextran-sulfate-sodium-induced experimental colitis. Ritonavir 48-57 ATP binding cassette subfamily B member 1 Homo sapiens 14-18 30098863-9 2018 Inhibition of MDR1, MRP4 and/or HIF-1alpha with ritonavir (RTV) reconstitutes AhR function in Th17-cells, enhancing therapeutic effects of UCB in dextran-sulfate-sodium-induced experimental colitis. Ritonavir 48-57 hypoxia inducible factor 1 subunit alpha Homo sapiens 32-42 30098863-9 2018 Inhibition of MDR1, MRP4 and/or HIF-1alpha with ritonavir (RTV) reconstitutes AhR function in Th17-cells, enhancing therapeutic effects of UCB in dextran-sulfate-sodium-induced experimental colitis. Ritonavir 48-57 aryl hydrocarbon receptor Homo sapiens 78-81 30098863-9 2018 Inhibition of MDR1, MRP4 and/or HIF-1alpha with ritonavir (RTV) reconstitutes AhR function in Th17-cells, enhancing therapeutic effects of UCB in dextran-sulfate-sodium-induced experimental colitis. Ritonavir 59-62 ATP binding cassette subfamily B member 1 Homo sapiens 14-18 30098863-9 2018 Inhibition of MDR1, MRP4 and/or HIF-1alpha with ritonavir (RTV) reconstitutes AhR function in Th17-cells, enhancing therapeutic effects of UCB in dextran-sulfate-sodium-induced experimental colitis. Ritonavir 59-62 hypoxia inducible factor 1 subunit alpha Homo sapiens 32-42 30098863-9 2018 Inhibition of MDR1, MRP4 and/or HIF-1alpha with ritonavir (RTV) reconstitutes AhR function in Th17-cells, enhancing therapeutic effects of UCB in dextran-sulfate-sodium-induced experimental colitis. Ritonavir 59-62 aryl hydrocarbon receptor Homo sapiens 78-81 30158154-7 2018 Similarly, treatment with the human immunodeficiency virus protease inhibitors lopinavir and ritonavir reduced the accumulation of Lamin-A at nuclear membranes and preserved nuclear membrane compliance after mechanical ventilation, mimicking the protective phenotype of Zmpste24-/- animals. Ritonavir 93-102 zinc metallopeptidase STE24 Homo sapiens 270-278 29283173-6 2018 The plasma exposure to triptolide and (5R)-5-hydroxytriptolide in the rats was significantly increased when co-administered with the CYP3a inhibitor ritonavir (30 mg/kg, po) with the values of AUC0- (area under the plasma concentration-time curve from time zero extrapolated to infinity) being increased by 6.84 and 1.83 times, respectively. Ritonavir 149-158 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 133-138 29649076-9 2018 Coadministration with P-gp/BCRP inhibitors such as cobicistat or PI-based regimens (ATV + RTV, LPV/r, or DRV + RTV) resulted in a range of 6%-183% increases in TAF and 105%-316% increases in TFV exposure, whereas coadministration with a P-gp inducer, efavirenz, resulted in a 15%-24% decrease in TAF and TFV exposure. Ritonavir 90-93 ATP binding cassette subfamily B member 1 Homo sapiens 22-26 29649076-9 2018 Coadministration with P-gp/BCRP inhibitors such as cobicistat or PI-based regimens (ATV + RTV, LPV/r, or DRV + RTV) resulted in a range of 6%-183% increases in TAF and 105%-316% increases in TFV exposure, whereas coadministration with a P-gp inducer, efavirenz, resulted in a 15%-24% decrease in TAF and TFV exposure. Ritonavir 111-114 ATP binding cassette subfamily B member 1 Homo sapiens 22-26 29851709-2 2018 Coadministration with strong CYP3A inhibitors (such as ketoconazole, posaconazole, and ritonavir) is contraindicated due to the risk of sedation and movement disorders from high levels of lurasidone. Ritonavir 87-96 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 29-34 30018638-6 2018 Patients with GD who underwent radioiodine therapy (RIT) had a lower lymphocytic expression level of p27Kip1, and those who took beta-blockers had higher expression levels of BID (BH3-interacting domain) and a lower Ki-67 expression level in thyrocytes than those who did not. Ritonavir 52-55 cyclin dependent kinase inhibitor 1B Homo sapiens 101-108 29288514-1 2018 BACKGROUND & AIMS: Limited data have shown high efficacy of co-formulated ombitasvir/paritaprevir/ritonavir (OBV/PTV/r) in the treatment of hepatitis C virus (HCV) genotype (GT)-4, and combined with dasabuvir (DSV) in GT1 patients, with chronic kidney disease (CKD) stages 4-5 (<30 mL/min/1.73 m2 ). Ritonavir 102-111 beta-1,4-galactosyltransferase 1 Homo sapiens 222-225 28891378-0 2018 In vitro inhibition of human UGT isoforms by ritonavir and cobicistat. Ritonavir 45-54 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 29-32 28891378-4 2018 However, the inhibitory effect of ritonavir and cobicistat on human UDP glucuronosyltransferase (UGT) enzymes in Phase II metabolism is not established. Ritonavir 34-43 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 68-95 28891378-4 2018 However, the inhibitory effect of ritonavir and cobicistat on human UDP glucuronosyltransferase (UGT) enzymes in Phase II metabolism is not established. Ritonavir 34-43 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 97-100 28891378-5 2018 This study evaluated the inhibition of human UGT isoforms by ritonavir versus cobicistat. Ritonavir 61-70 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 45-48 29869311-1 2018 Ritonavir is an anti-viral compound that has also been employed extensively as a CYP3A4 and P-glycoprotein (Pgp) inhibitor to boost the pharmacokinetic performance of compounds that undergo first pass metabolism. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 81-87 29869311-1 2018 Ritonavir is an anti-viral compound that has also been employed extensively as a CYP3A4 and P-glycoprotein (Pgp) inhibitor to boost the pharmacokinetic performance of compounds that undergo first pass metabolism. Ritonavir 0-9 ATP binding cassette subfamily B member 1 Homo sapiens 92-106 29869311-1 2018 Ritonavir is an anti-viral compound that has also been employed extensively as a CYP3A4 and P-glycoprotein (Pgp) inhibitor to boost the pharmacokinetic performance of compounds that undergo first pass metabolism. Ritonavir 0-9 ATP binding cassette subfamily B member 1 Homo sapiens 108-111 29848702-7 2018 CONCLUSION: The effectiveness of combination of ritonavir and delanzomib appears to be due to the induction of ER stress and inhibition of the mTOR pathway. Ritonavir 48-57 mechanistic target of rapamycin kinase Homo sapiens 143-147 29451681-1 2018 Ritonavir is one of several ketoconazole alternatives used to evaluate strong CYP3A4 inhibition potential in clinical drug-drug interaction (DDI) studies. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 78-84 29318452-0 2018 Correction to: CSF inflammatory markers and neurocognitive function after addition of maraviroc to monotherapy darunavir/ritonavir in stable HIV patients: the CINAMMON study. Ritonavir 121-130 colony stimulating factor 2 Homo sapiens 15-18 29272369-7 2018 Initiation with a nonrecommended nonnucleoside reverse transcriptase inhibitor-based vs a ritonavir-boosted protease inhibitor-based regimen resulted in a much smaller gain of around 100 CD4 cells/muL after 1 year. Ritonavir 90-99 CD4 molecule Homo sapiens 187-190 29476044-3 2018 Plasma drug concentrations are boosted by CYP3A inhibitors such as cobisistat and ritonavir (RTV). Ritonavir 82-91 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 42-47 29476044-3 2018 Plasma drug concentrations are boosted by CYP3A inhibitors such as cobisistat and ritonavir (RTV). Ritonavir 93-96 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 42-47 29691457-2 2018 To determine whether adaptations occur in response to GLUT inhibition in the failing adult heart, we chronically treated TG9 mice, a transgenic model of dilated cardiomyopathy and heart failure, with the GLUT inhibitor ritonavir. Ritonavir 219-228 solute carrier family 1 (glial high affinity glutamate transporter), member 3 Mus musculus 204-208 29691457-5 2018 GLUT1 and -12 protein expression was significantly increased in left ventricular (LV) myocardium in ritonavir-treated animals. Ritonavir 100-109 solute carrier family 1 (glial high affinity glutamate transporter), member 3 Mus musculus 0-5 29427135-2 2018 Ritonavir (RTV) is a potent mechanism-based and reversible CYP3A inhibitor and moderate inducer that is used as a pharmacokinetic enhancer in several antiviral treatment regimens. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 59-64 29427135-2 2018 Ritonavir (RTV) is a potent mechanism-based and reversible CYP3A inhibitor and moderate inducer that is used as a pharmacokinetic enhancer in several antiviral treatment regimens. Ritonavir 11-14 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 59-64 29427135-3 2018 Drug-drug interaction (DDI) between RTV and amlodipine is due to mixed inhibition and induction of CYP3A4, which is challenging to predict without a mechanistic model that accounts for the complexity of both mechanisms occurring simultaneously. Ritonavir 36-39 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 99-105 29661866-8 2018 In human hepatocytes and adipocytes, known target cells of FGF21 action, efavirenz, elvitegravir, and the lopinavir-ritonavir combination exerted inhibitory effects on KLB gene expression. Ritonavir 116-125 klotho beta Homo sapiens 168-171 29661866-11 2018 Moreover, pharmacological inhibitors of either ER or oxidative stress significantly impaired lopinavir-ritonavir-induced regulation of FGF21, but not KLB. Ritonavir 103-112 fibroblast growth factor 21 Homo sapiens 135-140 29634071-2 2018 There is a known pharmacological interaction between ritonavir and those corticosteroids which are metabolised by the CYP3A4 pathway. Ritonavir 53-62 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 118-124 29302721-0 2018 Impact of ritonavir dose and schedule on CYP3A inhibition and venetoclax clinical pharmacokinetics. Ritonavir 10-19 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 41-46 29302721-3 2018 The study objective was to determine the effect of different dosage regimens of ritonavir, a strong CYP3A inhibitor, on the pharmacokinetics of venetoclax in 20 healthy subjects. Ritonavir 80-89 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 100-105 29302721-8 2018 Administration of 50 mg ritonavir daily saturated CYP3A inhibition and completely inhibited the formation of the major venetoclax metabolite M27. Ritonavir 24-33 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 50-55 29302721-9 2018 Time-dependent CYP3A inhibition with daily 50 mg ritonavir was offset by ritonavir CYP3A induction, resulting in a limited net increase in CYP3A inhibition with multiple doses. Ritonavir 49-58 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 15-20 29302721-9 2018 Time-dependent CYP3A inhibition with daily 50 mg ritonavir was offset by ritonavir CYP3A induction, resulting in a limited net increase in CYP3A inhibition with multiple doses. Ritonavir 73-82 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 83-88 29302721-9 2018 Time-dependent CYP3A inhibition with daily 50 mg ritonavir was offset by ritonavir CYP3A induction, resulting in a limited net increase in CYP3A inhibition with multiple doses. Ritonavir 73-82 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 83-88 29451681-0 2018 Verification of a physiologically based pharmacokinetic model of ritonavir to estimate drug-drug interaction potential of CYP3A4 substrates. Ritonavir 65-74 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 122-128 29451681-2 2018 In this study, four physiologically based pharmacokinetic (PBPK) models of ritonavir as an in vivo time-dependent inhibitor of CYP3A4 were created and verified for oral doses of 20, 50, 100 and 200 mg using the fraction absorbed (Fa ) and oral clearance (CLoral ) values reported in the literature, because transporter and CYP enzyme reaction phenotyping data were not available. Ritonavir 75-84 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 127-133 29451681-3 2018 The models were used subsequently to predict and compare the magnitude of the AUC increase in nine reference DDI studies evaluating the effect of ritonavir at steady-state on midazolam (CYP3A4 substrate) exposure. Ritonavir 146-155 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 186-192 29451681-5 2018 Simulations of the hepatic and gut CYP3A4 abundance after multiple oral dosing of ritonavir indicated that a 3-day treatment with ritonavir 100 mg twice daily is sufficient to reach maximal CYP3A4 inhibition and subsequent systemic exposure increase of a CYP3A4 substrate, resulting in the reliable estimation of fm,CYP3A4 . Ritonavir 130-139 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 35-41 29451681-5 2018 Simulations of the hepatic and gut CYP3A4 abundance after multiple oral dosing of ritonavir indicated that a 3-day treatment with ritonavir 100 mg twice daily is sufficient to reach maximal CYP3A4 inhibition and subsequent systemic exposure increase of a CYP3A4 substrate, resulting in the reliable estimation of fm,CYP3A4 . Ritonavir 130-139 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 190-196 29451681-5 2018 Simulations of the hepatic and gut CYP3A4 abundance after multiple oral dosing of ritonavir indicated that a 3-day treatment with ritonavir 100 mg twice daily is sufficient to reach maximal CYP3A4 inhibition and subsequent systemic exposure increase of a CYP3A4 substrate, resulting in the reliable estimation of fm,CYP3A4 . Ritonavir 130-139 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 190-196 29451681-5 2018 Simulations of the hepatic and gut CYP3A4 abundance after multiple oral dosing of ritonavir indicated that a 3-day treatment with ritonavir 100 mg twice daily is sufficient to reach maximal CYP3A4 inhibition and subsequent systemic exposure increase of a CYP3A4 substrate, resulting in the reliable estimation of fm,CYP3A4 . Ritonavir 130-139 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 190-196 29280108-0 2018 CSF inflammatory markers and neurocognitive function after addition of maraviroc to monotherapy darunavir/ritonavir in stable HIV patients: the CINAMMON study. Ritonavir 106-115 colony stimulating factor 2 Homo sapiens 0-3 29434718-5 2018 Consistent with previous studies, treatment with lopinavir/ritonavir for 2 weeks decreased body weight, adipose tissue mass, levels of plasma adiponectin and leptin, and increased plasma levels of triglycerides, total cholesterol and insulin. Ritonavir 59-68 adiponectin, C1Q and collagen domain containing Mus musculus 142-153 29093019-0 2018 The X-Ray Crystal Structure of the Human Mono-Oxygenase Cytochrome P450 3A5-Ritonavir Complex Reveals Active Site Differences between P450s 3A4 and 3A5. Ritonavir 76-85 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 56-75 29117017-9 2018 CONCLUSION: Among patients who initiated atazanavir/ritonavir-containing regimens, UGT1A1 slow metabolizer genotype rs887829 T/T was associated with increased bilirubin-related discontinuation of atazanavir in White but not in Black patients, this despite T/T genotype being more frequent in Black patients. Ritonavir 52-61 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 83-89 29216208-7 2017 RESULTS: Overall we showed that patients receiving tenofovir (TDF) with a ritonavir boosted protease inhibitor (rbPI) exhibited a higher risk of CKD compared with patients who received TDF with a non-nucleosidic reverse transcriptase inhibitor (NNRTI). Ritonavir 74-83 sex determining region Y Homo sapiens 62-65 29412384-1 2017 OBJECTIVES: We sought to assess the relationship between stimulated thyroglobulin (sTg) before radioactive iodine therapy (RIT), and the dynamic risk stratification 1 year after treatment, and to establish the utility of the sTg as a predictor of response to therapy in these patients. Ritonavir 123-126 thyroglobulin Homo sapiens 68-81 28960344-1 2017 OBJECTIVES: Ritonavir and cobicistat are strong inhibitors of human cytochrome P450-3A (CYP3A) isoforms, and are used clinically as pharmacokinetic boosting agents for other antiretroviral drugs. Ritonavir 12-21 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 68-86 28960344-1 2017 OBJECTIVES: Ritonavir and cobicistat are strong inhibitors of human cytochrome P450-3A (CYP3A) isoforms, and are used clinically as pharmacokinetic boosting agents for other antiretroviral drugs. Ritonavir 12-21 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 88-93 28960344-6 2017 KEY FINDINGS: Ritonavir and cobicistat both were strong inhibitors of CYP3A4, with IC50 values of 0.014 and 0.032 mum, respectively. Ritonavir 14-23 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 70-76 28960344-10 2017 CONCLUSIONS: Consistent with previous reports, both ritonavir and cobicistat were highly potent inhibitors of CYP3A. Ritonavir 52-61 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 110-115 29135577-4 2018 RESULTS: Among women conceiving on ART, lopinavir/RTV was associated with increased PTD risk in those with CD4 cell count 350 cells/mul or less [odds ratio 1.99 (1.02, 3.85)] and with CD4 cell count more than 350 cells/mul [odds ratio 1.61 (1.07, 2.43)] vs. women on nonnucleoside reverse transcriptase inhibitors-based (mainly efavirenz and nevirapine) regimens in the same CD4 subgroup. Ritonavir 50-53 CD4 molecule Homo sapiens 107-110 29135577-4 2018 RESULTS: Among women conceiving on ART, lopinavir/RTV was associated with increased PTD risk in those with CD4 cell count 350 cells/mul or less [odds ratio 1.99 (1.02, 3.85)] and with CD4 cell count more than 350 cells/mul [odds ratio 1.61 (1.07, 2.43)] vs. women on nonnucleoside reverse transcriptase inhibitors-based (mainly efavirenz and nevirapine) regimens in the same CD4 subgroup. Ritonavir 50-53 CD4 molecule Homo sapiens 184-187 29135577-4 2018 RESULTS: Among women conceiving on ART, lopinavir/RTV was associated with increased PTD risk in those with CD4 cell count 350 cells/mul or less [odds ratio 1.99 (1.02, 3.85)] and with CD4 cell count more than 350 cells/mul [odds ratio 1.61 (1.07, 2.43)] vs. women on nonnucleoside reverse transcriptase inhibitors-based (mainly efavirenz and nevirapine) regimens in the same CD4 subgroup. Ritonavir 50-53 CD4 molecule Homo sapiens 184-187 29232137-0 2018 Inhibition of Human CYP3A4 by Rationally Designed Ritonavir-Like Compounds: Impact and Interplay of the Side Group Functionalities. Ritonavir 50-59 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 20-26 29232137-1 2018 Structure-function relationships of nine rationally designed ritonavir-like compounds were investigated to better understand the ligand binding and inhibitory mechanism in human drug-metabolizing cytochrome P450 3A4 (CYP3A4). Ritonavir 61-70 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 196-215 29232137-1 2018 Structure-function relationships of nine rationally designed ritonavir-like compounds were investigated to better understand the ligand binding and inhibitory mechanism in human drug-metabolizing cytochrome P450 3A4 (CYP3A4). Ritonavir 61-70 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 217-223 28901962-0 2017 Ability of polymer-bound P-glycoprotein inhibitor ritonavir to overcome multidrug resistance in various resistant neuroblastoma cell lines. Ritonavir 50-59 ATP binding cassette subfamily B member 1 Homo sapiens 25-39 28627229-8 2017 Ritonavir induces CYP1A2, 2B6, 2C9, 2C19, and uridine 5"-diphospho-glucuronosyltransferase, whereas cobicistat does not. Ritonavir 0-9 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 18-24 28901962-2 2017 Here, we present the effect of the N-(2-hydroxypropyl) methacrylamide-based polymer conjugate with P-gp inhibitor ritonavir (RIT) on the increase of free doxorubicin (DOX) and polymer-bound DOX cytotoxicity in the human neuroblastoma 4 cell line and its resistant clones to different cytostatics. Ritonavir 114-123 ATP binding cassette subfamily B member 1 Homo sapiens 99-103 28901962-2 2017 Here, we present the effect of the N-(2-hydroxypropyl) methacrylamide-based polymer conjugate with P-gp inhibitor ritonavir (RIT) on the increase of free doxorubicin (DOX) and polymer-bound DOX cytotoxicity in the human neuroblastoma 4 cell line and its resistant clones to different cytostatics. Ritonavir 125-128 ATP binding cassette subfamily B member 1 Homo sapiens 99-103 29108449-2 2017 More rarely, it has been described in HIV-positive patients on ritonavir (RTV) while using the inhaled corticosteroid fluticasone, which is metabolized through the cytochrome P450 3A4 (CYP3A4) enzyme system. Ritonavir 63-72 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 164-183 29108449-2 2017 More rarely, it has been described in HIV-positive patients on ritonavir (RTV) while using the inhaled corticosteroid fluticasone, which is metabolized through the cytochrome P450 3A4 (CYP3A4) enzyme system. Ritonavir 63-72 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 185-191 29108449-2 2017 More rarely, it has been described in HIV-positive patients on ritonavir (RTV) while using the inhaled corticosteroid fluticasone, which is metabolized through the cytochrome P450 3A4 (CYP3A4) enzyme system. Ritonavir 74-77 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 164-183 29108449-2 2017 More rarely, it has been described in HIV-positive patients on ritonavir (RTV) while using the inhaled corticosteroid fluticasone, which is metabolized through the cytochrome P450 3A4 (CYP3A4) enzyme system. Ritonavir 74-77 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 185-191 29108449-3 2017 In the presence of RTV, a known CYP3A4 enzyme inhibitor, the interaction can result in impaired metabolism and systemic accumulation of inhaled fluticasone resulting in iatrogenic CS. Ritonavir 19-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 32-38 29118985-13 2017 Learning points: Drug-drug interaction between ritonavir and triamcinolone can cause Cushing syndrome.Although triamcinolone has a half-life of 3 h, an intra-articular injection may be systematically absorbed for 3 weeks after injection, and adrenal suppression may last as long as 30 days.Co-administration of ritonavir and corticosteroids may result in an increase of plasma levels of corticosteroids levels, as they are both eliminated by CYP3A metabolism, and this interaction has the potential to prolong the half-life of triamcinolone several fold.No specific guidelines are available for the management of iatrogenic Cushing syndrome secondary to ritonavir and corticosteroids.One treatment option includes replacing ritonavir with a non-protease inhibitor-based regimen.Initiating hydrocortisone replacement therapy to prevent an adrenal crisis is also an alternate option. Ritonavir 47-56 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 442-447 29088262-7 2017 Mice with targeted deletion of TGF-beta1 in megakaryocytes/platelets (PF4CreTgfb1flox/flox) were partially protected from ritonavir-induced cardiac dysfunction and fibrosis. Ritonavir 122-131 transforming growth factor, beta 1 Mus musculus 31-40 29088262-7 2017 Mice with targeted deletion of TGF-beta1 in megakaryocytes/platelets (PF4CreTgfb1flox/flox) were partially protected from ritonavir-induced cardiac dysfunction and fibrosis. Ritonavir 122-131 transforming growth factor, beta 1 Mus musculus 70-81 29088262-12 2017 These results suggest that platelet-derived TGF-beta1 contributes to ritonavir-associated cardiac dysfunction and fibrosis, extending the relevance of our findings to other antiretrovirals that also activate platelets. Ritonavir 69-78 transforming growth factor, beta 1 Mus musculus 44-53 28951049-8 2017 To prove the applicability of the procedure, the effect of amprenavir, indinavir, nelfinavir, ritonavir, and saquinavir on the hBCRP ATPase activity was tested. Ritonavir 94-103 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 127-132 28951049-9 2017 Nelfinavir, ritonavir, and saquinavir were identified as hBCRP ATPase inhibitors and none of the five HIV protease inhibitors turned out to be an hBCRP substrate. Ritonavir 12-21 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 57-62 27590127-8 2017 Finally, we compared the phospholipids with Tween 80 and the competitive P-gp inhibitor verapamil in an in vivo study, testing their effects on the oral bioavailability of the P-gp substrate drug ritonavir. Ritonavir 197-206 phosphoglycolate phosphatase Homo sapiens 177-181 28848011-0 2017 Differential Influence of the Antiretroviral Pharmacokinetic Enhancers Ritonavir and Cobicistat on Intestinal P-Glycoprotein Transport and the Pharmacokinetic/Pharmacodynamic Disposition of Dabigatran. Ritonavir 71-80 ATP binding cassette subfamily B member 1 Homo sapiens 110-124 28837922-6 2017 GLUT4 inhibition by knockdown or treatment with the FDA-approved HIV protease inhibitor ritonavir leads to cytostatic and/or cytotoxic and chemosensitizing effects in tumor cells both in vitro and in vivo. Ritonavir 88-97 solute carrier family 2 member 4 Homo sapiens 0-5 28483778-4 2017 When perpetrator interactions were assessed, ritonavir was responsible for the strong increase in exposure of sensitive CYP3A substrates, whereas paritaprevir (an OATP1B1/1B3 inhibitor) greatly increased the exposure of sensitive OATP1B1/1B3 substrates. Ritonavir 45-54 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 120-125 28978797-6 2017 METHODS: We conducted a phase 1B study of 12 HIV-positive individuals on a ritonavir-containing regimen (HIV viral load less than 40 copies/ml and CD4 > 400 cells/mul). Ritonavir 75-84 CD4 molecule Homo sapiens 147-150 28688088-2 2017 However, due to the acute effect of I-131 on thyrocytes, Tg measured after radioiodine therapy (RIT) would not accurately reflect the thyroid tissue burden. Ritonavir 96-99 thyroglobulin Homo sapiens 57-59 28683645-1 2017 OBJECTIVES: An observational, prospective, cohort study was performed to compare efficacy and safety of a switch from ritonavir-boosted protease inhibitor (PI/r) to nevirapine or raltegravir with that of rosuvastatin addition to current antiretroviral therapy in HIV-infected patients with hyperlipidaemia. Ritonavir 118-127 serpin family A member 13, pseudogene Homo sapiens 136-154 28683645-1 2017 OBJECTIVES: An observational, prospective, cohort study was performed to compare efficacy and safety of a switch from ritonavir-boosted protease inhibitor (PI/r) to nevirapine or raltegravir with that of rosuvastatin addition to current antiretroviral therapy in HIV-infected patients with hyperlipidaemia. Ritonavir 118-127 serpin family A member 13, pseudogene Homo sapiens 156-160 28718515-2 2017 Although primarily metabolized by CYP2B6 and -3A, efavirenz (EFV) and lopinavir/ritonavir (LPV/r) are substrates of P-glycoprotein and the solute carrier organic (SLCO) anion transporter, respectively. Ritonavir 80-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-40 28550055-12 2017 However, ritonavir showed the highest DDI index (1.9) for OATP4C1, suggesting that it may strongly influence this transporter and thus cause drug interactions seen in clinical settings. Ritonavir 9-18 solute carrier organic anion transporter family member 4C1 Homo sapiens 58-65 29062559-1 2017 Ritonavir is a powerful inhibitor of the cytochrome P450 3A4 (CYP3A4) isoenzyme. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 41-60 29062559-1 2017 Ritonavir is a powerful inhibitor of the cytochrome P450 3A4 (CYP3A4) isoenzyme. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 62-68 29062559-3 2017 However, when co-administered with other drugs that are metabolised via the CYP3A4 pathway, ritonavir can potentially cause serious drug-drug interactions. Ritonavir 92-101 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 76-82 28403692-9 2017 Our hypothesis is that the hypokalemia could be a result of a tenofovir-mediated tubular damage triggered by the increased vinblastine serum levels secondary to a CYP3A4 inhibition by ritonavir. Ritonavir 184-193 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 163-169 28483778-6 2017 Paritaprevir, ritonavir, and dasabuvir are BCRP inhibitors. Ritonavir 14-23 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 43-47 28483778-8 2017 Plasma exposures of the 3D regimen were reduced by strong CYP3A inducers (paritaprevir and ritonavir; major CYP3A substrates) but were not affected by strong CYP3A4 inhibitors, since ritonavir (a CYP3A inhibitor) is already present in the regimen. Ritonavir 91-100 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 58-63 28443391-4 2017 Bioequivalence between cobicistat (COBI) and ritonavir (RTV) as a pharmacoenhancer of ATV was established. Ritonavir 56-59 nibrin Homo sapiens 86-89 28458002-6 2017 It was invigorating to identify that chlorohexidine, paromomycin and deferoxamine could inhibit the wild-type ABL1, while chlorohexidine and ritonavir could inhibit the T315I mutant ABL1. Ritonavir 141-150 ABL proto-oncogene 1, non-receptor tyrosine kinase Homo sapiens 182-186 28060049-2 2017 We describe a case of symptomatic Cushing"s syndrome in an adolescent male with sight-threatening vernal keratoconjunctivitis on antiretroviral therapy for HIV-1 infection that included ritonavir, a potent cytochrome p450 CYP3A4 inhibitor. Ritonavir 186-195 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 222-228 27798431-0 2017 Hemeoxygenase-1 as a Novel Driver in Ritonavir-Induced Insulin Resistance in HIV-1-Infected Patients. Ritonavir 37-46 insulin Homo sapiens 55-62 27798431-6 2017 RESULTS: We show that, in contrast to RAL, ritonavir treatment significantly increases mRNA expression levels of HO-1, IL-8, TNFalpha, CCL5, and MCP-1 in vitro in a dose-dependent manner. Ritonavir 43-52 C-X-C motif chemokine ligand 8 Homo sapiens 119-123 27798431-6 2017 RESULTS: We show that, in contrast to RAL, ritonavir treatment significantly increases mRNA expression levels of HO-1, IL-8, TNFalpha, CCL5, and MCP-1 in vitro in a dose-dependent manner. Ritonavir 43-52 tumor necrosis factor Homo sapiens 125-133 27798431-6 2017 RESULTS: We show that, in contrast to RAL, ritonavir treatment significantly increases mRNA expression levels of HO-1, IL-8, TNFalpha, CCL5, and MCP-1 in vitro in a dose-dependent manner. Ritonavir 43-52 C-C motif chemokine ligand 5 Homo sapiens 135-139 27798431-6 2017 RESULTS: We show that, in contrast to RAL, ritonavir treatment significantly increases mRNA expression levels of HO-1, IL-8, TNFalpha, CCL5, and MCP-1 in vitro in a dose-dependent manner. Ritonavir 43-52 C-C motif chemokine ligand 2 Homo sapiens 145-150 28521396-0 2017 Inhibition of MMP-9 expression by ritonavir or saquinavir is associated with inactivation of the AKT/Fra-1 pathway in cervical intraepithelial neoplasia cells. Ritonavir 34-43 matrix metallopeptidase 9 Homo sapiens 14-19 28521396-0 2017 Inhibition of MMP-9 expression by ritonavir or saquinavir is associated with inactivation of the AKT/Fra-1 pathway in cervical intraepithelial neoplasia cells. Ritonavir 34-43 AKT serine/threonine kinase 1 Homo sapiens 97-100 28579812-13 2017 Detailed analysis of the costs for GT 1 showed the treatment based on ritonavir boosted paritaprevir/ombitasvir + dasabuvir+-RBV with an average cost of $24,978 (RBV+) and $25,448 (RBV-) per patient was the most cost-effective. Ritonavir 70-79 beta-1,4-galactosyltransferase 1 Homo sapiens 35-39 28341759-3 2017 Strong CYP3A4 inhibition by ritonavir may contribute to the observed sorafenib toxicity.Alternate antiretroviral agents without predicted interactions are preferred for co-administration in patients with HIV and cancers for which sorafenib is indicated. Ritonavir 28-37 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 7-13 28341759-5 2017 We evaluated drug-drug interactions between sorafenib and ritonavir, an HIV medication with strong CYP3A4 inhibitory activity. Ritonavir 58-67 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 99-105 28341759-16 2017 Strong CYP3A4 inhibitors may contribute to sorafenib toxicity, and ritonavir has previously been shown to be a CYP3A4 inhibitor. Ritonavir 67-76 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 111-117 27999204-5 2017 Here, we show that this favorable interaction occurs in cell culture, is dependent on the dose and duration of RG7787 exposure, requires the catalytically active PE, and still occurs with RIT targeting a non-MSLN surface antigen. Ritonavir 188-191 mesothelin Mus musculus 208-212 27165046-2 2017 The objective of this analysis was to evaluate the effect of renal function as determined by creatinine clearance (CrCL) on the pharmacokinetics of the DAAs, ritonavir, and ribavirin in HCV genotype 1-infected patients with or without cirrhosis. Ritonavir 158-167 CRCL Homo sapiens 115-119 28979315-4 2017 Since lymphocytes and lymphoma cells are highly radiosensitive, low grade NHL that has relapsed or refractory to standard therapy is treated by RIT in which a beta-emitting radionuclide coupled to anti-CD20 antibody. Ritonavir 144-147 keratin 20 Homo sapiens 202-206 27909750-8 2017 The data suggest that ritonavir, in addition to containing viral replication, could inhibit the expression of virulence factors in opportunistic yeast, as proteases and Hsp90. Ritonavir 22-31 Hsp90 family chaperone HSP82 Saccharomyces cerevisiae S288C 169-174 26976380-2 2017 We evaluated the p24 levels in patients on triple therapy and after switching to ritonavir-boosted protease inhibitor monotherapy (mtPI/rtv), as well as the relationships with virological and immunological evolution. Ritonavir 81-90 transmembrane p24 trafficking protein 2 Homo sapiens 17-20 28134057-3 2017 OBJECTIVE: To compare the effect of raltegravir + ritonavir boosted lopinavir (RAL + LPV/r) to efavirenz/tenofovir disoproxil fumarate/emtricitabine (EFV/TDF/FTC) on HIV kinetics and T cell dynamics. Ritonavir 50-59 RAS like proto-oncogene A Homo sapiens 79-82 28458904-6 2017 Pharmacogenetic analysis revealed that the pediatric patient carried the CYP3A4 *1B/*1G and CYP3A5 *3/*3 genotype (associated with a partial and complete loss of enzyme activity, respectively) which is associated with the development of iatrogenic Cushing"s syndrome in patients treated for HIV due to the strong inhibition of CYP3 enzymes by Ritonavir. Ritonavir 343-352 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 73-79 28458904-6 2017 Pharmacogenetic analysis revealed that the pediatric patient carried the CYP3A4 *1B/*1G and CYP3A5 *3/*3 genotype (associated with a partial and complete loss of enzyme activity, respectively) which is associated with the development of iatrogenic Cushing"s syndrome in patients treated for HIV due to the strong inhibition of CYP3 enzymes by Ritonavir. Ritonavir 343-352 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 92-98 28458904-6 2017 Pharmacogenetic analysis revealed that the pediatric patient carried the CYP3A4 *1B/*1G and CYP3A5 *3/*3 genotype (associated with a partial and complete loss of enzyme activity, respectively) which is associated with the development of iatrogenic Cushing"s syndrome in patients treated for HIV due to the strong inhibition of CYP3 enzymes by Ritonavir. Ritonavir 343-352 peptidylprolyl isomerase F Homo sapiens 73-77 27144920-1 2017 BACKGROUND: Stimulated thyroglobulin (STg) levels in patients with differentiated thyroid carcinomas (DTCs) after total thyroidectomy (TT) and before radioactive iodine (131I) ablation/therapy (RIT) are predictive of therapeutic success but can be influenced by the thyroid-stimulating hormone (TSH) level. Ritonavir 194-197 thyroglobulin Homo sapiens 23-36 28280605-9 2017 The most frequent activated signaling pathways were: HER2, fibroblast growth factor receptor (FGFR), p38 through BRAF-MEK cascade via RIT and RIN, ARMS-mediated activation of MAPK cascade, and VEGFR2. Ritonavir 134-137 mitogen-activated protein kinase 14 Homo sapiens 101-104 28038962-2 2017 The antiretrovirals ritonavir-boosted lopinavir (LPV/r) and nevirapine inhibit and induce CYP3A4, respectively. Ritonavir 20-29 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 90-96 27432796-0 2017 CYP2C19 Genotype-Dependent Pharmacokinetic Drug Interaction Between Voriconazole and Ritonavir-Boosted Atazanavir in Healthy Subjects. Ritonavir 85-94 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 0-7 27432796-2 2017 Genetic polymorphism of CYP2C19 not only plays a prominent role in its disposition but may also influence potential drug interactions with CYP450 modulators such as ritonavir. Ritonavir 165-174 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 24-31 28280605-9 2017 The most frequent activated signaling pathways were: HER2, fibroblast growth factor receptor (FGFR), p38 through BRAF-MEK cascade via RIT and RIN, ARMS-mediated activation of MAPK cascade, and VEGFR2. Ritonavir 134-137 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 113-117 28280605-9 2017 The most frequent activated signaling pathways were: HER2, fibroblast growth factor receptor (FGFR), p38 through BRAF-MEK cascade via RIT and RIN, ARMS-mediated activation of MAPK cascade, and VEGFR2. Ritonavir 134-137 mitogen-activated protein kinase kinase 7 Homo sapiens 118-121 27924729-6 2017 trans10,cis12 + ritonavir down-regulated Pparg (-1.55) and Adipoq (-2.95), as well as differentiation (Fcor (-4.78-fold), Arl4a (-4.84), Itga6 (-2.45), Id4 (-2.01)) and triglyceride storage genes (Mrap (- 8.25), Scd1 (-4.34), Lipin1 (-2.52)). Ritonavir 16-25 adiponectin, C1Q and collagen domain containing Homo sapiens 59-65 27404500-9 2017 Efavirenz, stavudine and ritonavir significantly down-regulated the bioactivating CYP2R1 and up-regulated the catabolic CYP24A1. Ritonavir 25-34 cytochrome P450 family 2 subfamily R member 1 Homo sapiens 82-88 27404500-9 2017 Efavirenz, stavudine and ritonavir significantly down-regulated the bioactivating CYP2R1 and up-regulated the catabolic CYP24A1. Ritonavir 25-34 cytochrome P450 family 24 subfamily A member 1 Homo sapiens 120-127 27774892-9 2017 CONCLUSION: These findings indicate that both ATV+COBI and ATV+RTV, each with FTC/TDF, are effective and well-tolerated treatment options across a wide demographic range of HIV-infected patients. Ritonavir 63-66 sex determining region Y Homo sapiens 82-85 27924729-6 2017 trans10,cis12 + ritonavir down-regulated Pparg (-1.55) and Adipoq (-2.95), as well as differentiation (Fcor (-4.78-fold), Arl4a (-4.84), Itga6 (-2.45), Id4 (-2.01)) and triglyceride storage genes (Mrap (- 8.25), Scd1 (-4.34), Lipin1 (-2.52)). Ritonavir 16-25 ADP ribosylation factor like GTPase 4A Homo sapiens 122-127 27924729-6 2017 trans10,cis12 + ritonavir down-regulated Pparg (-1.55) and Adipoq (-2.95), as well as differentiation (Fcor (-4.78-fold), Arl4a (-4.84), Itga6 (-2.45), Id4 (-2.01)) and triglyceride storage genes (Mrap (- 8.25), Scd1 (-4.34), Lipin1 (-2.52)). Ritonavir 16-25 integrin subunit alpha 6 Homo sapiens 137-142 27924729-6 2017 trans10,cis12 + ritonavir down-regulated Pparg (-1.55) and Adipoq (-2.95), as well as differentiation (Fcor (-4.78-fold), Arl4a (-4.84), Itga6 (-2.45), Id4 (-2.01)) and triglyceride storage genes (Mrap (- 8.25), Scd1 (-4.34), Lipin1 (-2.52)). Ritonavir 16-25 inhibitor of DNA binding 4, HLH protein Homo sapiens 152-155 27924729-6 2017 trans10,cis12 + ritonavir down-regulated Pparg (-1.55) and Adipoq (-2.95), as well as differentiation (Fcor (-4.78-fold), Arl4a (-4.84), Itga6 (-2.45), Id4 (-2.01)) and triglyceride storage genes (Mrap (- 8.25), Scd1 (-4.34), Lipin1 (-2.52)). Ritonavir 16-25 melanocortin 2 receptor accessory protein Homo sapiens 197-201 27924729-6 2017 trans10,cis12 + ritonavir down-regulated Pparg (-1.55) and Adipoq (-2.95), as well as differentiation (Fcor (-4.78-fold), Arl4a (-4.84), Itga6 (-2.45), Id4 (-2.01)) and triglyceride storage genes (Mrap (- 8.25), Scd1 (-4.34), Lipin1 (-2.52)). Ritonavir 16-25 stearoyl-CoA desaturase Homo sapiens 212-216 27924729-6 2017 trans10,cis12 + ritonavir down-regulated Pparg (-1.55) and Adipoq (-2.95), as well as differentiation (Fcor (-4.78-fold), Arl4a (-4.84), Itga6 (-2.45), Id4 (-2.01)) and triglyceride storage genes (Mrap (- 8.25), Scd1 (-4.34), Lipin1 (-2.52)). Ritonavir 16-25 lipin 1 Homo sapiens 226-232 27924729-8 2017 cis9,trans11 + ritonavir increased PPAR-gamma nuclear binding to its gene response element (P=0.038). Ritonavir 15-24 peroxisome proliferator activated receptor gamma Homo sapiens 35-45 27378623-9 2016 PON1 activity toward paraoxon was decreased in groups receiving PIs (control: 149 +- 5 U/ml; PIs-treated: RTV at doses 10 mg/kg 133 +- 9.5 U/ml, RTV at doses 50 mg/kg 134 +- 10.8 U/ml, SQV at doses 10 mg/kg 131 +- 9.2 U/ml, ATV at doses 10 mg/kg 132 +- 11.8 U/ml, ATV at doses 100 mg/kg 108 +- 7.8 U/ml). Ritonavir 106-109 paraoxonase 1 Rattus norvegicus 0-4 27378623-9 2016 PON1 activity toward paraoxon was decreased in groups receiving PIs (control: 149 +- 5 U/ml; PIs-treated: RTV at doses 10 mg/kg 133 +- 9.5 U/ml, RTV at doses 50 mg/kg 134 +- 10.8 U/ml, SQV at doses 10 mg/kg 131 +- 9.2 U/ml, ATV at doses 10 mg/kg 132 +- 11.8 U/ml, ATV at doses 100 mg/kg 108 +- 7.8 U/ml). Ritonavir 146-149 paraoxonase 1 Rattus norvegicus 0-4 27098343-8 2016 Ritonavir and dexamethasone both induced transcription of CYP27B1, the enzyme responsible for the formation of 1,25D3 . Ritonavir 0-9 cytochrome P450 family 27 subfamily B member 1 Homo sapiens 58-65 27889708-3 2016 In cultured ECs, miR-34a expression was significantly increased by HIV-Tat protein and by the antiretroviral agents, lopinavir/ritonavir. Ritonavir 127-136 microRNA 34a Mus musculus 17-24 27697800-1 2016 Ritonavir and cobicistat, used as pharmacokinetic enhancers in combination with some antiretrovirals (ARVs) for the treatment of HIV, are potent inhibitors of the CYP3A4 isoenzyme. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 163-169 27230021-10 2016 Mediation analysis suggested that CE (38%) and RTV (44%) substantially mediated the association between inattention and the T-allele of SNP rs3785157 in the norepinephrine transporter gene (SLC6A2) and the T-allele of SNP rs7984966 in HTR2A, respectively. Ritonavir 47-50 solute carrier family 6 member 2 Homo sapiens 157-183 27230021-10 2016 Mediation analysis suggested that CE (38%) and RTV (44%) substantially mediated the association between inattention and the T-allele of SNP rs3785157 in the norepinephrine transporter gene (SLC6A2) and the T-allele of SNP rs7984966 in HTR2A, respectively. Ritonavir 47-50 solute carrier family 6 member 2 Homo sapiens 190-196 27230021-10 2016 Mediation analysis suggested that CE (38%) and RTV (44%) substantially mediated the association between inattention and the T-allele of SNP rs3785157 in the norepinephrine transporter gene (SLC6A2) and the T-allele of SNP rs7984966 in HTR2A, respectively. Ritonavir 47-50 5-hydroxytryptamine receptor 2A Homo sapiens 235-240 27697800-2 2016 Most glucocorticoids are metabolised via the CYP3A4 pathway and iatrogenic Cushing"s syndrome (ICS), with possible secondary adrenal insufficiency (SAI), is a recognised complication following co-administration with ritonavir or cobicistat. Ritonavir 216-225 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 45-51 26626330-10 2016 Both compounds induced several drug metabolizing genes (including CYP2B6, CYP3A4 and UGT1A1), mediated by CAR activation in Nevirapine and PXR in Ritonavir. Ritonavir 146-155 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-72 27458223-2 2016 Three studies were conducted to evaluate the potential drug-drug interactions between delamanid and antiretroviral drugs, including ritonavir, a strong inhibitor of CYP3A4, and selected anti-TB drugs, including rifampin, a strong inducer of cytochrome P450 (CYP) isozymes. Ritonavir 132-141 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 165-171 26626330-10 2016 Both compounds induced several drug metabolizing genes (including CYP2B6, CYP3A4 and UGT1A1), mediated by CAR activation in Nevirapine and PXR in Ritonavir. Ritonavir 146-155 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 74-80 26626330-10 2016 Both compounds induced several drug metabolizing genes (including CYP2B6, CYP3A4 and UGT1A1), mediated by CAR activation in Nevirapine and PXR in Ritonavir. Ritonavir 146-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-91 26626330-10 2016 Both compounds induced several drug metabolizing genes (including CYP2B6, CYP3A4 and UGT1A1), mediated by CAR activation in Nevirapine and PXR in Ritonavir. Ritonavir 146-155 nuclear receptor subfamily 1 group I member 2 Homo sapiens 139-142 26945713-3 2016 Cobicistat and ritonavir are equally strong inhibitors of cytochrome P450 (CYP) 3A4 and consequently were shown to be equivalent pharmacokinetic enhancers for elvitegravir and for the PIs atazanavir and darunavir. Ritonavir 15-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-73 26640142-10 2016 Inhibition of glucose metabolism with the GLUT4 inhibitor ritonavir elicits variable cytotoxicity in MM that is marginally enhanced with venetoclax treatment, however, targeting glutamine metabolism with 6-diazo-5-oxo-l-norleucine uniformly sensitized MM cell lines and relapse/refractory patient samples to venetoclax. Ritonavir 58-67 solute carrier family 2 member 4 Homo sapiens 42-47 26945713-3 2016 Cobicistat and ritonavir are equally strong inhibitors of cytochrome P450 (CYP) 3A4 and consequently were shown to be equivalent pharmacokinetic enhancers for elvitegravir and for the PIs atazanavir and darunavir. Ritonavir 15-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-78 27348592-0 2016 Response to First-Line Ritonavir-Boosted Protease Inhibitors (PI/r)-Based Regimens in HIV Positive Patients Presenting to Care with Low CD4 Counts: Data from the Icona Foundation Cohort. Ritonavir 23-32 CD4 molecule Homo sapiens 136-139 27264208-6 2016 As a result, a 48-hr exposure of human erythrocytes to ritonavir significantly increased the percentage of Annexin-V-binding cells (>=5 mug/ml), significantly decreased forward scatter (>=5 mug/ml), significantly increased Fluo3 fluorescence (20 mug/ml), slightly, but significantly increased DCFDA fluorescence (20 mug/ml) and slightly, but significantly increased ceramide abundance (20 mug/ml). Ritonavir 55-64 annexin A5 Homo sapiens 107-116 27264208-7 2016 The effect of ritonavir on Annexin-V binding was significantly blunted, but not fully abolished by the removal of extracellular Ca(2+) . Ritonavir 14-23 annexin A5 Homo sapiens 27-36 27126611-2 2016 Herein we describe a spectroscopic investigation of the interaction of CYP3A4 with N-methylritonavir, an analog of ritonavir, widely used as a pharmacoenhancer. Ritonavir 91-100 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 71-77 27167198-4 2016 Our goal is to produce a new RIT that targets mesothelin and is non-immunogenic by combining mutations that decrease B- and T-cell epitopes. Ritonavir 29-32 mesothelin Homo sapiens 46-56 26709605-1 2016 OBJECTIVES: The aim of this analysis was to review the evidence and update a meta-analysis evaluating the efficacy and safety results from randomized controlled trials of ritonavir-boosted protease inhibitor (PI/r) monotherapy. Ritonavir 171-180 serpin family A member 13, pseudogene Homo sapiens 189-207 26371436-3 2016 On the basis of our investigations of analogs of ritonavir, a potent CYP3A4 inactivator and pharmacoenhancer, we have built a pharmacophore model for a CYP3A4-specific inhibitor. Ritonavir 49-58 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 69-75 26371436-3 2016 On the basis of our investigations of analogs of ritonavir, a potent CYP3A4 inactivator and pharmacoenhancer, we have built a pharmacophore model for a CYP3A4-specific inhibitor. Ritonavir 49-58 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 152-158 26968795-4 2016 We found that three distinct PIs, including ritonavir, atazanavir, and lopinavir, stimulated the expression of HO-1 protein and mRNA. Ritonavir 44-53 heme oxygenase 1 Homo sapiens 111-115 26709605-1 2016 OBJECTIVES: The aim of this analysis was to review the evidence and update a meta-analysis evaluating the efficacy and safety results from randomized controlled trials of ritonavir-boosted protease inhibitor (PI/r) monotherapy. Ritonavir 171-180 serpin family A member 13, pseudogene Homo sapiens 209-213 26939705-5 2016 H89 increased the rate of ADP-ribosylation of eukaryotic elongation factor 2, enhancing the arrest of protein synthesis and the reduction of MCL1 in synergy with the RIT. Ritonavir 166-169 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 141-145 27019957-4 2016 One approach to overcome NVS123 developability issues was to increase plasma exposure by coadministrating it with an inhibitor of CYP3A4 such as ritonavir. Ritonavir 145-154 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 130-136 27029846-8 2016 Concomitant administration of non-ritonavir-boosted atazanavir decreased elvitegravir clearance by 35%, likely due to UDP-glucuronosyl transferase (UGT) 1A1 inhibition. Ritonavir 34-43 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 118-156 27196356-1 2016 Cobicistat and ritonavir are structurally distinct compounds that both potently inhibit cytochrome P450 (CYP) 3A, the metabolizing enzyme primarily responsible for the elimination of several antiretroviral medications, and, as such, are pharmacokinetic boosters for antiretroviral agents that require longer dosing intervals. Ritonavir 15-24 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 88-112 26833162-2 2016 Here we investigated the phenotypic consequences of amino acid changes in Gag and protease on lopinavir (LPV) and ritonavir (RTV) susceptibility among pediatric patients failing PI therapy. Ritonavir 125-128 Pr55(Gag) Human immunodeficiency virus 1 74-77 26369773-5 2016 The Michaelis-Menten constant (K m) and the maximum rate of metabolite formation (V max) for cytochrome P450 3A4-mediated darunavir biotransformation and inhibition by ritonavir were determined experimentally, while the contributions of hepatocyte influx and efflux transporters were assessed by sensitivity analysis. Ritonavir 168-177 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 93-112 26719540-3 2016 To enhance the therapeutic efficacy of RITs, we conducted a kinome RNAi sensitization screen, which identified discoidin domain receptor 1 (DDR1), a collagen-activated tyrosine kinase, as a potential target. Ritonavir 39-43 discoidin domain receptor tyrosine kinase 1 Homo sapiens 111-138 26719540-3 2016 To enhance the therapeutic efficacy of RITs, we conducted a kinome RNAi sensitization screen, which identified discoidin domain receptor 1 (DDR1), a collagen-activated tyrosine kinase, as a potential target. Ritonavir 39-43 discoidin domain receptor tyrosine kinase 1 Homo sapiens 140-144 26719540-5 2016 Therefore, we investigated the effects of DDR1 on RIT. Ritonavir 50-53 discoidin domain receptor tyrosine kinase 1 Homo sapiens 42-46 26668209-8 2016 The results indicate that, like ketoconazole, the alternative clinical CYP3A4/5 inhibitors ritonavir, clarithromycin, and itraconazole each have unique transporter inhibition profiles. Ritonavir 91-100 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 71-77 26353895-3 2016 It is susceptible to metabolic drug-drug interactions with drugs that are moderate or strong CYP3A inhibitors (e.g. ritonavir and erythromycin) or CYP3A inducers (e.g. rifampin and efavirenz); coadministration of these drugs may increase or decrease plasma concentrations of simeprevir, respectively, and should be avoided. Ritonavir 116-125 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 93-98 26239522-0 2016 Evidence-based choice of ritonavir as index CYP3A inhibitor in drug-drug interaction studies. Ritonavir 25-34 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 44-49 26741368-9 2016 Our results showed that methamphetamine alters the ritonavir binding to CYP3A4 by decreasing both the deltaAmax (0.0038+-0.0003 vs. 0.0055+-0.0003) and KD (0.043+-0.0001 vs. 0.065+-0.001 nM), while indinavir showed only reduced KD in presence of methamphetamine (0.086+-0.01 vs. 0.174+-0.03 nM). Ritonavir 51-60 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 72-78 26818566-3 2016 Artemether and lumefantrine are metabolised by cytochrome P450 isoenzyme CYP3A4, which lopinavir/ritonavir inhibits, potentially causing clinically important drug-drug interactions. Ritonavir 97-106 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 73-79 26684581-9 2016 Furthermore, uptake of compound 2 was inhibited by two inhibitors of OATP1B1*1a and OATP1B3: rifampicin and ritonavir. Ritonavir 108-117 solute carrier organic anion transporter family member 1B1 Homo sapiens 69-76 26684581-9 2016 Furthermore, uptake of compound 2 was inhibited by two inhibitors of OATP1B1*1a and OATP1B3: rifampicin and ritonavir. Ritonavir 108-117 solute carrier organic anion transporter family member 1B3 Homo sapiens 84-91 26741368-11 2016 Finally, CYP3A4 docking with lopinavir and ritonavir in the absence and presence of methamphetamine showed that methamphetamine alters the docking of ritonavir, which is consistent with the results obtained from spectral binding and metabolism studies. Ritonavir 43-52 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 9-15 26741368-11 2016 Finally, CYP3A4 docking with lopinavir and ritonavir in the absence and presence of methamphetamine showed that methamphetamine alters the docking of ritonavir, which is consistent with the results obtained from spectral binding and metabolism studies. Ritonavir 150-159 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 9-15 26727525-4 2016 METHODS: This study examined the chronic effects of EtOH and ART (darunavir/ritonavir), alone and in combination, on expression/levels of cytochrome P450 enzymes (CYPs), antioxidant enzymes (AOEs), reactive oxygen species (ROS), and cytotoxicity in U937 cells. Ritonavir 76-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 138-153 26872331-0 2016 Effectiveness of Ritonavir-Boosted Protease Inhibitor Monotherapy in Clinical Practice Even with Previous Virological Failures to Protease Inhibitor-Based Regimens. Ritonavir 17-26 serpin family A member 13, pseudogene Homo sapiens 35-53 26872331-0 2016 Effectiveness of Ritonavir-Boosted Protease Inhibitor Monotherapy in Clinical Practice Even with Previous Virological Failures to Protease Inhibitor-Based Regimens. Ritonavir 17-26 serpin family A member 13, pseudogene Homo sapiens 130-148 26872331-1 2016 BACKGROUND AND OBJECTIVE: Significant controversy still exists about ritonavir-boosted protease inhibitor monotherapy (mtPI/rtv) as a simplification strategy that is used up to now to treat patients that have not experienced previous virological failure (VF) while on protease inhibitor (PI) -based regimens. Ritonavir 69-78 serpin family A member 13, pseudogene Homo sapiens 87-105 26872331-1 2016 BACKGROUND AND OBJECTIVE: Significant controversy still exists about ritonavir-boosted protease inhibitor monotherapy (mtPI/rtv) as a simplification strategy that is used up to now to treat patients that have not experienced previous virological failure (VF) while on protease inhibitor (PI) -based regimens. Ritonavir 69-78 serpin family A member 13, pseudogene Homo sapiens 268-286 26452722-10 2016 Rifampin-mediated CYP3A4 induction increased midazolam clearance by 73%, while CYP3A4 inhibition with ritonavir decreased midazolam clearance by 79%. Ritonavir 102-111 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 79-85 26245760-13 2016 PPAR-gamma binding to its nuclear consensus site, adiponectin secretion and triglyceride accumulation were all reduced by ritonavir. Ritonavir 122-131 peroxisome proliferator activated receptor gamma Mus musculus 0-10 26245760-13 2016 PPAR-gamma binding to its nuclear consensus site, adiponectin secretion and triglyceride accumulation were all reduced by ritonavir. Ritonavir 122-131 adiponectin, C1Q and collagen domain containing Mus musculus 50-61 27712589-2 2016 The human immunodeficiency virus protease inhibitor ritonavir, on the other hand, not only increases unfolded proteins by suppressing HSP90 but also acts as a proteasome inhibitor. Ritonavir 52-61 heat shock protein 90 alpha family class A member 1 Homo sapiens 134-139 26443804-5 2015 In this work, we describe the development and characterization of LMB-T20, a highly potent RIT targeted at mesothelin-expressing cancers with low immunogenicity due to removal of its eight T-cell epitopes. Ritonavir 91-94 mesothelin Homo sapiens 107-117 26463060-9 2016 The discrepancy between the methods was also apparent for conducted trials, e.g., lopinavir/ritonavir was predicted to increased BDQ exposure 22% by NCA and 188% by model-based methods. Ritonavir 92-101 CEA cell adhesion molecule 6 Homo sapiens 149-152 26559816-9 2015 Seventy-eight percent (n = 2772) of patients received a prescription for PEP, consisting of Tenofovir/Emtracitabine (TVD) + Lopinavir/Ritonavir (LPV) in 74% of cases, followed by Zidovudine/Lamivudine (CBV) + LPV (10%) and TVD + Raltegravir (RAL) (8%). Ritonavir 134-143 prolyl endopeptidase Homo sapiens 73-76 25923589-7 2015 CONCLUSIONS: Ritonavir produces CYP3A inhibition equivalent to or greater than ketoconazole, and is the best index CYP3A inhibitor alternative to ketoconazole. Ritonavir 13-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 32-37 26348830-0 2015 Augmented Inhibition of CYP3A4 in Human Primary Hepatocytes by Ritonavir Solid Drug Nanoparticles. Ritonavir 63-72 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 24-30 26348830-12 2015 These data provide in vitro proof of concept for improved inhibition of CYP3A4 by ritonavir through formation of solid drug nanoparticles. Ritonavir 82-91 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 72-78 27747723-9 2015 Rivaroxaban, a substrate for cytochrome P450 (CYP) 3A4 and P-glycoprotein (P-gp), is contraindicated in patients concomitantly treated with strong inhibitors of both these systems, e.g. protease inhibitors (PIs) such as ritonavir (based on in vitro data and a pharmacokinetic study in healthy volunteers). Ritonavir 220-229 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 29-54 27747723-9 2015 Rivaroxaban, a substrate for cytochrome P450 (CYP) 3A4 and P-glycoprotein (P-gp), is contraindicated in patients concomitantly treated with strong inhibitors of both these systems, e.g. protease inhibitors (PIs) such as ritonavir (based on in vitro data and a pharmacokinetic study in healthy volunteers). Ritonavir 220-229 ATP binding cassette subfamily B member 1 Homo sapiens 75-79 26330331-12 2015 RESULTS: Administration of erlotinib with CYP3A4 inducers (dexamethasone) and inhibitors (ketoconazole and ritonavir) resulted in significant alterations in erlotinib exposure. Ritonavir 107-116 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 42-48 25923589-7 2015 CONCLUSIONS: Ritonavir produces CYP3A inhibition equivalent to or greater than ketoconazole, and is the best index CYP3A inhibitor alternative to ketoconazole. Ritonavir 13-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 115-120 26322261-1 2015 Ritonavir is a potent inhibitor of the cytochrome P450 enzyme CYP3A4 and is subject to multiple drug-drug interactions. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 62-68 26327754-9 2015 There are already some approved drugs available, including irbesartan, ezetimibe, and ritonavir and cyclosporin A, with documented inhibitory effects on NTCP"s metabolic function. Ritonavir 86-95 solute carrier family 10 member 1 Homo sapiens 153-157 25831464-7 2015 RESULTS: In the on-treatment analysis, the median (interquartile range) increase in the CD4 count after 48 weeks was +193 (129-349) cells per microliter in the efavirenz arm, +197 (146-238) cells per microliter in the ritonavir-boosted atazanavir arm, and +205 (178-327) cells per microliter in the ritonavir-boosted lopinavir arm (P = 0.73). Ritonavir 299-308 CD4 molecule Homo sapiens 88-91 25831464-7 2015 RESULTS: In the on-treatment analysis, the median (interquartile range) increase in the CD4 count after 48 weeks was +193 (129-349) cells per microliter in the efavirenz arm, +197 (146-238) cells per microliter in the ritonavir-boosted atazanavir arm, and +205 (178-327) cells per microliter in the ritonavir-boosted lopinavir arm (P = 0.73). Ritonavir 218-227 CD4 molecule Homo sapiens 88-91 26196688-7 2015 On multivariate analysis, cumulative months on protease inhibitor regimens and use of ritonavir as single protease inhibitor remained significant (p = 0.008; p = 0.033). Ritonavir 86-95 serpin family A member 13, pseudogene Homo sapiens 106-124 25989229-10 2015 Exposure of rat and human hepatocytes to 0.5 muM ritonavir resulted in Cu,cell of 12 +- 1 and 8 +- 1 nM. Ritonavir 49-58 latexin Homo sapiens 45-48 25691652-2 2015 GRL-0739 blocked the infectivity and replication of HIV-1NL4-3 variants selected by concentrations of up to 5 muM ritonavir or atazanavir (EC50, 0.035 to 0.058 muM). Ritonavir 114-123 nuclear receptor subfamily 3 group C member 1 Homo sapiens 0-3 26009829-5 2015 Cystatin C decreased significantly within each arm; however, ritonavir-boosted atazanavir attenuated the beneficial effects of ART on cystatin C compared to EFV. Ritonavir 61-70 cystatin C Homo sapiens 134-144 25733917-4 2015 As a substrate of CYP 3A4, dolutegravir is affected by rifampin, efavirenz, tipranavir/ritonavir, fosamprenavir/ritonavir, and dose increase is required. Ritonavir 87-96 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 18-25 25929922-1 2015 The purpose of this article is to evaluate the evolution of microbial translocation (MT) and its role in CD4 and CD8 T cells immune activation (IA) in HIV-1-infected patients on ritonavir-boosted darunavir monotherapy (mtDRV/rtv).Prospective study of consecutive HIV-1-infected patients switched to mtDRV/rtv as a simplification regimen. Ritonavir 178-187 CD4 molecule Homo sapiens 105-108 25929922-1 2015 The purpose of this article is to evaluate the evolution of microbial translocation (MT) and its role in CD4 and CD8 T cells immune activation (IA) in HIV-1-infected patients on ritonavir-boosted darunavir monotherapy (mtDRV/rtv).Prospective study of consecutive HIV-1-infected patients switched to mtDRV/rtv as a simplification regimen. Ritonavir 178-187 CD8a molecule Homo sapiens 113-116 25742730-1 2015 BACKGROUND: Saquinavir/ritonavir (1000/100 mg twice daily [BID]) is associated with dose- and exposure-dependent prolongation of the QT interval. Ritonavir 23-32 BH3 interacting domain death agonist Homo sapiens 59-62 25608583-0 2015 Antiviral activity and CSF concentrations of 600/100 mg of darunavir/ritonavir once daily in HIV-1 patients with plasma viral suppression. Ritonavir 69-78 colony stimulating factor 2 Homo sapiens 23-26 25608583-1 2015 OBJECTIVES: The objective of this study was to assess whether a lower dose than the currently used one of darunavir/ritonavir might achieve good CSF concentrations and contribute to inhibition of CNS viral replication. Ritonavir 116-125 colony stimulating factor 2 Homo sapiens 145-148 25542900-9 2015 Ritonavir and metformin effectively suppressed AKT and mTORC1 phosphorylation and prosurvival BCL-2 family member MCL-1 expression in multiple myeloma cell lines in vitro and in vivo. Ritonavir 0-9 AKT serine/threonine kinase 1 Homo sapiens 47-50 25542900-9 2015 Ritonavir and metformin effectively suppressed AKT and mTORC1 phosphorylation and prosurvival BCL-2 family member MCL-1 expression in multiple myeloma cell lines in vitro and in vivo. Ritonavir 0-9 CREB regulated transcription coactivator 1 Mus musculus 55-61 25542900-9 2015 Ritonavir and metformin effectively suppressed AKT and mTORC1 phosphorylation and prosurvival BCL-2 family member MCL-1 expression in multiple myeloma cell lines in vitro and in vivo. Ritonavir 0-9 BCL2 apoptosis regulator Homo sapiens 94-99 25542900-9 2015 Ritonavir and metformin effectively suppressed AKT and mTORC1 phosphorylation and prosurvival BCL-2 family member MCL-1 expression in multiple myeloma cell lines in vitro and in vivo. Ritonavir 0-9 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 114-119 25733917-4 2015 As a substrate of CYP 3A4, dolutegravir is affected by rifampin, efavirenz, tipranavir/ritonavir, fosamprenavir/ritonavir, and dose increase is required. Ritonavir 112-121 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 18-25 25451053-5 2015 In a 3-day monotherapy study with HCV genotype 1-infected patients, ABT-450 was coadministered with ritonavir, a cytochrome P450 3A4 inhibitor shown previously to markedly increase peak, trough, and overall drug exposures of ABT-450. Ritonavir 100-109 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 113-132 25801567-11 2015 After adjusting for weight, eGFR, and the concomitant use of ritonavir-boosted protease inhibitors, a 30% increase in the mean tenofovir plasma concentration was observed in patients having the ABCC4 4131 TG or GG genotype. Ritonavir 61-70 ATP binding cassette subfamily C member 4 Homo sapiens 194-199 25545854-0 2015 Anti-HIV drugs, lopinavir/ritonavir and atazanavir, modulate innate immune response triggered by Leishmania in macrophages: the role of NF-kappaB and PPAR-gamma. Ritonavir 26-35 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 136-145 24828872-6 2015 Treatment of CLL cells with human immunodeficiency (HIV) protease inhibitor ritonavir, which inhibits GLUT4, elicits toxicity similar to that elicited upon glucose deprivation. Ritonavir 76-85 solute carrier family 2 member 4 Homo sapiens 102-107 25623401-6 2015 RESULTS: Expression of the ER stress markers of BiP, CHOP, and SERCA and the autophagy marker LC3 was significantly changed in PMH in response to combined alcohol, RIT, and LOP, which was companied by increased cell death compared with control. Ritonavir 164-167 heat shock protein 5 Mus musculus 48-51 24992294-14 2015 CONCLUSIONS: Despite 34% lower atazanavir exposure during pregnancy, atazanavir/ritonavir 300/100 mg once daily generates effective concentrations for protease inhibitor (PI)-naive patients, even if co-administered with tenofovir. Ritonavir 80-89 serpin family A member 13, pseudogene Homo sapiens 151-169 25283307-6 2014 The relative vitality, indicating the selective advantage of polymorphisms, of the C-SA protease relative to the subtype B protease in the presence of ritonavir and darunavir was four- and tenfold greater, respectively. Ritonavir 151-160 heat shock protein family A (Hsp70) member 9 Homo sapiens 83-87 25531880-0 2014 mTORC2 is required for rit-mediated oxidative stress resistance. Ritonavir 23-26 CREB regulated transcription coactivator 2 Mus musculus 0-6 25391550-1 2015 Cytochrome P450 (CYP) 3A4 has been considered to be the most important enzyme system for metabolism of lopinavir/ritonavir (LPV/r), a widely used HIV protease inhibitor (PI) recommended during pregnancy. Ritonavir 113-122 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-25 25151207-3 2014 METHODS: Nineteen SNPs in the ABCB1, NR1I2, UGT1A1, SLCO1B1 and CYP3A5 genes were examined in case patients with atazanavir/ritonavir-induced nephrolithiasis (n = 31) and controls (n = 47). Ritonavir 124-133 ATP binding cassette subfamily B member 1 Homo sapiens 30-35 25216238-8 2014 Possible alternatives to ketoconazole as prototype CYP3A inhibitors include ritonavir and potentially itraconazole, but not clarithromycin. Ritonavir 76-85 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 51-56 25151207-3 2014 METHODS: Nineteen SNPs in the ABCB1, NR1I2, UGT1A1, SLCO1B1 and CYP3A5 genes were examined in case patients with atazanavir/ritonavir-induced nephrolithiasis (n = 31) and controls (n = 47). Ritonavir 124-133 nuclear receptor subfamily 1 group I member 2 Homo sapiens 37-42 25151207-3 2014 METHODS: Nineteen SNPs in the ABCB1, NR1I2, UGT1A1, SLCO1B1 and CYP3A5 genes were examined in case patients with atazanavir/ritonavir-induced nephrolithiasis (n = 31) and controls (n = 47). Ritonavir 124-133 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 44-50 25151207-3 2014 METHODS: Nineteen SNPs in the ABCB1, NR1I2, UGT1A1, SLCO1B1 and CYP3A5 genes were examined in case patients with atazanavir/ritonavir-induced nephrolithiasis (n = 31) and controls (n = 47). Ritonavir 124-133 solute carrier organic anion transporter family member 1B1 Homo sapiens 52-59 25151207-3 2014 METHODS: Nineteen SNPs in the ABCB1, NR1I2, UGT1A1, SLCO1B1 and CYP3A5 genes were examined in case patients with atazanavir/ritonavir-induced nephrolithiasis (n = 31) and controls (n = 47). Ritonavir 124-133 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 64-70 24846165-3 2014 Treatments of HIV patients with CML are with HAART drugs, ritonavir and efavirenz, may cause complex drug interactions through CYP3A inhibition or induction. Ritonavir 58-67 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 127-132 25274602-2 2014 Given the potent inhibition of CYP3A4 by ritonavir, subtherapeutic doses of ritonavir are used to increase plasma concentrations of other HIV drugs oxidized by CYP3A4, thereby extending their clinical efficacy. Ritonavir 41-50 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 31-37 25274602-2 2014 Given the potent inhibition of CYP3A4 by ritonavir, subtherapeutic doses of ritonavir are used to increase plasma concentrations of other HIV drugs oxidized by CYP3A4, thereby extending their clinical efficacy. Ritonavir 76-85 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 31-37 25274602-2 2014 Given the potent inhibition of CYP3A4 by ritonavir, subtherapeutic doses of ritonavir are used to increase plasma concentrations of other HIV drugs oxidized by CYP3A4, thereby extending their clinical efficacy. Ritonavir 76-85 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 160-166 25274602-3 2014 However, the mechanism of inhibition of CYP3A4 by ritonavir remains unclear. Ritonavir 50-59 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 40-46 25274602-5 2014 The results presented here demonstrate that inhibition of CYP3A4 by ritonavir occurs by CYP3A4-mediated activation and subsequent formation of a covalent bond to the apoprotein. Ritonavir 68-77 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 58-64 25274602-5 2014 The results presented here demonstrate that inhibition of CYP3A4 by ritonavir occurs by CYP3A4-mediated activation and subsequent formation of a covalent bond to the apoprotein. Ritonavir 68-77 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 88-94 25274602-6 2014 Incubations of [(3)H]ritonavir with reconstituted CYP3A4 and human liver microsomes resulted in a covalent binding stoichiometry equal to 0.93 +- 0.04 moles of ritonavir bound per mole of inactivated CYP3A4. Ritonavir 21-30 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 50-56 25274602-6 2014 Incubations of [(3)H]ritonavir with reconstituted CYP3A4 and human liver microsomes resulted in a covalent binding stoichiometry equal to 0.93 +- 0.04 moles of ritonavir bound per mole of inactivated CYP3A4. Ritonavir 21-30 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 200-206 25274602-8 2014 Tryptic digestion of the CYP3A4-[(3)H]ritonavir incubations exhibited an adducted peptide (255-RM K: ESRLEDTQKHR-268) associated with a radiochromatic peak and a mass consistent with ritonavir plus 16 Da, in agreement with the whole-protein mass spectrometry. Ritonavir 38-47 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 25-31 25274602-8 2014 Tryptic digestion of the CYP3A4-[(3)H]ritonavir incubations exhibited an adducted peptide (255-RM K: ESRLEDTQKHR-268) associated with a radiochromatic peak and a mass consistent with ritonavir plus 16 Da, in agreement with the whole-protein mass spectrometry. Ritonavir 183-192 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 25-31 25274602-10 2014 In conclusion, ritonavir exhibited potent time-dependent inactivation of CYP3A, with the mechanism of inactivation occurring though a covalent bond to Lys257 of the CYP3A4 apoprotein. Ritonavir 15-24 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 165-171 25394078-2 2014 Previous studies suggested that TDF toxicity is higher when it is co-administered with ritonavir-boosted protease inhibitors (PI/r)[3, 4]. Ritonavir 87-96 sex determining region Y Homo sapiens 32-35 24997317-0 2014 Intracellular accumulation of atazanavir/ritonavir according to plasma concentrations and OATP1B1, ABCB1 and PXR genetic polymorphisms. Ritonavir 41-50 solute carrier organic anion transporter family member 1B1 Homo sapiens 90-97 24997317-0 2014 Intracellular accumulation of atazanavir/ritonavir according to plasma concentrations and OATP1B1, ABCB1 and PXR genetic polymorphisms. Ritonavir 41-50 ATP binding cassette subfamily B member 1 Homo sapiens 99-104 24997317-11 2014 Median ritonavir intracellular concentrations were higher for OATP1B1 521 T C TC or CC carriers and for PXR 44477 A G AG or GG carriers. Ritonavir 7-16 solute carrier organic anion transporter family member 1B1 Homo sapiens 62-69 24997317-11 2014 Median ritonavir intracellular concentrations were higher for OATP1B1 521 T C TC or CC carriers and for PXR 44477 A G AG or GG carriers. Ritonavir 7-16 nuclear receptor subfamily 1 group I member 2 Homo sapiens 104-107 24997317-14 2014 Ritonavir intracellular concentrations were associated with OATP1B1 521 and PXR 44477 SNPs while intracellular atazanavir exposure was associated with the ABCB1 2677 SNP. Ritonavir 0-9 solute carrier organic anion transporter family member 1B1 Homo sapiens 60-67 24997317-14 2014 Ritonavir intracellular concentrations were associated with OATP1B1 521 and PXR 44477 SNPs while intracellular atazanavir exposure was associated with the ABCB1 2677 SNP. Ritonavir 0-9 nuclear receptor subfamily 1 group I member 2 Homo sapiens 76-79 25397574-13 2014 CONCLUSIONS: Antiretroviral regimens containing atazanavir with or without ritonavir were durable and well tolerated, an elevated viral load and CD4 <200 cells/microL at baseline resulted related to regimen discontinuation and reduced CD4 recovery. Ritonavir 75-84 CD4 molecule Homo sapiens 145-148 23883365-11 2014 The permeability of felodipine was increased in presence of naringenin and ritonavir (standard P-glycoprotein (P-gp) and Cytochrome P450 (CYP)3A4 inhibitor). Ritonavir 75-84 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 95-109 25164142-6 2014 Both agents inhibit CYP3A4, with cobicistat being a more specific CYP inhibitor than ritonavir. Ritonavir 85-94 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 20-26 23883365-11 2014 The permeability of felodipine was increased in presence of naringenin and ritonavir (standard P-glycoprotein (P-gp) and Cytochrome P450 (CYP)3A4 inhibitor). Ritonavir 75-84 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 111-115 24516079-1 2014 Hyperbilirubinaemia with or without jaundice is one of the side effects of atazanavir boosted with low-dose ritonavir (ATV/rit) related to the drug plasma levels, as a result of its metabolism by UGT1A1 - uridine diphosphate-glucuronosyl transferase. Ritonavir 108-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 196-202 24516079-8 2014 Of the ATV/rit-treated patients, 14 were found to be carriers of the UGT1A1*28 variant (54%), and maximum serum bilirubin levels were significantly higher in the carrier population (4.71 vs. 2.69 mg/dL, p = 0.026). Ritonavir 11-14 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-75 24929949-4 2014 High-dose ritonavir has been shown to increase olanzapine elimination through induction of CYP1A2 and/or UGT, but the effect of low-dose ritonavir on olanzapine pharmacokinetics is unknown. Ritonavir 10-19 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 91-97 25079492-1 2014 To investigate the effect of ritonavir on hepatocyte proliferation, we detected the change of cleaved caspase-3 expression level in the hepatocytes. Ritonavir 29-38 caspase 3 Mus musculus 102-111 25079492-3 2014 The results showed that ritonavir can evidently inhibit hepatocyte proliferation and increase cleaved caspase-3 expression level. Ritonavir 24-33 caspase 3 Mus musculus 102-111 27129005-6 2014 The modest increases in AUC and Cmax following co-administration of GSK2336805 plus ritonavir suggest that GSK2336805 when given concomitantly with a single CYP3A/Pgp inhibiting drug will not likely require dose adjustment. Ritonavir 84-93 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 157-162 27129005-6 2014 The modest increases in AUC and Cmax following co-administration of GSK2336805 plus ritonavir suggest that GSK2336805 when given concomitantly with a single CYP3A/Pgp inhibiting drug will not likely require dose adjustment. Ritonavir 84-93 phosphoglycolate phosphatase Homo sapiens 163-166 24973543-0 2014 Investigating the contribution of CYP2J2 to ritonavir metabolism in vitro and in vivo. Ritonavir 44-53 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 34-40 24973543-2 2014 Ritonavir pharmacokinetics are complicated by inhibition, induction and pharmacogenetics of cytochrome P450 (CYP) enzymes mediating its clearance. Ritonavir 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-107 24973543-2 2014 Ritonavir pharmacokinetics are complicated by inhibition, induction and pharmacogenetics of cytochrome P450 (CYP) enzymes mediating its clearance. Ritonavir 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-112 24973543-3 2014 This investigation revealed that CYP2J2, along with CYP3A4/5 and CYP2D6, efficiently metabolizes ritonavir, and to a CYP2J2-specific (minor) metabolite. Ritonavir 97-106 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 33-39 24973543-3 2014 This investigation revealed that CYP2J2, along with CYP3A4/5 and CYP2D6, efficiently metabolizes ritonavir, and to a CYP2J2-specific (minor) metabolite. Ritonavir 97-106 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 52-58 24973543-3 2014 This investigation revealed that CYP2J2, along with CYP3A4/5 and CYP2D6, efficiently metabolizes ritonavir, and to a CYP2J2-specific (minor) metabolite. Ritonavir 97-106 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 65-71 24973543-8 2014 A modest (2-6%) contribution of CYP2J2 to ritonavir clearance is predicted which increases to more than 20% in subjects carrying CYP2D6 poor metabolizer polymorphisms and CYP3A4 irreversible inhibition. Ritonavir 42-51 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 32-38 24973543-8 2014 A modest (2-6%) contribution of CYP2J2 to ritonavir clearance is predicted which increases to more than 20% in subjects carrying CYP2D6 poor metabolizer polymorphisms and CYP3A4 irreversible inhibition. Ritonavir 42-51 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 129-135 24973543-8 2014 A modest (2-6%) contribution of CYP2J2 to ritonavir clearance is predicted which increases to more than 20% in subjects carrying CYP2D6 poor metabolizer polymorphisms and CYP3A4 irreversible inhibition. Ritonavir 42-51 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 171-177 24950369-8 2014 A simulated dose-reduction scenario showed that 200/100 mg lopinavir/ritonavir was adequate to achieve therapeutic concentration in individuals with CYP3A4*22/*22 alone or in combination with SLCO1B1 521CC. Ritonavir 69-78 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 149-155 24950369-8 2014 A simulated dose-reduction scenario showed that 200/100 mg lopinavir/ritonavir was adequate to achieve therapeutic concentration in individuals with CYP3A4*22/*22 alone or in combination with SLCO1B1 521CC. Ritonavir 69-78 solute carrier organic anion transporter family member 1B1 Homo sapiens 192-199 24929949-4 2014 High-dose ritonavir has been shown to increase olanzapine elimination through induction of CYP1A2 and/or UGT, but the effect of low-dose ritonavir on olanzapine pharmacokinetics is unknown. Ritonavir 10-19 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 105-108 24986243-2 2014 In particular, CYP3A4 and CYP3A5 (interacting with more than 60% of licensed drugs) exhibit the most individual variations of gene expression, mostly caused by single nucleotide polymorphisms (SNPs) within the regulatory region of the CYP3A4 and CYP3A5 genes which might affect the level of enzyme production.In this study, we sought to improve the performance of sensitive screening for CYP3A polymorphism detection in twenty HIV-1 infected patients undergoing lopinavir/ritonavir (LPV/r) monotherapy. Ritonavir 472-481 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 15-21 24986243-2 2014 In particular, CYP3A4 and CYP3A5 (interacting with more than 60% of licensed drugs) exhibit the most individual variations of gene expression, mostly caused by single nucleotide polymorphisms (SNPs) within the regulatory region of the CYP3A4 and CYP3A5 genes which might affect the level of enzyme production.In this study, we sought to improve the performance of sensitive screening for CYP3A polymorphism detection in twenty HIV-1 infected patients undergoing lopinavir/ritonavir (LPV/r) monotherapy. Ritonavir 472-481 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 26-32 24986243-2 2014 In particular, CYP3A4 and CYP3A5 (interacting with more than 60% of licensed drugs) exhibit the most individual variations of gene expression, mostly caused by single nucleotide polymorphisms (SNPs) within the regulatory region of the CYP3A4 and CYP3A5 genes which might affect the level of enzyme production.In this study, we sought to improve the performance of sensitive screening for CYP3A polymorphism detection in twenty HIV-1 infected patients undergoing lopinavir/ritonavir (LPV/r) monotherapy. Ritonavir 472-481 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 235-241 24986243-2 2014 In particular, CYP3A4 and CYP3A5 (interacting with more than 60% of licensed drugs) exhibit the most individual variations of gene expression, mostly caused by single nucleotide polymorphisms (SNPs) within the regulatory region of the CYP3A4 and CYP3A5 genes which might affect the level of enzyme production.In this study, we sought to improve the performance of sensitive screening for CYP3A polymorphism detection in twenty HIV-1 infected patients undergoing lopinavir/ritonavir (LPV/r) monotherapy. Ritonavir 472-481 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 246-252 24986243-2 2014 In particular, CYP3A4 and CYP3A5 (interacting with more than 60% of licensed drugs) exhibit the most individual variations of gene expression, mostly caused by single nucleotide polymorphisms (SNPs) within the regulatory region of the CYP3A4 and CYP3A5 genes which might affect the level of enzyme production.In this study, we sought to improve the performance of sensitive screening for CYP3A polymorphism detection in twenty HIV-1 infected patients undergoing lopinavir/ritonavir (LPV/r) monotherapy. Ritonavir 472-481 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 15-20 24434120-7 2014 In male Lewis-Brown Norway rats, RTV administration (75 mg/kg/day, 5 weeks) resulted in an 85% increase in plasma 8-isoprostane and a 23% decrease in hepatic eNOS mRNA; concomitantly, eNOS protein decreased 75%, whereas plasma nitrite level was reduced 48%. Ritonavir 33-36 nitric oxide synthase 3 Rattus norvegicus 158-162 24648507-0 2014 Low-density lipoprotein and ritonavir: an interaction between antiretrovirals and lipids mediated by P-glycoprotein. Ritonavir 28-37 ATP binding cassette subfamily B member 1 Homo sapiens 101-115 24648507-9 2014 Notably, ritonavir induced reduced cholesterol mobilization in THP-1 cells, probably due to its inhibitory action on P-gp activity. Ritonavir 9-18 ATP binding cassette subfamily B member 1 Homo sapiens 117-121 24648507-10 2014 CONCLUSIONS: Our data indicate a potential interplay between LDL and ritonavir mediated by P-gp. Ritonavir 69-78 ATP binding cassette subfamily B member 1 Homo sapiens 91-95 24259240-0 2014 Insulin resistance and lipid profiles in HIV-infected Thai children receiving lopinavir/ritonavir-based highly active antiretroviral therapy. Ritonavir 88-97 insulin Homo sapiens 0-7 24259240-1 2014 BACKGROUND: Lopinavir/ritonavir (LPV/r) is associated with insulin resistance (IR). Ritonavir 22-31 insulin Homo sapiens 59-66 24488374-16 2014 However, the CYP3A4 inhibitors ritonavir and ketoconazole resulted in a 6.9- and 3.1-fold increase in AUC, respectively. Ritonavir 31-40 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 13-19 24075526-9 2014 Lopinavir/ritonavir, etravirine, and lamivudine administered through a jejunostomy resulted effective in decreasing HIV viral load and increasing CD4 lymphocyte count in a HIV patient who could not receive treatment orally. Ritonavir 10-19 CD4 molecule Homo sapiens 146-149 24474568-3 2014 METHODS: In a modified phase 1 study of sunitinib, patients were stratified into 2 treatment arms based on whether they were receiving therapy with ritonavir, a potent CYP3A4 inhibitor. Ritonavir 148-157 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 168-174 24434120-7 2014 In male Lewis-Brown Norway rats, RTV administration (75 mg/kg/day, 5 weeks) resulted in an 85% increase in plasma 8-isoprostane and a 23% decrease in hepatic eNOS mRNA; concomitantly, eNOS protein decreased 75%, whereas plasma nitrite level was reduced 48%. Ritonavir 33-36 nitric oxide synthase 3 Rattus norvegicus 184-188 24399452-2 2014 As afatinib is a substrate for the P-glycoprotein (P-gp) pump transporter the three studies presented here investigated the pharmacokinetics of afatinib in the presence of a potent inhibitor (ritonavir) or inducer [rifampicin (rifampin)] of P-gp. Ritonavir 192-201 ATP binding cassette subfamily B member 1 Homo sapiens 51-55 24696866-8 2014 Although boosting it with a strong CYP3A inhibitor such as ritonavir substantially increases its plasma exposure and prolongs its elimination half-life, other combinations or even monotherapy could also be considered. Ritonavir 59-68 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 35-40 24411125-1 2014 Ritonavir (RTV), an HIV-1 protease inhibitor (PI), is also a potent mechanism-based inhibitor of human cytochrome P450 3A (CYP3A) and has been widely prescribed as a pharmacoenhancer. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 103-121 24412072-2 2014 The HIV protease inhibitor (PI) ritonavir (RTV) has been widely used as a pharmacoenhancer for other PIs, which are substrates of cytochrome P450 3A (CYP3A). Ritonavir 32-41 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 130-148 24411125-1 2014 Ritonavir (RTV), an HIV-1 protease inhibitor (PI), is also a potent mechanism-based inhibitor of human cytochrome P450 3A (CYP3A) and has been widely prescribed as a pharmacoenhancer. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 123-128 24412072-2 2014 The HIV protease inhibitor (PI) ritonavir (RTV) has been widely used as a pharmacoenhancer for other PIs, which are substrates of cytochrome P450 3A (CYP3A). Ritonavir 32-41 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 150-155 24412072-2 2014 The HIV protease inhibitor (PI) ritonavir (RTV) has been widely used as a pharmacoenhancer for other PIs, which are substrates of cytochrome P450 3A (CYP3A). Ritonavir 43-46 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 130-148 24412072-2 2014 The HIV protease inhibitor (PI) ritonavir (RTV) has been widely used as a pharmacoenhancer for other PIs, which are substrates of cytochrome P450 3A (CYP3A). Ritonavir 43-46 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 150-155 24411125-1 2014 Ritonavir (RTV), an HIV-1 protease inhibitor (PI), is also a potent mechanism-based inhibitor of human cytochrome P450 3A (CYP3A) and has been widely prescribed as a pharmacoenhancer. Ritonavir 11-14 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 103-121 24411125-1 2014 Ritonavir (RTV), an HIV-1 protease inhibitor (PI), is also a potent mechanism-based inhibitor of human cytochrome P450 3A (CYP3A) and has been widely prescribed as a pharmacoenhancer. Ritonavir 11-14 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 123-128 24290411-2 2014 It is co-administered with low-dose ritonavir, a potent CYP3A inhibitor, to enhance danoprevir pharmacokinetics. Ritonavir 36-45 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 56-61 24343782-2 2014 Cobicistat has a lower potential for off-target drug interactions than the standard boosting agent ritonavir, due to its more selective inhibition of CYP3A and lower likelihood for enzymatic induction, and is devoid of anti-HIV activity. Ritonavir 99-108 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 150-155 24315932-8 2014 We found that ritonavir administration caused intestinal damage and its co-administration with naproxen or ASA exacerbated the severity of injury and intestinal inflammation, as assessed by measuring haematocrit, MPO, mucosal levels of PGE2 and mRNA levels of iNOS, MCP-1 and VLA-1. Ritonavir 14-23 myeloperoxidase Homo sapiens 213-216 24333052-0 2014 Effect of ritonavir-induced cytochrome P450 3A4 inhibition on plasma fentanyl concentrations during patient-controlled epidural labor analgesia: a pharmacokinetic simulation. Ritonavir 10-19 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 28-47 24333052-1 2014 BACKGROUND: Ritonavir inhibition of cytochrome P450 3A4 decreases the elimination clearance of fentanyl by 67%. Ritonavir 12-21 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 36-55 24333052-2 2014 We used a pharmacokinetic model developed from published data to simulate the effect of sample patient-controlled epidural labor analgesic regimens on plasma fentanyl concentrations in the absence and presence of ritonavir-induced cytochrome P450 3A4 inhibition. Ritonavir 213-222 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 231-250 24333052-12 2014 CONCLUSION: Our model predicts that even with maximal clinical dosing regimens of epidural fentanyl over 24 h, ritonavir-induced cytochrome P450 3A4 inhibition is unlikely to produce plasma fentanyl concentrations associated with a decrease in minute ventilation. Ritonavir 111-120 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 129-148 24242708-8 2014 Inhibition of CYP3A activity using the specific inhibitor ritonavir resulted in alleviation of the anti-proliferative response of VDR ligands in prostate cancer cells. Ritonavir 58-67 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 14-19 24242708-8 2014 Inhibition of CYP3A activity using the specific inhibitor ritonavir resulted in alleviation of the anti-proliferative response of VDR ligands in prostate cancer cells. Ritonavir 58-67 vitamin D receptor Homo sapiens 130-133 24212378-4 2014 From a set of potential P-gp inhibitors, clarithromycin, cyclosporin A, itraconazole, ketoconazole, quinidine, and ritonavir inhibited P-gp-mediated transport of dabigatran etexilate over a concentration range that may hypothetically occur in the intestine. Ritonavir 115-124 ATP binding cassette subfamily B member 1 Homo sapiens 24-28 24212378-4 2014 From a set of potential P-gp inhibitors, clarithromycin, cyclosporin A, itraconazole, ketoconazole, quinidine, and ritonavir inhibited P-gp-mediated transport of dabigatran etexilate over a concentration range that may hypothetically occur in the intestine. Ritonavir 115-124 ATP binding cassette subfamily B member 1 Homo sapiens 135-139 24315932-8 2014 We found that ritonavir administration caused intestinal damage and its co-administration with naproxen or ASA exacerbated the severity of injury and intestinal inflammation, as assessed by measuring haematocrit, MPO, mucosal levels of PGE2 and mRNA levels of iNOS, MCP-1 and VLA-1. Ritonavir 14-23 nitric oxide synthase 2 Homo sapiens 260-264 24315932-8 2014 We found that ritonavir administration caused intestinal damage and its co-administration with naproxen or ASA exacerbated the severity of injury and intestinal inflammation, as assessed by measuring haematocrit, MPO, mucosal levels of PGE2 and mRNA levels of iNOS, MCP-1 and VLA-1. Ritonavir 14-23 C-C motif chemokine ligand 2 Homo sapiens 266-271 24315932-8 2014 We found that ritonavir administration caused intestinal damage and its co-administration with naproxen or ASA exacerbated the severity of injury and intestinal inflammation, as assessed by measuring haematocrit, MPO, mucosal levels of PGE2 and mRNA levels of iNOS, MCP-1 and VLA-1. Ritonavir 14-23 integrin subunit alpha 1 Homo sapiens 276-281 24202050-0 2014 Ritonavir inhibits HIF-1alpha-mediated VEGF expression in retinal pigment epithelial cells in vitro. Ritonavir 0-9 hypoxia inducible factor 1 subunit alpha Homo sapiens 19-29 24451403-0 2014 Ritonavir-Mediated Induction of Apoptosis in Pancreatic Cancer Occurs via the RB/E2F-1 and AKT Pathways. Ritonavir 0-9 E2F transcription factor 1 Homo sapiens 81-86 24451403-0 2014 Ritonavir-Mediated Induction of Apoptosis in Pancreatic Cancer Occurs via the RB/E2F-1 and AKT Pathways. Ritonavir 0-9 AKT serine/threonine kinase 1 Homo sapiens 91-94 24379203-11 2014 In Cyp mice pretreated with ritonavir, significant (P<0.05) increases in atazanavir Cbrain/Cplasma (1.8-fold) and Ctestes/Cplasma (9.5-fold) ratios compared to those in vehicle-treated WT mice were observed. Ritonavir 28-37 peptidyl-prolyl isomerase G (cyclophilin G) Mus musculus 3-6 24372550-7 2014 Interestingly, atazanavir and ritonavir also activated LXRalpha/beta in reporter assays, while tipranavir enhanced transcriptional activity of ERalpha. Ritonavir 30-39 nuclear receptor subfamily 1 group H member 3 Homo sapiens 55-63 24805065-0 2014 Ritonavir analogues as a probe for deciphering the cytochrome P450 3A4 inhibitory mechanism. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 51-70 24805065-3 2014 Currently, the CYP3A4 inhibitor ritonavir and its derivative cobicistat are prescribed to HIV patients as pharmacoenhancers. Ritonavir 32-41 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 15-21 24805065-5 2014 To unravel the structural basis of CYP3A4 inhibition, we compared the binding modes of ritonavir and ten analogues using biochemical, mutagenesis and x-ray crystallography techniques. Ritonavir 87-96 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 35-41 24805065-6 2014 This review summarizes our findings on the relative contribution of the heme-ligating moiety, side chains and the terminal group of ritonavir-like molecules to the ligand binding process, and highlights strategies for a structure-guided design of CYP3A4 inactivators. Ritonavir 132-141 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 247-253 24202050-0 2014 Ritonavir inhibits HIF-1alpha-mediated VEGF expression in retinal pigment epithelial cells in vitro. Ritonavir 0-9 vascular endothelial growth factor A Homo sapiens 39-43 24202050-3 2014 Hence, the primary objective of this study was to examine the expression of HIF-1alpha and VEGF in human retinal pigment epithelial cells treated with ritonavir under hypoxic and normoxic conditions. Ritonavir 151-160 hypoxia inducible factor 1 subunit alpha Homo sapiens 76-86 24202050-3 2014 Hence, the primary objective of this study was to examine the expression of HIF-1alpha and VEGF in human retinal pigment epithelial cells treated with ritonavir under hypoxic and normoxic conditions. Ritonavir 151-160 vascular endothelial growth factor A Homo sapiens 91-95 24202050-8 2014 Presence of ritonavir in the culture medium strongly inhibited VEGF expression in a concentration-dependent manner under hypoxic conditions. Ritonavir 12-21 vascular endothelial growth factor A Homo sapiens 63-67 24202050-9 2014 Immunoblot analysis demonstrated a substantially reduced protein expression of HIF-1alpha in the presence of ritonavir. Ritonavir 109-118 hypoxia inducible factor 1 subunit alpha Homo sapiens 79-89 24202050-10 2014 Further, hypoxic exposure-induced VEGF secretion was also inhibited by ritonavir, as demonstrated using ELISA. Ritonavir 71-80 vascular endothelial growth factor A Homo sapiens 34-38 24202050-13 2014 CONCLUSIONS: This study demonstrates for the first time that ritonavir can inhibit HIF-1alpha and VEGF in ARPE-19 and D407 cells. Ritonavir 61-70 hypoxia inducible factor 1 subunit alpha Homo sapiens 83-93 24202050-13 2014 CONCLUSIONS: This study demonstrates for the first time that ritonavir can inhibit HIF-1alpha and VEGF in ARPE-19 and D407 cells. Ritonavir 61-70 vascular endothelial growth factor A Homo sapiens 98-102 23811743-6 2014 FPV 45 mg/kg boosted with RTV 7 to 10 mg/kg BID achieved average plasma amprenavir area under curve(0-tau) values 26% to 28% lower and Cmax similar to historical adult data for FPV/RTV 700/100 mg BID; amprenavir Ctau values were lower in the subjects <6 months of age. Ritonavir 26-29 BH3 interacting domain death agonist Homo sapiens 196-199 24577112-2 2014 We reported previously that prophylactic postnatal ritonavir-PI therapy in HIV-exposed neonates was associated with increases in plasma 17-hydroxyprogesterone (17-OHP) and dehydroepiandrosterone sulfate (DHEA-S). Ritonavir 51-60 sulfotransferase family 2A member 1 Homo sapiens 204-210 23811744-8 2014 Median increases in absolute and relative (percentage) CD4 counts from baseline to week 48 occurred in both the fosamprenavir (340 cells/mm; 8%) and fosamprenavir/ritonavir group (190 cells/mm; 8%). Ritonavir 163-172 CD4 molecule Homo sapiens 55-58 23973637-6 2013 A comparison among the most clinically used PIs, ritonavir (RTV), LPV, and DRV, revealed that LPV potently and RTV moderately but not DRV induced ER stress via ROS-dependent JNK activation rather than proteasome inhibition. Ritonavir 60-63 mitogen-activated protein kinase 8 Homo sapiens 174-177 25715484-4 2014 A combination of once daily darunavir/ritonavir 800/100 mg ensured a more marked immunological improvement: a difference in the increment (77.1 in 1 mul) of CD4+ lymphocyte count in patients who took darunavir (362.2 in mul) and in those who used atazanavir (285.1 in mul). Ritonavir 38-47 CD4 molecule Homo sapiens 157-160 24396625-14 2013 Darunavir/ritonavir-based therapy demonstrated improvements in CD4+ cell recovery and association with progressive functional immune recovery over 48 weeks. Ritonavir 10-19 CD4 molecule Homo sapiens 63-66 24334181-1 2013 BACKGROUND: ACTG A5202 randomized treatment-naive individuals to tenofovir-emtricitabine (TDF/FTC) or abacavir-lamivudine (ABC/3TC) combined with efavirenz (EFV) or atazanavir/ritonavir (ATV/r). Ritonavir 176-185 actin gamma 1 Homo sapiens 12-16 23872824-0 2013 A randomised study of the effect of danoprevir/ritonavir or ritonavir on substrates of cytochrome P450 (CYP) 3A and 2C9 in chronic hepatitis C patients using a drug cocktail. Ritonavir 60-69 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 87-111 23872824-8 2013 CONCLUSIONS: Substantial inhibition of CYP3A- and modest induction of CYP2C9- activity were observed with danoprevir/r and low-dose ritonavir. Ritonavir 132-141 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 39-44 23872824-8 2013 CONCLUSIONS: Substantial inhibition of CYP3A- and modest induction of CYP2C9- activity were observed with danoprevir/r and low-dose ritonavir. Ritonavir 132-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 24014652-5 2013 Caco-2 cell uptake of the P-gp substrate R6G was increased by the P-gp inhibitors cyclosporine A (CSA) and ritonavir but not by DLM, CPM, and TPM. Ritonavir 107-116 ATP binding cassette subfamily B member 1 Homo sapiens 26-30 24014652-5 2013 Caco-2 cell uptake of the P-gp substrate R6G was increased by the P-gp inhibitors cyclosporine A (CSA) and ritonavir but not by DLM, CPM, and TPM. Ritonavir 107-116 ATP binding cassette subfamily B member 1 Homo sapiens 66-70 23872824-0 2013 A randomised study of the effect of danoprevir/ritonavir or ritonavir on substrates of cytochrome P450 (CYP) 3A and 2C9 in chronic hepatitis C patients using a drug cocktail. Ritonavir 47-56 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 87-111 23892274-12 2013 CONCLUSION: Rh2 inhibits the efflux of ritonavir through P-gp in vitro. Ritonavir 39-48 ATP binding cassette subfamily B member 1 Homo sapiens 57-61 23939663-2 2013 In 18 healthy participants individual CYP3A activity was quantified using midazolam metabolic clearance both alone and during CYP3A inhibition with 40 mg ritonavir. Ritonavir 154-163 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 38-43 23703578-10 2013 All HIV PIs except nelfinavir are coadministered with a low dose of ritonavir, a potent CYP3A inhibitor to improve their pharmacokinetic properties. Ritonavir 68-77 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 88-93 23703578-14 2013 Simvastatin and lovastatin metabolized through CYP3A have the highest potency for drug-drug interaction with potent CYP3A inhibitors such as ritonavir- or cobicistat-boosted HIV-PI or the hepatitis C virus (HCV) PI, telaprevir or boceprevir, and therefore their coadministration is contraindicated. Ritonavir 141-150 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 47-52 23703578-14 2013 Simvastatin and lovastatin metabolized through CYP3A have the highest potency for drug-drug interaction with potent CYP3A inhibitors such as ritonavir- or cobicistat-boosted HIV-PI or the hepatitis C virus (HCV) PI, telaprevir or boceprevir, and therefore their coadministration is contraindicated. Ritonavir 141-150 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 116-121 23886699-0 2013 The effect of ritonavir on human CYP2B6 catalytic activity: heme modification contributes to the mechanism-based inactivation of CYP2B6 and CYP3A4 by ritonavir. Ritonavir 14-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-39 23873286-11 2013 In addition, for both victim drugs, the extent of DDI when co-administered was greater with ritonavir compared with ketoconazole, in line with their in vitro CYP3A inhibition potency in RLM. Ritonavir 92-101 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 158-163 23886699-0 2013 The effect of ritonavir on human CYP2B6 catalytic activity: heme modification contributes to the mechanism-based inactivation of CYP2B6 and CYP3A4 by ritonavir. Ritonavir 14-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 129-135 23886699-0 2013 The effect of ritonavir on human CYP2B6 catalytic activity: heme modification contributes to the mechanism-based inactivation of CYP2B6 and CYP3A4 by ritonavir. Ritonavir 14-23 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 140-146 23886699-0 2013 The effect of ritonavir on human CYP2B6 catalytic activity: heme modification contributes to the mechanism-based inactivation of CYP2B6 and CYP3A4 by ritonavir. Ritonavir 150-159 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-39 23886699-0 2013 The effect of ritonavir on human CYP2B6 catalytic activity: heme modification contributes to the mechanism-based inactivation of CYP2B6 and CYP3A4 by ritonavir. Ritonavir 150-159 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 129-135 24114622-10 2013 Moreover, concomitant use with strong CYP3A4 inhibitors (such as ketoconazole, itraconazole, clarithromycin, ritonavir, telithromycin, etc.) Ritonavir 109-118 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 38-44 23886699-0 2013 The effect of ritonavir on human CYP2B6 catalytic activity: heme modification contributes to the mechanism-based inactivation of CYP2B6 and CYP3A4 by ritonavir. Ritonavir 150-159 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 140-146 23886699-1 2013 The mechanism-based inactivation of human CYP2B6 by ritonavir (RTV) in a reconstituted system was investigated. Ritonavir 52-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-48 23886699-1 2013 The mechanism-based inactivation of human CYP2B6 by ritonavir (RTV) in a reconstituted system was investigated. Ritonavir 63-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-48 23886699-4 2013 Inactivation of CYP2B6 by incubation with 10 muM RTV for 10 min resulted in an approximately 50% loss of catalytic activity and native heme, but no modification of the apoprotein was observed. Ritonavir 49-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 23886699-9 2013 Furthermore, LC-MS/MS analysis revealed that both CYP3A4 and human liver microsomes form an RTV-glutathione conjugate having a MH+ at m/z 858 during metabolism of RTV, suggesting the formation of an isocyanate intermediate leading to formation of the conjugate. Ritonavir 92-95 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 50-56 23886699-9 2013 Furthermore, LC-MS/MS analysis revealed that both CYP3A4 and human liver microsomes form an RTV-glutathione conjugate having a MH+ at m/z 858 during metabolism of RTV, suggesting the formation of an isocyanate intermediate leading to formation of the conjugate. Ritonavir 163-166 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 50-56 23305158-7 2013 A significant increase in rivaroxaban exposure was demonstrated with the strong CYP3A4, P-gp/Bcrp (ABCG2) inhibitors (and potential CYP2J2 inhibitors) ketoconazole (158% increase [95% CI 136%, 182%] for a 400 mg once daily dose) and ritonavir (153% increase [95% CI 134%, 174%]). Ritonavir 233-242 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 80-86 23836171-8 2013 Amprenavir, atazanavir, darunavir, efavirenz, ritonavir, and lopinavir were found to activate hPXR, whereas abacavir, efavirenz, and nevirapine were found to activate hCAR. Ritonavir 46-55 nuclear receptor subfamily 1 group I member 2 Homo sapiens 94-98 23836171-8 2013 Amprenavir, atazanavir, darunavir, efavirenz, ritonavir, and lopinavir were found to activate hPXR, whereas abacavir, efavirenz, and nevirapine were found to activate hCAR. Ritonavir 46-55 CXADR Ig-like cell adhesion molecule Homo sapiens 167-171 23748748-0 2013 Concentration effect relationship of CYP3A inhibition by ritonavir in humans. Ritonavir 57-66 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 37-42 23748748-1 2013 PURPOSE: To investigate the dose and concentration dependency of CYP3A inhibition by ritonavir using the established limited sampling strategy with midazolam for CYP3A activity. Ritonavir 85-94 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 65-70 23748748-1 2013 PURPOSE: To investigate the dose and concentration dependency of CYP3A inhibition by ritonavir using the established limited sampling strategy with midazolam for CYP3A activity. Ritonavir 85-94 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 162-167 23748748-3 2013 Single ascending doses of ritonavir (0.1-300 mg) were evaluated for CYP3A inhibition in two cohorts using midazolam as a marker substance. Ritonavir 26-35 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 68-73 23748748-4 2013 RESULTS: Ritonavir administered as a single oral dose produced a dose-dependent CYP3A inhibition with an ID50 of 3.4 mg. Ritonavir 9-18 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 80-85 23748748-7 2013 CONCLUSIONS: Ritonavir shows a dose and concentration effect relationship of CYP3A inhibition. Ritonavir 13-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 77-82 23305158-7 2013 A significant increase in rivaroxaban exposure was demonstrated with the strong CYP3A4, P-gp/Bcrp (ABCG2) inhibitors (and potential CYP2J2 inhibitors) ketoconazole (158% increase [95% CI 136%, 182%] for a 400 mg once daily dose) and ritonavir (153% increase [95% CI 134%, 174%]). Ritonavir 233-242 ATP binding cassette subfamily B member 1 Homo sapiens 88-92 23440887-5 2013 Among them, saquinavir and ritonavir in addition to CsA exhibited long-lasting inhibitory effects on OATP1B1-mediated transport of E1 S at >= 5 and 25 muM, respectively, even after they were washed out from the incubation buffer. Ritonavir 27-36 solute carrier organic anion transporter family member 1B1 Homo sapiens 101-108 23712757-3 2013 OBJECTIVE: The objective of this study was to evaluate the effect of a potent OATP inhibitor, ciclosporin, on danoprevir pharmacokinetics, when administered as danoprevir/ritonavir. Ritonavir 171-180 solute carrier organic anion transporter family member 1A2 Homo sapiens 78-82 23712757-16 2013 Therefore, co-administration of danoprevir/ritonavir with potent OATP inhibitors should be undertaken with appropriate precautions. Ritonavir 43-52 solute carrier organic anion transporter family member 1A2 Homo sapiens 65-69 23440887-0 2013 Long-lasting inhibitory effects of saquinavir and ritonavir on OATP1B1-mediated uptake. Ritonavir 50-59 solute carrier organic anion transporter family member 1B1 Homo sapiens 63-70 23440887-8 2013 The present study firstly showed that saquinavir and ritonavir as well as CsA have long-lasting inhibitory effects on OATP1B1. Ritonavir 53-62 solute carrier organic anion transporter family member 1B1 Homo sapiens 118-125 23479398-3 2013 Efavirenz and protease inhibitor boosted with ritonavir are major components of the antiretroviral therapy and are inducers and/or inhibitors of several cytochrome P450 (CYP) isoforms. Ritonavir 46-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 153-168 23867790-2 2013 METHODS: The effects of IGF-I on ritonavir-induced adipocyte cell death were studied in vitro. Ritonavir 33-42 insulin like growth factor 1 Homo sapiens 24-29 23441978-2 2013 Darunavir/ritonavir is a substrate and inhibitor of CYP3A. Ritonavir 10-19 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 52-57 23746300-0 2013 Dissecting cytochrome P450 3A4-ligand interactions using ritonavir analogues. Ritonavir 57-66 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 11-30 23746300-1 2013 Cytochrome P450 3A4 (CYP3A4) inhibitors ritonavir and cobicistat, currently administered to HIV patients as pharmacoenhancers, were designed on the basis of the chemical structure/activity relationships rather than the CYP3A4 crystal structure. Ritonavir 40-49 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-19 23746300-1 2013 Cytochrome P450 3A4 (CYP3A4) inhibitors ritonavir and cobicistat, currently administered to HIV patients as pharmacoenhancers, were designed on the basis of the chemical structure/activity relationships rather than the CYP3A4 crystal structure. Ritonavir 40-49 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 21-27 23479398-3 2013 Efavirenz and protease inhibitor boosted with ritonavir are major components of the antiretroviral therapy and are inducers and/or inhibitors of several cytochrome P450 (CYP) isoforms. Ritonavir 46-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 170-173 23597294-7 2013 At a similar supersaturation ratio of ~1.2, ritonavir and celecoxib had slower normalized crystal growth rates (0.20 and 0.91 mg min(-1) m(-2), respectively), while the normalized crystal growth rate of efavirenz was significantly higher (2.97 mg min(-1) m(-2)), resulting in lower levels of crystal growth inhibition by the polymers for efavirenz. Ritonavir 44-53 CD59 molecule (CD59 blood group) Homo sapiens 129-135 26251758-0 2013 Lopinavir/Ritonavir Impairs Physical Strength in Association with Reduced Igf1 Expression in Skeletal Muscle of Older Mice. Ritonavir 10-19 insulin-like growth factor 1 Mus musculus 74-78 23814462-8 2013 In the SPIRIT study (a switch study), significantly greater improvements from baseline in serum total cholesterol, low-density lipoprotein cholesterol, and trygliceride were demonstrated in patients switching to RPV/FTC/TDF from a ritonavir-boosted protease inhibitor (PI/r)-based regimen, than in those who continued treatment with a PI/r regimen. Ritonavir 231-240 sex determining region Y Homo sapiens 220-223 23597294-7 2013 At a similar supersaturation ratio of ~1.2, ritonavir and celecoxib had slower normalized crystal growth rates (0.20 and 0.91 mg min(-1) m(-2), respectively), while the normalized crystal growth rate of efavirenz was significantly higher (2.97 mg min(-1) m(-2)), resulting in lower levels of crystal growth inhibition by the polymers for efavirenz. Ritonavir 44-53 CD59 molecule (CD59 blood group) Homo sapiens 247-253 23589366-0 2013 Time-dependent interaction of ritonavir in chronic use: the power balance between inhibition and induction of P-glycoprotein and cytochrome P450 3A. Ritonavir 30-39 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 110-124 23364475-9 2013 Although not statistically significant, a trend was observed towards lower maraviroc, darunavir and ritonavir concentrations in period 2 versus period 1; total maraviroc exposure was 3579 ng h/mL (95% CI: 2983-4294) and 2996 ng h/mL (95% CI: 2374-3782) in periods 1 and 2, respectively, and the GM ratio was 0.84 (95% CI: 0.67-1.05). Ritonavir 100-109 period circadian regulator 2 Homo sapiens 128-136 23381882-1 2013 We investigated the mechanisms of ritonavir-mediated enhancement effect on the pharmacokinetics of saquinavir using in vivo probes for CYP3A4 (midazolam), p-glycoprotein (fexofenadine), and OATP1B1 (pravastatin) following oral micro/small dosing. Ritonavir 34-43 solute carrier organic anion transporter family member 1B1 Homo sapiens 190-197 23589366-1 2013 Ritonavir (RTV) is not only an inhibitor but also an immunoreactive inducer of both P-glycoprotein (Pgp) and cytochrome P450 (CYP) 3A in terms of its chronic use. Ritonavir 0-9 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 84-98 23589366-1 2013 Ritonavir (RTV) is not only an inhibitor but also an immunoreactive inducer of both P-glycoprotein (Pgp) and cytochrome P450 (CYP) 3A in terms of its chronic use. Ritonavir 0-9 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 100-103 23589366-1 2013 Ritonavir (RTV) is not only an inhibitor but also an immunoreactive inducer of both P-glycoprotein (Pgp) and cytochrome P450 (CYP) 3A in terms of its chronic use. Ritonavir 0-9 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 109-133 23589366-1 2013 Ritonavir (RTV) is not only an inhibitor but also an immunoreactive inducer of both P-glycoprotein (Pgp) and cytochrome P450 (CYP) 3A in terms of its chronic use. Ritonavir 11-14 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 84-98 23589366-1 2013 Ritonavir (RTV) is not only an inhibitor but also an immunoreactive inducer of both P-glycoprotein (Pgp) and cytochrome P450 (CYP) 3A in terms of its chronic use. Ritonavir 11-14 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 100-103 23589366-1 2013 Ritonavir (RTV) is not only an inhibitor but also an immunoreactive inducer of both P-glycoprotein (Pgp) and cytochrome P450 (CYP) 3A in terms of its chronic use. Ritonavir 11-14 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 109-133 23589366-2 2013 The aim of present study was to test the hypothesis that the power balance between inhibition effects of RTV and induced activities of Pgp and CYP3A depends on the time after last RTV treatment (TimeR) in the chronic use of RTV; rhodamine 123 (Rho) and midazolam (MDZ) were administered at predetermined TimeR to rats pretreated with RTV for 7 days. Ritonavir 180-183 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 135-138 23589366-2 2013 The aim of present study was to test the hypothesis that the power balance between inhibition effects of RTV and induced activities of Pgp and CYP3A depends on the time after last RTV treatment (TimeR) in the chronic use of RTV; rhodamine 123 (Rho) and midazolam (MDZ) were administered at predetermined TimeR to rats pretreated with RTV for 7 days. Ritonavir 180-183 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 143-148 23589366-2 2013 The aim of present study was to test the hypothesis that the power balance between inhibition effects of RTV and induced activities of Pgp and CYP3A depends on the time after last RTV treatment (TimeR) in the chronic use of RTV; rhodamine 123 (Rho) and midazolam (MDZ) were administered at predetermined TimeR to rats pretreated with RTV for 7 days. Ritonavir 180-183 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 135-138 23589366-2 2013 The aim of present study was to test the hypothesis that the power balance between inhibition effects of RTV and induced activities of Pgp and CYP3A depends on the time after last RTV treatment (TimeR) in the chronic use of RTV; rhodamine 123 (Rho) and midazolam (MDZ) were administered at predetermined TimeR to rats pretreated with RTV for 7 days. Ritonavir 180-183 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 143-148 23090875-3 2013 Unlike paclitaxel, docetaxel is extensively metabolized by CYP3A4 and its oral bioavailability can be enhanced in mice and humans by coadministration of the potent CYP3A inhibitor ritonavir. Ritonavir 180-189 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 59-65 23633271-0 2013 Efficacy of the CCR5 antagonist maraviroc in reducing early, ritonavir-induced atherogenesis and advanced plaque progression in mice. Ritonavir 61-70 chemokine (C-C motif) receptor 5 Mus musculus 16-20 23633271-4 2013 METHODS AND RESULTS: Exposure to maraviroc limits the evolution and associated systemic inflammation of ritonavir-induced atherosclerotic in ApoE(-/-) mice and inhibits plaques development in a late model of atherosclerosis in which dyslipidemia plays the main pathogenic role. Ritonavir 104-113 apolipoprotein E Mus musculus 141-145 23633271-5 2013 In ritonavir-treated mice, maraviroc reduced plaque areas and macrophage infiltration; downregulated the local expression of vascular cell adhesion molecule-1, intercellular adhesion molecule-1, monocyte chemoattractant protein-1, and interleukin-17A; and reduced tumor necrosis factor-alpha and RANTES (regulated on activation, normal T cell expressed, and secreted). Ritonavir 3-12 vascular cell adhesion molecule 1 Mus musculus 125-193 23633271-5 2013 In ritonavir-treated mice, maraviroc reduced plaque areas and macrophage infiltration; downregulated the local expression of vascular cell adhesion molecule-1, intercellular adhesion molecule-1, monocyte chemoattractant protein-1, and interleukin-17A; and reduced tumor necrosis factor-alpha and RANTES (regulated on activation, normal T cell expressed, and secreted). Ritonavir 3-12 interleukin 17A Mus musculus 235-250 23633271-5 2013 In ritonavir-treated mice, maraviroc reduced plaque areas and macrophage infiltration; downregulated the local expression of vascular cell adhesion molecule-1, intercellular adhesion molecule-1, monocyte chemoattractant protein-1, and interleukin-17A; and reduced tumor necrosis factor-alpha and RANTES (regulated on activation, normal T cell expressed, and secreted). Ritonavir 3-12 tumor necrosis factor Mus musculus 264-291 23633271-5 2013 In ritonavir-treated mice, maraviroc reduced plaque areas and macrophage infiltration; downregulated the local expression of vascular cell adhesion molecule-1, intercellular adhesion molecule-1, monocyte chemoattractant protein-1, and interleukin-17A; and reduced tumor necrosis factor-alpha and RANTES (regulated on activation, normal T cell expressed, and secreted). Ritonavir 3-12 chemokine (C-C motif) ligand 5 Mus musculus 296-302 23090875-3 2013 Unlike paclitaxel, docetaxel is extensively metabolized by CYP3A4 and its oral bioavailability can be enhanced in mice and humans by coadministration of the potent CYP3A inhibitor ritonavir. Ritonavir 180-189 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 59-64 23090875-11 2013 Coadministered ritonavir inhibited Cyp3a-mediated metabolism, but not P-gp-mediated transport of paclitaxel. Ritonavir 15-24 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 35-40 23090875-12 2013 Owing to the dominant effect of P-gp, ritonavir enhanced only paclitaxel plasma concentrations in P-gp-deficient mice. Ritonavir 38-47 phosphoglycolate phosphatase Mus musculus 98-102 23090875-14 2013 Accordingly, ritonavir could enhance the oral bioavailability of paclitaxel in CYP3A4-humanized mice, despite the fact that these mice are P-gp-proficient. Ritonavir 13-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 79-85 23090875-15 2013 Our results show that CYP3A4 inhibition most likely underlies the boosting effect of ritonavir on oral paclitaxel bioavailability in humans. Ritonavir 85-94 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 22-28 23586711-0 2013 Pyridine-substituted desoxyritonavir is a more potent inhibitor of cytochrome P450 3A4 than ritonavir. Ritonavir 27-36 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 67-86 23586711-1 2013 Utilization of the cytochrome P450 3A4 (CYP3A4) inhibitor ritonavir as a pharmacoenhancer for anti-HIV drugs revolutionized the treatment of HIV infection. Ritonavir 58-67 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 19-38 23586711-1 2013 Utilization of the cytochrome P450 3A4 (CYP3A4) inhibitor ritonavir as a pharmacoenhancer for anti-HIV drugs revolutionized the treatment of HIV infection. Ritonavir 58-67 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 40-46 23586711-2 2013 However, owing to ritonavir-related complications, there is a need for development of new CYP3A4 inhibitors with improved pharmacochemical properties, which requires a full understanding of the CYP3A4 inactivation mechanisms and the unraveling of possible inhibitor binding modes. Ritonavir 18-27 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 90-96 23586711-5 2013 Additionally, Ser119 was identified as a key residue assisting binding of ritonavir-like inhibitors, which emphasizes the importance of polar interactions in the CYP3A4-ligand association. Ritonavir 74-83 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 162-168 23503447-1 2013 OBJECTIVE: Among HIV-positive patients prescribed ritonavir-boosted lopinavir, SLCO1B1 521T C (rs4149056) is associated with increased plasma lopinavir exposure. Ritonavir 50-59 solute carrier organic anion transporter family member 1B1 Homo sapiens 79-86 23403426-2 2013 GRL-0519 blocked the infectivity and replication of HIV-1NL4-3 variants selected by up to a 5 muM concentration of ritonavir, lopinavir, or atazanavir (EC50, 0.0028 to 0.0033 muM). Ritonavir 115-124 nuclear receptor subfamily 3 group C member 1 Homo sapiens 0-3 23201460-8 2013 Disulfiram, used to treat alcoholism for half-a century now, is a potent ALDH inhibitor and the old anti-viral drug ritonavir inhibits HSP90. Ritonavir 116-125 heat shock protein 90 alpha family class A member 1 Homo sapiens 135-140 23531330-2 2013 In addition to its antiviral effect, Ritonavir directly inhibits the insulin-regulated glucose transporter GLUT4 and blocks glucose entry into fat and muscle cells. Ritonavir 37-46 solute carrier family 2 (facilitated glucose transporter), member 4 Mus musculus 107-112 23341120-5 2013 Half-maximal effects were observed for ritonavir in a concentration of 3 muM. Ritonavir 39-48 latexin Homo sapiens 73-76 23341120-6 2013 The ritonavir-induced stimulated GSH export from astrocytes was completely prevented by MK571, an inhibitor of the multidrug resistance protein 1. Ritonavir 4-13 ATP binding cassette subfamily B member 1 Homo sapiens 115-145 23531330-10 2013 On the other hand, administration of ritonavir, a nonspecific GLUT inhibitor has been found to exhibit complete protection as observed by normalisation of heart/body weight ratio, serum markers, antioxidant enzymes activities and histopathological alterations. Ritonavir 37-46 solute carrier family 1 (glial high affinity glutamate transporter), member 3 Mus musculus 62-66 23531330-12 2013 CONCLUSIONS: Our study concluded that ritonavir, a nonspecific GLUT inhibitors showed complete protection in catecholamine induced myocardial necrosis. Ritonavir 38-47 solute carrier family 1 (glial high affinity glutamate transporter), member 3 Mus musculus 63-67 23148286-0 2013 Impact of UGT1A1 Gilbert variant on discontinuation of ritonavir-boosted atazanavir in AIDS Clinical Trials Group Study A5202. Ritonavir 55-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 10-16 22900583-2 2013 Ritonavir is a potent CYP3A inhibitor and was shown in vitro as a CYP2B6 inhibitor. Ritonavir 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-72 22900583-9 2013 Midazolam metabolic ratio (MR) dramatically decreased in presence of ritonavir (6.7 +- 2.6 versus 0.13 +- 0.07) reflecting an almost complete inhibition of CYP3A4, whereas omeprazole, flurbiprofen and bupropion MR were not affected. Ritonavir 69-78 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 156-162 22900583-10 2013 These data demonstrate that ritonavir is able to block prasugrel CYP3A4 bioactivation. Ritonavir 28-37 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 65-71 23165478-4 2013 The adverse immunostimulatory responses were abrogated by selective B cell-targeted delivery and early endosomal compartmentalization of G3139-encapsulated RIT-INPs, resulting in reduced NF-kappaB activation, robust Bcl-2 down-regulation, and enhanced sensitivity to fludarabine-induced cytotoxicity. Ritonavir 156-159 BCL2 apoptosis regulator Homo sapiens 216-221 23234359-11 2013 Preincubation of THLE-3A4 cells with the cytochrome P450 3A4 inhibitor ritonavir blocked the selective toxicity of rimonabant to these cells. Ritonavir 71-80 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 41-60 23032410-12 2013 CONCLUSION: Despite full viral load-suppression in blood and CSF, antiretroviral monotherapy with lopinavir/ritonavir can raise CSF levels of S100B, suggesting astrocytic damage. Ritonavir 108-117 S100 calcium binding protein B Homo sapiens 142-147 23091188-2 2013 Antiretroviral protease inhibitors lopinavir (LPV) and ritonavir (RTV) are reported BSEP inhibitors. Ritonavir 55-64 ATP binding cassette subfamily B member 11 Rattus norvegicus 84-88 23802035-0 2013 Putting the Rit in cellular resistance: Rit, p38 MAPK and oxidative stress. Ritonavir 12-15 mitogen-activated protein kinase 14 Mus musculus 45-53 23802035-8 2013 Together, our studies establish Rit as the central regulator of an evolutionarily conserved, p38-dependent signaling cascade that functions as a critical survival mechanism in response to stress. Ritonavir 32-35 mitogen-activated protein kinase 14 Mus musculus 93-96 23091188-2 2013 Antiretroviral protease inhibitors lopinavir (LPV) and ritonavir (RTV) are reported BSEP inhibitors. Ritonavir 66-69 ATP binding cassette subfamily B member 11 Rattus norvegicus 84-88 23349724-1 2013 OBJECTIVE: In the ANRS 139 TRIO trial, the use of 3 new active drugs (raltegravir, etravirine, and darunavir/ritonavir), resulted in a potent and sustained inhibition of viral replication in multidrug-resistant treatment-experienced patients. Ritonavir 109-118 trio Rho guanine nucleotide exchange factor Homo sapiens 27-31 23548184-7 2013 Ritonavir exhibits strong inhibition of hepatic enzyme CYP 3A4, which is part of the major metabolic pathway of most glucocorticoids. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 55-62 23533630-8 2013 We also identified through the use of primary adipocytes of CHOP(-/-) mice that CHOP, the major transcriptional factor of the ER stress signaling pathway, is involved in lopinavir/ritonavir-induced inhibition of cell differentiation in adipocytes. Ritonavir 180-189 DNA-damage inducible transcript 3 Mus musculus 60-64 23533630-8 2013 We also identified through the use of primary adipocytes of CHOP(-/-) mice that CHOP, the major transcriptional factor of the ER stress signaling pathway, is involved in lopinavir/ritonavir-induced inhibition of cell differentiation in adipocytes. Ritonavir 180-189 DNA-damage inducible transcript 3 Mus musculus 80-84 23061604-10 2012 However, the low frequency of the TAM-1 pathway in our cohort data suggests that these patients should respond well to second-line regimens containing a ritonavir-boosted protease inhibitor. Ritonavir 153-162 stromal interaction molecule 1 Homo sapiens 34-39 23843142-1 2013 OBJECTIVES: To analyse the progression of liver fibrosis as measured by elastography and biochemical testing in prisoninmates co-infected by HIV and HCVwho started on ritonavir-boosted protease inhibitor (PI) therapy. Ritonavir 167-176 serpin family A member 13, pseudogene Homo sapiens 185-204 23843142-1 2013 OBJECTIVES: To analyse the progression of liver fibrosis as measured by elastography and biochemical testing in prisoninmates co-infected by HIV and HCVwho started on ritonavir-boosted protease inhibitor (PI) therapy. Ritonavir 167-176 serpin family A member 13, pseudogene Homo sapiens 205-207 23115227-1 2012 OBJECTIVE: To report a probable drug interaction between the antiretroviral TRIO regimen (ritonavir-boosted darunavir, etravirine, and raltegravir) and warfarin in an HIV-infected patient. Ritonavir 90-99 trio Rho guanine nucleotide exchange factor Homo sapiens 76-80 23017355-1 2012 OBJECTIVE: To analyze the effect of switching the ritonavir-boosted protease inhibitor (PI/r) in a stable combined antiretroviral therapy (cART) regimen to raltegravir on low-density lipoprotein (LDL) particles, and lipoprotein-associated phospholipase A2 (Lp-PLA2). Ritonavir 50-59 phospholipase A2 group VII Homo sapiens 216-255 22381043-5 2012 The potent CYP3A4 inhibitor ritonavir alters the sequential metabolism of tilidine, substantially reducing the partial metabolic clearances of tilidine to nortilidine and nortilidine to bisnortilidine, which increases the nortilidine exposure twofold. Ritonavir 28-37 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 11-17 22381043-14 2012 CONCLUSIONS: The sequential metabolism of tilidine is inhibited by the potent CYP3A4 inhibitor, ritonavir, independent of the CYP2C19 genotype, with a twofold increase in the exposure of the active nortilidine. Ritonavir 96-105 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 78-84 23017355-1 2012 OBJECTIVE: To analyze the effect of switching the ritonavir-boosted protease inhibitor (PI/r) in a stable combined antiretroviral therapy (cART) regimen to raltegravir on low-density lipoprotein (LDL) particles, and lipoprotein-associated phospholipase A2 (Lp-PLA2). Ritonavir 50-59 phospholipase A2 group VII Homo sapiens 257-264 22174437-0 2012 Influence of SLCO1B1 polymorphisms on the drug-drug interaction between darunavir/ritonavir and pravastatin. Ritonavir 82-91 solute carrier organic anion transporter family member 1B1 Homo sapiens 13-20 22174437-8 2012 In sum, the influence of SLCO1B1*15 and *17 haplotypes on pravastatin pharmacokinetics was maintained in the presence of darunavir/ritonavir. Ritonavir 131-140 solute carrier organic anion transporter family member 1B1 Homo sapiens 25-32 22562466-2 2012 SS1P is a RIT that targets mesothelin on the surface of cancer cells and is being evaluated in patients with mesothelioma. Ritonavir 10-13 mesothelin Homo sapiens 27-37 22809809-9 2012 Ritonavir resulted in a decrease in CYP3A/5 activity, yet a concomitant increase in mRNA and protein levels of CYP3A4. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 36-43 22809809-9 2012 Ritonavir resulted in a decrease in CYP3A/5 activity, yet a concomitant increase in mRNA and protein levels of CYP3A4. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 111-117 22308076-2 2012 Ritonavir (RTV) is a known inhibitor of both P-gp and CYP3A and is co-administered with LPV in anti-HIV therapy. Ritonavir 0-9 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 54-59 22308076-2 2012 Ritonavir (RTV) is a known inhibitor of both P-gp and CYP3A and is co-administered with LPV in anti-HIV therapy. Ritonavir 11-14 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 54-59 22661571-6 2012 After ritonavir removal, a reduction was observed in total bilirubin (from 4.09 to 1.82 mg/dL; P < 0.001), gamma-glutamyl transpeptidase (P = 0.015), triglycerides (P = 0.03) and total cholesterol (P = 0.05). Ritonavir 6-15 inactive glutathione hydrolase 2 Homo sapiens 110-139 21719718-5 2012 The lopinavir/ritonavir dose of 500/125 mg bid administered with efavirenz most closely approximates the pharmacokinetic exposure of lopinavir/ritonavir 400/100 mg bid administered alone. Ritonavir 14-23 BH3 interacting domain death agonist Homo sapiens 43-46 21719718-5 2012 The lopinavir/ritonavir dose of 500/125 mg bid administered with efavirenz most closely approximates the pharmacokinetic exposure of lopinavir/ritonavir 400/100 mg bid administered alone. Ritonavir 14-23 BH3 interacting domain death agonist Homo sapiens 164-167 21719718-5 2012 The lopinavir/ritonavir dose of 500/125 mg bid administered with efavirenz most closely approximates the pharmacokinetic exposure of lopinavir/ritonavir 400/100 mg bid administered alone. Ritonavir 143-152 BH3 interacting domain death agonist Homo sapiens 43-46 22410611-1 2012 Ritonavir is a HIV protease inhibitor that also potently inactivates cytochrome P450 3A4 (CYP3A4), a major human drug-metabolizing enzyme. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 69-88 21826404-5 2012 Atazanavir, ritonavir, and saquinavir modestly inhibited of Abeta degradation, while lopinavir, nelfinavir, and ritonavir enhanced secretion of undigested Abeta after phagocytosis. Ritonavir 12-21 amyloid beta precursor protein Homo sapiens 60-65 21826404-5 2012 Atazanavir, ritonavir, and saquinavir modestly inhibited of Abeta degradation, while lopinavir, nelfinavir, and ritonavir enhanced secretion of undigested Abeta after phagocytosis. Ritonavir 112-121 amyloid beta precursor protein Homo sapiens 155-160 21826404-6 2012 Lopinavir, nelfinavir, ritonavir, and saquinavir inhibited endogenous Abeta40 production from primary cultured human cortical neurons, which were associated with reduction in Beta-site APP Converting Enzyme 1 (BACE1) and gamma-secretase enzyme activities. Ritonavir 23-32 beta-secretase 1 Homo sapiens 175-208 21826404-6 2012 Lopinavir, nelfinavir, ritonavir, and saquinavir inhibited endogenous Abeta40 production from primary cultured human cortical neurons, which were associated with reduction in Beta-site APP Converting Enzyme 1 (BACE1) and gamma-secretase enzyme activities. Ritonavir 23-32 beta-secretase 1 Homo sapiens 210-215 22470109-0 2012 17-Allylamino-17-demethoxygeldanamycin and ritonavir inhibit renal cancer growth by inhibiting the expression of heat shock factor-1. Ritonavir 43-52 heat shock transcription factor 1 Homo sapiens 113-132 22452979-6 2012 As proof of principle regarding the therapeutic potential of GLUT-targeted compounds, we include evidence of the antimyeloma effects elicited against both cell lines and primary cells by the FDA-approved HIV protease inhibitor ritonavir, which exerts a selective off-target inhibitory effect on GLUT4. Ritonavir 227-236 solute carrier family 2 member 1 Homo sapiens 61-65 22452979-6 2012 As proof of principle regarding the therapeutic potential of GLUT-targeted compounds, we include evidence of the antimyeloma effects elicited against both cell lines and primary cells by the FDA-approved HIV protease inhibitor ritonavir, which exerts a selective off-target inhibitory effect on GLUT4. Ritonavir 227-236 solute carrier family 2 member 4 Homo sapiens 295-300 22410611-1 2012 Ritonavir is a HIV protease inhibitor that also potently inactivates cytochrome P450 3A4 (CYP3A4), a major human drug-metabolizing enzyme. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 90-96 22410611-2 2012 To better understand the mechanism of ligand binding and to find strategies for improvement of the inhibitory potency of ritonavir, currently administered to enhance pharmacokinetics of other anti-HIV drugs that are quickly metabolized by CYP3A4, we compared the manner of CYP3A4 interaction with the drug and two analogs lacking either the heme-ligating thiazole nitrogen or the entire thiazole group. Ritonavir 121-130 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 239-245 22410611-2 2012 To better understand the mechanism of ligand binding and to find strategies for improvement of the inhibitory potency of ritonavir, currently administered to enhance pharmacokinetics of other anti-HIV drugs that are quickly metabolized by CYP3A4, we compared the manner of CYP3A4 interaction with the drug and two analogs lacking either the heme-ligating thiazole nitrogen or the entire thiazole group. Ritonavir 121-130 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 273-279 22094027-2 2012 Herein, modeling studies with dirlotapide, a CYP3A4 substrate, indicated that a substantial conformational change of CYP3A4 was necessary to accommodate it within the active site cavity, which is in good agreement with a new published CYP3A4 ritonavir co-crystal structure. Ritonavir 242-251 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 45-51 22328106-0 2012 Hypercortisolism caused by ritonavir associated inhibition of CYP 3A4 under inhalative glucocorticoid therapy. Ritonavir 27-36 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 62-69 22328106-3 2012 The PI ritonavir is described as the most potent compound within these CYP3A4 inhibitors. Ritonavir 7-16 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 71-77 22297608-0 2012 Ritonavir-induced lipoatrophy and dyslipidaemia is reversed by the anti-inflammatory drug leflunomide in a PPAR-gamma-dependent manner. Ritonavir 0-9 peroxisome proliferator activated receptor gamma Homo sapiens 107-117 22297608-9 2012 Histopathology analysis shows that exposure to ritonavir causes inflammation of epididymal fat as demonstrated by dense leukocytes infiltration as well as by increased levels of proinflammatory mediators and reduced expression and activity of peroxisome proliferator-activated receptor-gamma (PPAR-gamma). Ritonavir 47-56 peroxisome proliferator activated receptor gamma Homo sapiens 243-291 22297608-9 2012 Histopathology analysis shows that exposure to ritonavir causes inflammation of epididymal fat as demonstrated by dense leukocytes infiltration as well as by increased levels of proinflammatory mediators and reduced expression and activity of peroxisome proliferator-activated receptor-gamma (PPAR-gamma). Ritonavir 47-56 peroxisome proliferator activated receptor gamma Homo sapiens 293-303 22161308-1 2012 PURPOSE: To predict and determine whether the protease inhibitors (PIs) nelfinavir, amprenavir, atazanavir, ritonavir, and saquinavir could serve as metabolic inhibitors of the human CES1 (hCES1) using both molecular modeling techniques and in vitro inhibition assays. Ritonavir 108-117 carboxylesterase 1 Homo sapiens 183-187 22161308-6 2012 Other PIs (amprenavir, atazanavir, ritonavir, saquinavir) were evaluated and also shown to be hCES1 inhibitors in vitro, although substantially less potent relative to nelfinavir. Ritonavir 35-44 carboxylesterase 1 Homo sapiens 94-99 22142846-0 2012 Noncanonical Wnt signaling promotes osteoclast differentiation and is facilitated by the human immunodeficiency virus protease inhibitor ritonavir. Ritonavir 137-146 Wnt family member 5A Homo sapiens 13-16 22142846-3 2012 While investigating bone mineral density loss in the setting of human immunodeficiency virus (HIV) infection and its treatment with the protease inhibitor ritonavir (RTV), we observed that RTV decreased nuclear localization of beta-catenin, critical to canonical Wnt signaling, in primary human and murine osteoclast precursors. Ritonavir 155-164 Wnt family member 5A Homo sapiens 263-266 22142846-3 2012 While investigating bone mineral density loss in the setting of human immunodeficiency virus (HIV) infection and its treatment with the protease inhibitor ritonavir (RTV), we observed that RTV decreased nuclear localization of beta-catenin, critical to canonical Wnt signaling, in primary human and murine osteoclast precursors. Ritonavir 189-192 Wnt family member 5A Homo sapiens 263-266 22142846-11 2012 This is the first demonstration of a direct role for Wnt signaling pathways and Ryk in regulation of osteoclast differentiation, and its modulation by a clinically important drug, ritonavir. Ritonavir 180-189 Wnt family member 5A Homo sapiens 53-56 22094027-2 2012 Herein, modeling studies with dirlotapide, a CYP3A4 substrate, indicated that a substantial conformational change of CYP3A4 was necessary to accommodate it within the active site cavity, which is in good agreement with a new published CYP3A4 ritonavir co-crystal structure. Ritonavir 242-251 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 117-123 22094027-2 2012 Herein, modeling studies with dirlotapide, a CYP3A4 substrate, indicated that a substantial conformational change of CYP3A4 was necessary to accommodate it within the active site cavity, which is in good agreement with a new published CYP3A4 ritonavir co-crystal structure. Ritonavir 242-251 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 117-123 21953914-0 2012 CYP3A4-mediated lopinavir bioactivation and its inhibition by ritonavir. Ritonavir 62-71 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-6 23189194-2 2012 METHODS: We studied patients from the Swiss HIV Cohort Study who failed cART with nucleoside reverse transcriptase inhibitors (NRTIs) and either a ritonavir-boosted PI (PI/r) or a non-nucleoside reverse transcriptase inhibitor (NNRTI). Ritonavir 147-156 pirin Homo sapiens 165-173 21930825-3 2011 Statistically significant induction of CYP1A2 (2.1-, 2.9-, and 2.2-fold), CYP2B6 (1.8-, 2.4-, and 4-fold), and CYP2C9 (1.3-, 1.8-, and 2.6-fold) was observed after NFV, RTV, or RIF treatment, respectively (as expected, CYP2D6 was not induced). Ritonavir 169-172 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 39-45 22128044-6 2011 Saquinavir, therapeutically boosted with the potent CYP3A4 inhibitor ritonavir, was the only PI shown to be associated with significant QT interval prolongation in studies with healthy volunteers. Ritonavir 69-78 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 52-58 21968131-1 2011 Protease inhibitors (PIs), such as atazanavir sulfate and ritonavir, are used clinically to prevent the progression of HIV and are known to induce insulin resistance. Ritonavir 58-67 insulin Homo sapiens 147-154 21968131-4 2011 Atazanavir sulfate + ritonavir treatment for 48 h completely prevented insulin stimulation of glucose uptake (P>0.05). Ritonavir 21-30 insulin Homo sapiens 71-78 21968131-7 2011 Atazanavir sulfate treatment alone or in combination with ritonavir significantly increased JNK1/2 phosphorylation when compared to the control or ritonavir group (P<0.05) and this was accompanied by a rise (P<0.05) in AKT(Ser473) phosphorylation in the basal state. Ritonavir 58-67 mitogen-activated protein kinase 8 Homo sapiens 92-96 21930825-0 2011 Complex drug interactions of HIV protease inhibitors 2: in vivo induction and in vitro to in vivo correlation of induction of cytochrome P450 1A2, 2B6, and 2C9 by ritonavir or nelfinavir. Ritonavir 163-172 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 126-150 21930825-3 2011 Statistically significant induction of CYP1A2 (2.1-, 2.9-, and 2.2-fold), CYP2B6 (1.8-, 2.4-, and 4-fold), and CYP2C9 (1.3-, 1.8-, and 2.6-fold) was observed after NFV, RTV, or RIF treatment, respectively (as expected, CYP2D6 was not induced). Ritonavir 169-172 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-80 21930825-3 2011 Statistically significant induction of CYP1A2 (2.1-, 2.9-, and 2.2-fold), CYP2B6 (1.8-, 2.4-, and 4-fold), and CYP2C9 (1.3-, 1.8-, and 2.6-fold) was observed after NFV, RTV, or RIF treatment, respectively (as expected, CYP2D6 was not induced). Ritonavir 169-172 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 21550074-13 2011 In this in-vitro study, we found a potent inhibition of prasugrel bioactivation by ritonavir compared to the specific inhibitors of CYP3A and CYP2B6 due to the simultaneous inhibition of CYP2B6 and CYP3A by ritonavir. Ritonavir 83-92 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 187-193 21732894-3 2011 In the CASTLE study, patients with the most advanced HIV disease (CD4 cell count <50 cells/mm(3)) showed that 78% (45/58) vs. 58% (28/48) of the patients achieved HIV RNA <50 copies/mL in the intent-to-treat analysis at week 96 for atazanavir/ritonavir and lopinavir/ritonavir, respectively. Ritonavir 249-258 CD4 molecule Homo sapiens 66-69 21732894-3 2011 In the CASTLE study, patients with the most advanced HIV disease (CD4 cell count <50 cells/mm(3)) showed that 78% (45/58) vs. 58% (28/48) of the patients achieved HIV RNA <50 copies/mL in the intent-to-treat analysis at week 96 for atazanavir/ritonavir and lopinavir/ritonavir, respectively. Ritonavir 273-282 CD4 molecule Homo sapiens 66-69 21732894-6 2011 Also in this CD4 cell count stratum, grades 2-4 treatment-related adverse events occurred in 25% in the atazanavir/ritonavir group and in 43% of lopinavir/ritonavir group, and the rate was also higher than in the higher CD4 cell count strata within the lopinavir/ritonavir treatment group (range: 29-34%). Ritonavir 155-164 CD4 molecule Homo sapiens 13-16 21732894-6 2011 Also in this CD4 cell count stratum, grades 2-4 treatment-related adverse events occurred in 25% in the atazanavir/ritonavir group and in 43% of lopinavir/ritonavir group, and the rate was also higher than in the higher CD4 cell count strata within the lopinavir/ritonavir treatment group (range: 29-34%). Ritonavir 155-164 CD4 molecule Homo sapiens 13-16 21550074-13 2011 In this in-vitro study, we found a potent inhibition of prasugrel bioactivation by ritonavir compared to the specific inhibitors of CYP3A and CYP2B6 due to the simultaneous inhibition of CYP2B6 and CYP3A by ritonavir. Ritonavir 83-92 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 198-203 21550074-13 2011 In this in-vitro study, we found a potent inhibition of prasugrel bioactivation by ritonavir compared to the specific inhibitors of CYP3A and CYP2B6 due to the simultaneous inhibition of CYP2B6 and CYP3A by ritonavir. Ritonavir 207-216 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 132-137 21550074-13 2011 In this in-vitro study, we found a potent inhibition of prasugrel bioactivation by ritonavir compared to the specific inhibitors of CYP3A and CYP2B6 due to the simultaneous inhibition of CYP2B6 and CYP3A by ritonavir. Ritonavir 207-216 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 142-148 21550074-13 2011 In this in-vitro study, we found a potent inhibition of prasugrel bioactivation by ritonavir compared to the specific inhibitors of CYP3A and CYP2B6 due to the simultaneous inhibition of CYP2B6 and CYP3A by ritonavir. Ritonavir 207-216 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 187-193 21550074-13 2011 In this in-vitro study, we found a potent inhibition of prasugrel bioactivation by ritonavir compared to the specific inhibitors of CYP3A and CYP2B6 due to the simultaneous inhibition of CYP2B6 and CYP3A by ritonavir. Ritonavir 207-216 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 198-203 21854345-6 2011 However, the coadministration of Ritonavir with inhaled (or intranasal) corticosteroids may result in an increase in the plasma corticosteroid levels due to the potent CYP3A4 inhibition by Ritonavir. Ritonavir 33-42 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 168-174 21854345-6 2011 However, the coadministration of Ritonavir with inhaled (or intranasal) corticosteroids may result in an increase in the plasma corticosteroid levels due to the potent CYP3A4 inhibition by Ritonavir. Ritonavir 189-198 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 168-174 22010421-6 2011 PXR-ligands include a wide variety of pharmaceutical agents, such as antiepileptic drugs, taxol, rifampicin, and human immunodeficiency virus protease inhibitors such as ritonavir and saquinavir. Ritonavir 170-179 nuclear receptor subfamily 1 group I member 2 Homo sapiens 0-3 21175357-0 2011 Efficacy of Tat-conjugated ritonavir-loaded nanoparticles in reducing HIV-1 replication in monocyte-derived macrophages and cytocompatibility with macrophages and human neurons. Ritonavir 27-36 Tat Human immunodeficiency virus 1 12-15 21444726-8 2011 Together, our studies establish Rit as a central regulator of a p38 MAPK-dependent signaling cascade that functions as a critical cellular survival mechanism in response to stress. Ritonavir 32-35 mitogen-activated protein kinase 14 Homo sapiens 64-67 21480191-2 2011 Seven separate drug-drug interaction (DDI) studies were performed to elucidate the in vivo effects of concomitant treatment with colchicine and known inhibitors of cytochrome P450 3A4 (CYP3A4)/P-glycoprotein (cyclosporine, ketoconazole, ritonavir, clarithromycin, azithromycin, verapamil ER [extended release]), and diltiazem ER) on the pharmacokinetics of colchicine. Ritonavir 237-246 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 185-191 21692667-1 2011 Darunavir is a new-generation nonpeptidic HIV protease inhibitor (PI), used with low doses of ritonavir for pharmacologic enhancement (boosting). Ritonavir 94-103 serpin family A member 13, pseudogene Homo sapiens 46-64 21491455-0 2011 An integrated pharmacokinetic model for the influence of CYP3A4 expression on the in vivo disposition of lopinavir and its modulation by ritonavir. Ritonavir 137-146 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 57-63 21422984-0 2011 High-sensitivity C-reactive protein levels fall during statin therapy in HIV-infected patients receiving ritonavir-boosted protease inhibitors. Ritonavir 105-114 C-reactive protein Homo sapiens 17-35 21602280-5 2011 We identified the cellular heat shock protein 90 kDa alpha (cytosolic), class B member 1 (HSP90AB1) as a host factor that can rescue impaired replication of ritonavir-resistant HIV. Ritonavir 157-166 heat shock protein 90 alpha family class B member 1 Homo sapiens 27-88 21602280-5 2011 We identified the cellular heat shock protein 90 kDa alpha (cytosolic), class B member 1 (HSP90AB1) as a host factor that can rescue impaired replication of ritonavir-resistant HIV. Ritonavir 157-166 heat shock protein 90 alpha family class B member 1 Homo sapiens 90-98 21602280-6 2011 Moreover, we show that pharmacologic inhibition of HSP90AB1 with 17-(allylamino)-17-demethoxygeldanamycin (tanespimycin) has potent in vitro anti-HIV activity and that ritonavir-resistant HIV is hypersensitive to the drug. Ritonavir 168-177 heat shock protein 90 alpha family class B member 1 Homo sapiens 51-59 21515813-7 2011 Of a set of potential P-gp inhibitors, ketoconazole and ritonavir, but not clarithromycin or erythromycin, inhibited P-gp-mediated transport of rivaroxaban, with half-maximal inhibitory concentration values in the range of therapeutic plasma concentrations. Ritonavir 56-65 phosphoglycolate phosphatase Mus musculus 22-26 21515813-7 2011 Of a set of potential P-gp inhibitors, ketoconazole and ritonavir, but not clarithromycin or erythromycin, inhibited P-gp-mediated transport of rivaroxaban, with half-maximal inhibitory concentration values in the range of therapeutic plasma concentrations. Ritonavir 56-65 phosphoglycolate phosphatase Mus musculus 117-121 21491455-8 2011 Hepatic CYP3A4 related systemic clearance was inversely related to ritonavir exposure and not only hepatic but also intestinal CYP3A4 expression contributed to systemic clearance of lopinavir. Ritonavir 67-76 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 8-14 21282435-0 2011 Positive impact of HIV-1 gag cleavage site mutations on the virological response to darunavir boosted with ritonavir. Ritonavir 107-116 Pr55(Gag) Human immunodeficiency virus 1 25-28 21949754-0 2011 Polymorphism in Gag gene cleavage sites of HIV-1 non-B subtype and virological outcome of a first-line lopinavir/ritonavir single drug regimen. Ritonavir 113-122 Pr55(Gag) Human immunodeficiency virus 1 16-19 21348537-6 2011 The presence of a strong CYP3A inhibitor such as ritonavir or cobicistat renders the potential for increase in systemic exposures of CYP3A substrates coadministered with boosted elvitegravir. Ritonavir 49-58 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 25-30 21348537-6 2011 The presence of a strong CYP3A inhibitor such as ritonavir or cobicistat renders the potential for increase in systemic exposures of CYP3A substrates coadministered with boosted elvitegravir. Ritonavir 49-58 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 133-138 21239995-1 2011 BACKGROUND: In resource-limited settings, many patients, with no prior protease inhibitor (PI) treatment on a second-line, high genetic barrier, ritonavir-boosted PI-containing regimen have virologic failure. Ritonavir 145-154 serpin family A member 13, pseudogene Homo sapiens 163-165 20942777-0 2011 Interaction involving tadalafil and CYP3A4 inhibition by ritonavir. Ritonavir 57-66 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 36-42 21359201-11 2011 Exenatide also abrogated the detrimental effect of the GLUT4 antagonist ritonavir on survival in TG9 mice. Ritonavir 72-81 solute carrier family 2 (facilitated glucose transporter), member 4 Mus musculus 55-60 21148235-6 2011 Mean change in CD4 cell count from baseline to week 96 was 265 cells/mm(3) for women and 269 cells/mm(3) for men on atazanavir/ritonavir and 298 cells/mm(3) for women and 286 cells/mm(3) for men on lopinavir/ritonavir. Ritonavir 127-136 CD4 molecule Homo sapiens 15-18 21148235-6 2011 Mean change in CD4 cell count from baseline to week 96 was 265 cells/mm(3) for women and 269 cells/mm(3) for men on atazanavir/ritonavir and 298 cells/mm(3) for women and 286 cells/mm(3) for men on lopinavir/ritonavir. Ritonavir 208-217 CD4 molecule Homo sapiens 15-18 21980965-2 2011 This was due to atazanavir and ritonavir therapy for her HIV inhibiting the CYP3A4 hepatic enzyme resulting in accumulation of tacrolimus. Ritonavir 31-40 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 76-82 20963404-8 2011 Three subjects discontinued tipranavir/ritonavir treatment as a result of drug-related increases in ALT/AST, including one subject who experienced mild upper abdominal pain. Ritonavir 39-48 solute carrier family 17 member 5 Homo sapiens 104-107 20861742-6 2010 Addition of low-dose ritonavir enhanced levels of CYP promoter activity for several protease inhibitors. Ritonavir 21-30 peptidylprolyl isomerase G Homo sapiens 50-53 21811554-2 2011 There is insufficient evidence whether simplification to ritonavir boosted protease inhibitor (PI/r) monotherapy in virologically suppressed HIV-infected patients is effective and safe to reduce cART side effects and costs. Ritonavir 57-66 serpin family A member 13, pseudogene Homo sapiens 95-99 20864532-0 2010 Effects of the HIV protease inhibitor ritonavir on GLUT4 knock-out mice. Ritonavir 38-47 solute carrier family 2 (facilitated glucose transporter), member 4 Mus musculus 51-56 21054396-9 2010 Ritonavir, a known P-gp substrate, proved to be less toxic in the P-gp expressing cell line than in the nonexpressing cell line at the cell-exposed concentrations and thus showed P-gp substrate properties. Ritonavir 0-9 phosphoglycolate phosphatase Homo sapiens 19-23 21054396-9 2010 Ritonavir, a known P-gp substrate, proved to be less toxic in the P-gp expressing cell line than in the nonexpressing cell line at the cell-exposed concentrations and thus showed P-gp substrate properties. Ritonavir 0-9 phosphoglycolate phosphatase Homo sapiens 66-70 21054396-9 2010 Ritonavir, a known P-gp substrate, proved to be less toxic in the P-gp expressing cell line than in the nonexpressing cell line at the cell-exposed concentrations and thus showed P-gp substrate properties. Ritonavir 0-9 phosphoglycolate phosphatase Homo sapiens 66-70 21054396-11 2010 CONCLUSIONS: The novel compounds have been shown to be prospective AIDS therapeutics, acting as effective and nontoxic P-gp inhibitors compared with ritonavir, which is a known P-gp inhibitor with unfavourable toxic and P-gp substrate properties. Ritonavir 149-158 phosphoglycolate phosphatase Homo sapiens 177-181 21054396-11 2010 CONCLUSIONS: The novel compounds have been shown to be prospective AIDS therapeutics, acting as effective and nontoxic P-gp inhibitors compared with ritonavir, which is a known P-gp inhibitor with unfavourable toxic and P-gp substrate properties. Ritonavir 149-158 phosphoglycolate phosphatase Homo sapiens 177-181 20864532-11 2010 These data confirm that a primary effect of ritonavir on peripheral glucose disposal is mediated through direct inhibition of GLUT4 activity in vivo. Ritonavir 44-53 solute carrier family 2 (facilitated glucose transporter), member 4 Mus musculus 126-131 20595906-1 2010 BACKGROUND: Some HIV protease inhibitors (PIs), including full-dose ritonavir (800 mg) and ritonavir-boosted lopinavir, acutely induce insulin resistance in the absence of HIV infection and changes in body composition. Ritonavir 68-77 insulin Homo sapiens 135-142 20595906-5 2010 CONCLUSIONS: A single boosting dose of ritonavir does not alter insulin sensitivity, suggesting lopinavir is likely responsible for the induction of insulin resistance demonstrated in prior short-term studies of lopinavir/ritonavir. Ritonavir 39-48 insulin Homo sapiens 149-156 20978219-10 2010 DISCUSSION: The Horn Drug Interaction Probability Scale demonstrated a possible relationship between the increased aripiprazole concentration and coadministration of darunavir/ritonavir and duloxetine, which inhibit CYP3A4 and 2D6. Ritonavir 176-185 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 216-230 20595906-6 2010 There is a dose-dependent effect of ritonavir on insulin sensitivity. Ritonavir 36-45 insulin Homo sapiens 49-56 20595906-1 2010 BACKGROUND: Some HIV protease inhibitors (PIs), including full-dose ritonavir (800 mg) and ritonavir-boosted lopinavir, acutely induce insulin resistance in the absence of HIV infection and changes in body composition. Ritonavir 91-100 insulin Homo sapiens 135-142 20551242-0 2010 In situ intestinal perfusion in knockout mice demonstrates inhibition of intestinal p-glycoprotein by ritonavir causing increased darunavir absorption. Ritonavir 102-111 phosphoglycolate phosphatase Mus musculus 84-98 20937904-0 2010 Structure and mechanism of the complex between cytochrome P4503A4 and ritonavir. Ritonavir 70-79 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 47-65 20937904-1 2010 Ritonavir is a HIV protease inhibitor routinely prescribed to HIV patients that also potently inactivates cytochrome P4503A4 (CYP3A4), the major human drug-metabolizing enzyme. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 106-124 20937904-1 2010 Ritonavir is a HIV protease inhibitor routinely prescribed to HIV patients that also potently inactivates cytochrome P4503A4 (CYP3A4), the major human drug-metabolizing enzyme. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 126-132 20937904-2 2010 By inhibiting CYP3A4, ritonavir increases plasma concentrations of other anti-HIV drugs oxidized by CYP3A4 thereby improving clinical efficacy. Ritonavir 22-31 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 14-20 20937904-2 2010 By inhibiting CYP3A4, ritonavir increases plasma concentrations of other anti-HIV drugs oxidized by CYP3A4 thereby improving clinical efficacy. Ritonavir 22-31 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 100-106 20937904-4 2010 The available data are inconsistent and suggest that ritonavir acts as a mechanism-based, competitive or mixed competitive-noncompetitive CYP3A4 inactivator. Ritonavir 53-62 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 138-144 20937904-6 2010 Our results show that ritonavir is a type II ligand that perfectly fits into the CYP3A4 active site cavity and irreversibly binds to the heme iron via the thiazole nitrogen, which decreases the redox potential of the protein and precludes its reduction with the redox partner, cytochrome P450 reductase. Ritonavir 22-31 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 81-87 20949026-0 2010 The bile acid sensor FXR protects against dyslipidemia and aortic plaques development induced by the HIV protease inhibitor ritonavir in mice. Ritonavir 124-133 nuclear receptor subfamily 1, group H, member 4 Mus musculus 21-24 20949026-3 2010 METHODOLOGY/PRINCIPAL FINDINGS: Administration of the HIV PI ritonavir to wild type mice increased plasma triacylglycerols and cholesterol levels and this effect was exacerbated by dosing ritonavir to mice harbouring a disrupted FXR. Ritonavir 61-70 nuclear receptor subfamily 1, group H, member 4 Mus musculus 229-232 20949026-5 2010 Treating wild type mice with the FXR agonist (chenodeoxycholic acid, CDCA) protected against development of dyslipidemia induced by ritonavir. Ritonavir 132-141 nuclear receptor subfamily 1, group H, member 4 Mus musculus 33-36 20949026-6 2010 Administration of ritonavir to ApoE(-/-) mice, a strain that develop spontaneously atherosclerosis, increased the extent of aortic plaques without worsening the dyslipidemia. Ritonavir 18-27 apolipoprotein E Mus musculus 31-35 20949026-11 2010 Natural and synthetic FXR ligands reduced the nuclear translocation of SREBP1c caused by ritonavir. Ritonavir 89-98 nuclear receptor subfamily 1, group H, member 4 Mus musculus 22-25 20949026-12 2010 CONCLUSIONS/SIGNIFICANCE: Activation of the bile acid sensor FXR protects against dyslipidemia and atherosclerotic caused by ritonavir, a widely used HIV PI. Ritonavir 125-134 nuclear receptor subfamily 1, group H, member 4 Mus musculus 61-64 20949026-14 2010 FXR ligands might hold promise in the treatment dyslipidemia induced by ritonavir. Ritonavir 72-81 nuclear receptor subfamily 1, group H, member 4 Mus musculus 0-3 20551242-3 2010 It has been shown that darunavir and ritonavir are substrates of P-glycoprotein (P-gp). Ritonavir 37-46 phosphoglycolate phosphatase Mus musculus 65-79 20551242-3 2010 It has been shown that darunavir and ritonavir are substrates of P-glycoprotein (P-gp). Ritonavir 37-46 phosphoglycolate phosphatase Mus musculus 81-85 20551242-9 2010 However, this P-gp-mediated darunavir transport is inhibited when it is combined with ritonavir. Ritonavir 86-95 phosphoglycolate phosphatase Mus musculus 14-18 21351567-8 2010 The selective CYP3A4 inhibitors, ketoconazole, troleandomycin and ritonavir demonstrated significant inhibitory effects on CMDCK intestinal metabolism, which suggested that co-administration of CMDCK with potent CYP3A inhibitors, such as ritonavir, might decrease its intestinal metabolic clearance and subsequently improve its bioavailability in body. Ritonavir 66-75 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 14-20 20507927-7 2010 Several PIs potently inhibited OATP2B1-mediated transport in Caco-2 cells at clinically relevant IC(50) concentrations for ritonavir (0.93 microM), atazanavir (2.2 microM), lopinavir (1.7 microM), tipranavir (0.77 microM), and nelfinavir (2.2 microM). Ritonavir 123-132 solute carrier organic anion transporter family member 2B1 Homo sapiens 31-38 20653496-15 2010 Ritonavir, a potent CYP3A4 inhibitor, may inhibit corticosteroid degradation and increase its accumulation. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 20-26 20630573-9 2010 CONCLUSIONS: Combination therapy using SAHA and ritonavir inhibited the proliferation of renal cancer cells effectively, perhaps by inhibiting both histone deacetylase function and expression. Ritonavir 48-57 histone deacetylase 9 Homo sapiens 148-167 21351567-8 2010 The selective CYP3A4 inhibitors, ketoconazole, troleandomycin and ritonavir demonstrated significant inhibitory effects on CMDCK intestinal metabolism, which suggested that co-administration of CMDCK with potent CYP3A inhibitors, such as ritonavir, might decrease its intestinal metabolic clearance and subsequently improve its bioavailability in body. Ritonavir 238-247 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 14-20 20551216-0 2010 Protease inhibitors atazanavir, lopinavir and ritonavir are potent blockers, but poor substrates, of ABC transporters in a broad panel of ABC transporter-overexpressing cell lines. Ritonavir 46-55 ATP binding cassette subfamily B member 6 (Langereis blood group) Homo sapiens 101-104 20551216-0 2010 Protease inhibitors atazanavir, lopinavir and ritonavir are potent blockers, but poor substrates, of ABC transporters in a broad panel of ABC transporter-overexpressing cell lines. Ritonavir 46-55 ATP binding cassette subfamily B member 6 (Langereis blood group) Homo sapiens 138-141 20551216-8 2010 Cells overexpressing MDR1 P-gp, MRP3, MRP4 and MRP5 displayed low levels of resistance to atazanavir (RF: 1.3-1.7); MRP7- and MRP9-overexpressing cells to lopinavir (RF: 1.4-1.5); and MRP9-overexpressing cells to ritonavir (RF: 1.4). Ritonavir 213-222 ATP binding cassette subfamily C member 3 Homo sapiens 32-36 20551216-5 2010 RESULTS: Atazanavir, lopinavir and ritonavir were highly effective in reversing resistance against established substrates in cells overexpressing MDR1 P-gp and MRP1, and, to a lesser extent, BCRP. Ritonavir 35-44 ATP binding cassette subfamily B member 1 Homo sapiens 146-150 20551216-5 2010 RESULTS: Atazanavir, lopinavir and ritonavir were highly effective in reversing resistance against established substrates in cells overexpressing MDR1 P-gp and MRP1, and, to a lesser extent, BCRP. Ritonavir 35-44 phosphoglycolate phosphatase Homo sapiens 151-155 20551216-8 2010 Cells overexpressing MDR1 P-gp, MRP3, MRP4 and MRP5 displayed low levels of resistance to atazanavir (RF: 1.3-1.7); MRP7- and MRP9-overexpressing cells to lopinavir (RF: 1.4-1.5); and MRP9-overexpressing cells to ritonavir (RF: 1.4). Ritonavir 213-222 ATP binding cassette subfamily C member 5 Homo sapiens 47-51 20551216-5 2010 RESULTS: Atazanavir, lopinavir and ritonavir were highly effective in reversing resistance against established substrates in cells overexpressing MDR1 P-gp and MRP1, and, to a lesser extent, BCRP. Ritonavir 35-44 ATP binding cassette subfamily B member 1 Homo sapiens 160-164 20551216-8 2010 Cells overexpressing MDR1 P-gp, MRP3, MRP4 and MRP5 displayed low levels of resistance to atazanavir (RF: 1.3-1.7); MRP7- and MRP9-overexpressing cells to lopinavir (RF: 1.4-1.5); and MRP9-overexpressing cells to ritonavir (RF: 1.4). Ritonavir 213-222 ATP binding cassette subfamily B member 1 Homo sapiens 21-25 20551216-8 2010 Cells overexpressing MDR1 P-gp, MRP3, MRP4 and MRP5 displayed low levels of resistance to atazanavir (RF: 1.3-1.7); MRP7- and MRP9-overexpressing cells to lopinavir (RF: 1.4-1.5); and MRP9-overexpressing cells to ritonavir (RF: 1.4). Ritonavir 213-222 ATP binding cassette subfamily C member 12 Homo sapiens 126-130 20375850-0 2010 Association between lipodystrophy and leptin in human immunodeficiency virus-1-infected children receiving lopinavir/ritonavir-based therapy. Ritonavir 117-126 leptin Homo sapiens 38-44 19835489-2 2010 Voriconazole induction, including the utilization of voriconazole therapeutic drug monitoring in both serum and CSF, with transition to voriconazole plus interferon-gamma (IFN-gamma) was successfully used in a patient receiving antiretroviral therapy with abacavir/lamivudine and lopinavir/ritonavir. Ritonavir 290-299 interferon gamma Homo sapiens 172-181 20467943-9 2010 In contrast to LPV/RTV, ATV/RTV treatment was associated with earlier and more positive improvements in QoL scores across CD4 sub-groups. Ritonavir 28-31 CD4 molecule Homo sapiens 122-125 18781283-6 2010 RESULTS: The patient after 10 years of antiretroviral therapy with 11 different regimens unable to produce full virological suppression and with a rapidly declining CD4 count, achieved a successful virological outcome with a scheme containing Tipranavir boosted with low dose of ritonavir. Ritonavir 279-288 CD4 molecule Homo sapiens 165-168 20573082-2 2010 Inhibition of the CYP3A4 enzymes by ritonavir resulted in increased oral bioavailability. Ritonavir 36-45 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 18-24 20590614-3 2010 We also evaluated whether ritonavir increases lopinavir oral bioavailability by inhibition of CYP3A, ABCB1 and/or ABCC2. Ritonavir 26-35 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 94-99 20590614-3 2010 We also evaluated whether ritonavir increases lopinavir oral bioavailability by inhibition of CYP3A, ABCB1 and/or ABCC2. Ritonavir 26-35 ATP-binding cassette, sub-family B (MDR/TAP), member 1B Mus musculus 101-106 20590614-11 2010 Ritonavir markedly increased lopinavir AUC(oral) in all CYP3A-containing mouse strains. Ritonavir 0-9 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 56-61 20590614-14 2010 Ritonavir increased lopinavir bioavailability primarily by inhibiting CYP3A. Ritonavir 0-9 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 70-75 20147896-0 2010 A phenotype-genotype approach to predicting CYP450 and P-glycoprotein drug interactions with the mixed inhibitor/inducer tipranavir/ritonavir. Ritonavir 132-141 ATP binding cassette subfamily B member 1 Homo sapiens 55-69 20228169-8 2010 Ritonavir-treated adipocytes expressed significantly more ATGL mRNA (P < 0.05) and protein (P < 0.05). Ritonavir 0-9 patatin like phospholipase domain containing 2 Homo sapiens 58-62 20228169-11 2010 In 3T3-L1 adipocytes, long-term ritonavir exposure perturbs FA metabolism by increasing ATGL-mediated partial TAG hydrolysis, thus increasing FA efflux, and leads to compensatory increases in FA reesterification with glycerol and acylglycerols. Ritonavir 32-41 patatin like phospholipase domain containing 2 Homo sapiens 88-92 20090545-0 2010 Prospective, randomized, open label trial of Efavirenz vs Lopinavir/Ritonavir in HIV+ treatment-naive subjects with CD4+<200 cell/mm3 in Mexico. Ritonavir 68-77 CD4 molecule Homo sapiens 116-119 20219970-6 2010 Importantly, Src inhibition, or the lack of functional Trk receptors, was found to inhibit PACAP-mediated Rit activation, whereas constitutively active Src alone was sufficient to stimulate Rit-guanosine triphosphate levels. Ritonavir 106-109 SRC proto-oncogene, non-receptor tyrosine kinase Homo sapiens 13-16 20219970-6 2010 Importantly, Src inhibition, or the lack of functional Trk receptors, was found to inhibit PACAP-mediated Rit activation, whereas constitutively active Src alone was sufficient to stimulate Rit-guanosine triphosphate levels. Ritonavir 106-109 adenylate cyclase activating polypeptide 1 Homo sapiens 91-96 19851115-7 2010 Upon coadministration, elvitegravir and ritonavir PK were unaltered, but maraviroc exposures were 2-fold to 4-fold higher presumably due to ritonavir-mediated CYP3A-/Pgp inhibition. Ritonavir 140-149 phosphoglycolate phosphatase Homo sapiens 166-169 20197013-0 2010 Effect of saquinavir/ritonavir on P-glycoprotein activity in healthy volunteers using digoxin as a probe. Ritonavir 21-30 ATP binding cassette subfamily B member 1 Homo sapiens 34-48 20197013-1 2010 BACKGROUND: Saquinavir and ritonavir, both human immunodeficiency virus-1 protease inhibitors, also inhibit the adenosine triphosphate-dependent efflux pump P-glycoprotein (P-gp), which is located at a variety of anatomic sites, including the human intestine. Ritonavir 27-36 ATP binding cassette subfamily B member 1 Homo sapiens 157-171 20197013-1 2010 BACKGROUND: Saquinavir and ritonavir, both human immunodeficiency virus-1 protease inhibitors, also inhibit the adenosine triphosphate-dependent efflux pump P-glycoprotein (P-gp), which is located at a variety of anatomic sites, including the human intestine. Ritonavir 27-36 ATP binding cassette subfamily B member 1 Homo sapiens 173-177 20197013-3 2010 This study investigated the inhibitory potential of multiple administrations of ritonavir-boosted saquinavir at the target therapeutic dose of 1,000 mg saquinavir/100 mg ritonavir twice daily on the pharmacokinetics of oral digoxin, a model P-gp substrate that is predominantly excreted as unchanged drug in the urine. Ritonavir 80-89 ATP binding cassette subfamily B member 1 Homo sapiens 241-245 20197013-18 2010 CONCLUSIONS: Pretreatment with saquinavir/ritonavir 1,000/100 mg twice daily increased digoxin exposure most likely via P-gp-inhibition. Ritonavir 42-51 ATP binding cassette subfamily B member 1 Homo sapiens 120-124 20238377-5 2010 In human hepatocytes, saquinavir, ritonavir and atazanavir were the most efficient inhibitors of ABCC2-mediated biliary excretion of CDF, whereas in rat hepatocytes indinavir, lopinavir and nelfinavir were the most efficient. Ritonavir 34-43 ATP binding cassette subfamily C member 2 Homo sapiens 97-102 19924124-0 2010 Effect of simultaneous induction and inhibition of CYP3A by St John"s Wort and ritonavir on CYP3A activity. Ritonavir 79-88 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 92-97 19924124-1 2010 We aimed to assess the effect of coadministration and withdrawal of a potent cytochrome P450 3A (CYP3A) inhibitor (ritonavir) and a potent CYP3A inducer (St John"s wort) on CYP3A enzyme activity in an open, fixed-sequence study design. Ritonavir 115-124 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 77-95 19924124-1 2010 We aimed to assess the effect of coadministration and withdrawal of a potent cytochrome P450 3A (CYP3A) inhibitor (ritonavir) and a potent CYP3A inducer (St John"s wort) on CYP3A enzyme activity in an open, fixed-sequence study design. Ritonavir 115-124 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 97-102 19898246-5 2010 RESULTS: Insulin-stimulated peripheral glucose disposal decreased by 25% after 3 months in patients on zidovudine/lamivudine/lopinavir/ritonavir (ZDV/3TC/LPV/r) (P < 0.001) and this decreased rate persisted thereafter, followed by a transient decrease in insulin-mediated inhibition of lipolysis. Ritonavir 135-144 insulin Homo sapiens 9-16 19898246-5 2010 RESULTS: Insulin-stimulated peripheral glucose disposal decreased by 25% after 3 months in patients on zidovudine/lamivudine/lopinavir/ritonavir (ZDV/3TC/LPV/r) (P < 0.001) and this decreased rate persisted thereafter, followed by a transient decrease in insulin-mediated inhibition of lipolysis. Ritonavir 135-144 insulin Homo sapiens 258-265 20122159-8 2010 We also evaluated the impact of T477A on the kinetics of intravirion Gag-Pol polyprotein processing of p51 downward arrowRNH cleavage site mutants using the protease inhibitor ritonavir. Ritonavir 176-185 Gag-Pol Human immunodeficiency virus 1 69-76 19732776-8 2010 CHOP silencing by specific small hairpin RNA prevented lopinavir- and ritonavir-induced barrier dysfunction in cultured intestinal epithelial cells, whereas CHOP(-)/(-) mice exhibited decreased mucosal injury after exposure to lopinavir and ritonavir. Ritonavir 70-79 DNA-damage inducible transcript 3 Mus musculus 0-4 19890203-3 2010 We studied the effects of the protease inhibitor combination lopinavir and ritonavir on insulin secretion, insulin sensitivity, and lipid metabolism in HIV-negative persons. Ritonavir 75-84 insulin Homo sapiens 88-95 20386083-0 2010 Darunavir/ritonavir and efavirenz exert differential effects on MRP1 transporter expression and function in healthy volunteers. Ritonavir 10-19 CD9 molecule Homo sapiens 64-68 20386083-2 2010 We studied MRP1 expression and function in healthy volunteers treated with darunavir/ritonavir and efavirenz. Ritonavir 85-94 CD9 molecule Homo sapiens 11-15 20386083-10 2010 MRP1 efflux function was increased after efavirenz administration (GMR 3.13, 95% CI 2.73-3.59; P<0.001) and darunavir/ritonavir plus efavirenz coadministration (GMR 4.35, 95% CI 3.35-5.68; P<0.001), but not after darunavir/ritonavir administration (GMR 1.06, 95% CI 0.80-1.42; P=0.42). Ritonavir 121-130 CD9 molecule Homo sapiens 0-4 20386083-10 2010 MRP1 efflux function was increased after efavirenz administration (GMR 3.13, 95% CI 2.73-3.59; P<0.001) and darunavir/ritonavir plus efavirenz coadministration (GMR 4.35, 95% CI 3.35-5.68; P<0.001), but not after darunavir/ritonavir administration (GMR 1.06, 95% CI 0.80-1.42; P=0.42). Ritonavir 229-238 CD9 molecule Homo sapiens 0-4 20386083-11 2010 CONCLUSIONS: Darunavir/ritonavir and efavirenz treatment exerted differential effects on MRP1 expression and function. Ritonavir 23-32 CD9 molecule Homo sapiens 89-93 20201776-4 2010 We determined the induction of CYP3A4 by EFV, RTV, ATV, EFV+RTV, EFV+ATV, EFV+RTV+ATV and rifampicin (RIF) employing primary human hepatocytes from 3 donors. Ritonavir 46-49 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 31-37 20201776-4 2010 We determined the induction of CYP3A4 by EFV, RTV, ATV, EFV+RTV, EFV+ATV, EFV+RTV+ATV and rifampicin (RIF) employing primary human hepatocytes from 3 donors. Ritonavir 60-63 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 31-37 20201776-4 2010 We determined the induction of CYP3A4 by EFV, RTV, ATV, EFV+RTV, EFV+ATV, EFV+RTV+ATV and rifampicin (RIF) employing primary human hepatocytes from 3 donors. Ritonavir 60-63 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 31-37 20201776-6 2010 CYP3A4 activity (testosterone-6beta-hydroxylation) was induced by EFV (3 fold) and RIF (4 fold), but was significantly suppressed in the presence of RTV and ATV. Ritonavir 149-152 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-6 20201776-8 2010 hPXR activation data in LS180 cells were consistent with the induction of transcripts and the estimated EC(50) values were 0.87 microM, 0.44 microM and 3.7 microM for RIF, RTV and EFV, respectively. Ritonavir 172-175 nuclear receptor subfamily 1 group I member 2 Homo sapiens 0-4 20201776-9 2010 However, in primary hepatocytes the net effect was suppression of EFV mediated CYP3A4 induction by RTV and ATV. Ritonavir 99-102 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 79-85 19732776-8 2010 CHOP silencing by specific small hairpin RNA prevented lopinavir- and ritonavir-induced barrier dysfunction in cultured intestinal epithelial cells, whereas CHOP(-)/(-) mice exhibited decreased mucosal injury after exposure to lopinavir and ritonavir. Ritonavir 241-250 DNA-damage inducible transcript 3 Mus musculus 0-4 19943026-4 2010 Among the protease inhibitors, ritonavir is the strongest inhibitor of CYP3A activity. Ritonavir 31-40 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 71-76 20610890-3 2010 The in vitro uptake of both drugs into HepG2 cells and precision cut rat liver slices significantly increased in the presence of Pgp and MRP-2 inhibitor ritonavir. Ritonavir 153-162 ATP binding cassette subfamily C member 2 Rattus norvegicus 137-142 21182340-1 2010 Darunavir boosted by low-dose ritonavir (DRV/r), at a daily dose of 600/100 mg twice a day (bid), has been shown to be superior to alternative highly active antiretroviral therapy (HAART) regimens for the management of treatment-experienced, HIV-infected adults in the phase IIb POWER trials and the phase III TITAN trial. Ritonavir 30-39 BH3 interacting domain death agonist Homo sapiens 92-95 21182349-1 2010 BACKGROUND: Using data from the phase IIb POWER trials, darunavir boosted with low-dose ritonavir (DRV/r; 600/100 mg twice daily; bid)-based highly active antiretroviral therapy (HAART) was shown to be significantly more efficacious and cost effective than other protease inhibitor (PI)-based therapy in highly treatment-experienced, HIV-1-infected adults. Ritonavir 88-97 BH3 interacting domain death agonist Homo sapiens 130-133 21182349-2 2010 Furthermore, in the phase III TITAN trial (TMC114-C214), DRV/r 600/100 mg bid-based HAART generated a superior 48-week virological response rate compared with standard-of-care lopinavir/ritonavir (LPV/r; 400/100 mg bid)-based therapy in treatment-experienced, lopinavir-naive patients, and in particular those with one or more International AIDS Society - USA (IAS-USA) primary PI resistance-associated mutations at baseline. Ritonavir 186-195 BH3 interacting domain death agonist Homo sapiens 74-77 21182351-2 2010 OBJECTIVES: To forecast the impact of the use of darunavir with low-dose ritonavir 600/100 mg twice a day (bid) in highly treatment-experienced, HIV-infected adults who have failed one or more protease inhibitor (PI)-containing regimen on the budget of the French Sickness Fund (French healthcare system) over a 3-year time horizon. Ritonavir 73-82 BH3 interacting domain death agonist Homo sapiens 107-110 20002087-4 2009 * Therefore ritonavir is an appropriate positive control inhibitor for clinical drug interaction studies involving CYP3A substrates. Ritonavir 12-21 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 115-120 20002087-11 2009 CONCLUSIONS: Low-dose ritonavir produces extensive CYP3A inhibition exceeding that of ketoconazole (typically 10- to 15-fold midazolam AUC enhancement), and is a suitable positive control index inhibitor for drug-drug interaction studies. Ritonavir 22-31 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 51-56 19710077-14 2009 Ritonavir lessened CYP3A5 expressor effects. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 19-25 19757141-9 2009 Ritonavir is a potent CYP3A5 isoenzyme and P-gp inhibitor. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 22-28 19757141-9 2009 Ritonavir is a potent CYP3A5 isoenzyme and P-gp inhibitor. Ritonavir 0-9 phosphoglycolate phosphatase Homo sapiens 43-47 19538959-6 2009 Ritonavir also decreased glutathione reductase activities and glutathione peroxidase activities in CQ-resistant parasite line. Ritonavir 0-9 glutathione-disulfide reductase Homo sapiens 25-46 19562329-3 2009 The aim of the present study was to investigate potential drug interactions between the benzimidazoles albendazole and mebendazole and the potent CYP3A4 inhibitor ritonavir. Ritonavir 163-172 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 146-152 19790146-5 2009 In period 2, the addition of lopinavir/ritonavir to the immunosuppressant regimen enabled a reduction in the dose of immunosuppressants required to maintain trough concentrations within the therapeutic range (to 0.3 mg/day for tacrolimus and 75 mg/day for cyclosporine). Ritonavir 39-48 period circadian regulator 2 Homo sapiens 3-11 19500085-8 2009 Both nelfinavir and ritonavir decreased both M1 and M3, consistent with their ability to inhibit CYP3A and 2C8. Ritonavir 20-29 cholinergic receptor muscarinic 1 Homo sapiens 45-54 19792991-15 2009 CONCLUSION: Administration of ritonavir-boosted saquinavir markedly increased the exposure of midazolam by inhibiting its metabolism, confirming that the combination of saquinavir and ritonavir at steady state strongly inhibits CYP3A4 activity. Ritonavir 30-39 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 228-234 19792991-15 2009 CONCLUSION: Administration of ritonavir-boosted saquinavir markedly increased the exposure of midazolam by inhibiting its metabolism, confirming that the combination of saquinavir and ritonavir at steady state strongly inhibits CYP3A4 activity. Ritonavir 184-193 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 228-234 19679477-1 2009 The HIV protease inhibitor ritonavir (RTV) is also a potent inhibitor of the metabolizing enzyme cytochrome P450 3A (CYP3A) and is clinically useful in HIV therapy in its ability to enhance human plasma levels of other HIV protease inhibitors (PIs). Ritonavir 27-36 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 97-115 19679477-1 2009 The HIV protease inhibitor ritonavir (RTV) is also a potent inhibitor of the metabolizing enzyme cytochrome P450 3A (CYP3A) and is clinically useful in HIV therapy in its ability to enhance human plasma levels of other HIV protease inhibitors (PIs). Ritonavir 27-36 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 117-122 19679477-1 2009 The HIV protease inhibitor ritonavir (RTV) is also a potent inhibitor of the metabolizing enzyme cytochrome P450 3A (CYP3A) and is clinically useful in HIV therapy in its ability to enhance human plasma levels of other HIV protease inhibitors (PIs). Ritonavir 38-41 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 97-115 19679477-1 2009 The HIV protease inhibitor ritonavir (RTV) is also a potent inhibitor of the metabolizing enzyme cytochrome P450 3A (CYP3A) and is clinically useful in HIV therapy in its ability to enhance human plasma levels of other HIV protease inhibitors (PIs). Ritonavir 38-41 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 117-122 19500085-8 2009 Both nelfinavir and ritonavir decreased both M1 and M3, consistent with their ability to inhibit CYP3A and 2C8. Ritonavir 20-29 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 97-102 19349850-1 2009 In 29 antiretroviral-naive HIV-2-infected patients starting lopinavir/ritonavir-containing regimen, the median CD4 cell count change from baseline (142 cells/microl) was +71 cells/microl at week 24 and +132 cells/microl at week 96. Ritonavir 70-79 CD4 molecule Homo sapiens 111-114 19649922-1 2009 The HIV protease inhibitor (PI) ritonavir is a potent, mechanism-based inhibitor of cytochrome P450 CYP3A4, an enzyme that is responsible for metabolizing most HIV PIs. Ritonavir 32-41 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 100-106 19349850-3 2009 This sustained elevation of CD4 cell count in the first 2 years of combination antiretroviral therapy shows the potential for lopinavir/ritonavir regimens as first-line therapy in HIV-2 infection. Ritonavir 136-145 CD4 molecule Homo sapiens 28-31 19543499-7 2009 The IFN-gamma/IL-5 ratio of the CD3+ and CD4+ cells were increased significantly after 8 weeks and maintained until 1 yr of RIT, while there were no changes in the control group. Ritonavir 124-127 interferon gamma Homo sapiens 4-13 19307295-2 2009 Over half of the compounds tested (14 of 24) were identified as time-dependent inhibitors of CYP3A4 and high mRNA/activity ratios (>10) were consistent with CYP3A4 time-dependent inhibition for compounds such as troleandomycin, ritonavir, and verapamil. Ritonavir 231-240 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 93-99 19307295-2 2009 Over half of the compounds tested (14 of 24) were identified as time-dependent inhibitors of CYP3A4 and high mRNA/activity ratios (>10) were consistent with CYP3A4 time-dependent inhibition for compounds such as troleandomycin, ritonavir, and verapamil. Ritonavir 231-240 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 160-166 19543499-7 2009 The IFN-gamma/IL-5 ratio of the CD3+ and CD4+ cells were increased significantly after 8 weeks and maintained until 1 yr of RIT, while there were no changes in the control group. Ritonavir 124-127 interleukin 5 Homo sapiens 14-18 19381336-2 2009 This study was designed to evaluate the steady-state pharmacokinetics and tolerability of the coadministration of the PIs saquinavir and ritonavir (a CYP3A4 inhibitor used as a pharmacoenhancer of other PIs) and rifampin when coadministered in healthy HIV-negative volunteers. Ritonavir 137-146 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 150-156 19144412-11 2009 The BALF IL-4:IFN-gamma ratio significantly decreased over time in the IN RIT group (mean+/-SEM, D1 2.4+/-0.2, M6 1.0+/-0.2). Ritonavir 74-77 interleukin 4 Homo sapiens 9-13 19144412-11 2009 The BALF IL-4:IFN-gamma ratio significantly decreased over time in the IN RIT group (mean+/-SEM, D1 2.4+/-0.2, M6 1.0+/-0.2). Ritonavir 74-77 interferon gamma Homo sapiens 14-23 19292590-6 2009 By the Cox proportional hazards model, higher reported adherence levels and higher baseline CD4(+) T cell counts were associated with a greater likelihood of maintaining virologic suppression while receiving lopinavir/ritonavir monotherapy. Ritonavir 218-227 CD4 molecule Homo sapiens 92-95 19292590-8 2009 This analysis suggests that adherence and higher baseline CD4(+) T cell counts may help to predict who will sustain virologic suppression with lopinavir/ritonavir monotherapy. Ritonavir 153-162 CD4 molecule Homo sapiens 58-61 19176623-5 2009 In contrast, the five Gag mutations significantly delayed the acquisition of HIV-1 resistance to ritonavir and nelfinavir (NFV). Ritonavir 97-106 Pr55(Gag) Human immunodeficiency virus 1 22-25 19218343-3 2009 Endothelium-dependent vasorelaxation in response to bradykinin was reduced significantly by the ritonavir in a concentration-dependent manner. Ritonavir 96-105 kininogen 1 Homo sapiens 52-62 19218343-5 2009 Five HAART drugs (ritonavir, indinavir, lamivudine, abacavir, and AZT) significantly decreased endothelial nitric oxide synthase (eNOS) expression and increased superoxide anion levels in both vessels and HPAECs. Ritonavir 18-27 nitric oxide synthase 3 Homo sapiens 95-128 19218343-5 2009 Five HAART drugs (ritonavir, indinavir, lamivudine, abacavir, and AZT) significantly decreased endothelial nitric oxide synthase (eNOS) expression and increased superoxide anion levels in both vessels and HPAECs. Ritonavir 18-27 nitric oxide synthase 3 Homo sapiens 130-134 19218343-6 2009 Furthermore, both ritonavir and AZT substantially activated ERK2 in HPAECs. Ritonavir 18-27 mitogen-activated protein kinase 1 Homo sapiens 60-64 19218343-8 2009 Inhibition of ERK1/2 also partially blocked ritonavir- and AZT-induced down-regulation of eNOS and vasomotor dysfunction. Ritonavir 44-53 mitogen-activated protein kinase 3 Homo sapiens 14-20 19218343-8 2009 Inhibition of ERK1/2 also partially blocked ritonavir- and AZT-induced down-regulation of eNOS and vasomotor dysfunction. Ritonavir 44-53 nitric oxide synthase 3 Homo sapiens 90-94 18855943-5 2009 Whereas the P-gp inhibitor, PSC833, increased atazanavir and ritonavir accumulation in hCMEC/D3 cells by 2-fold, the MRP inhibitor MK571 had no effect. Ritonavir 61-70 phosphoglycolate phosphatase Homo sapiens 12-16 19270151-1 2009 Protease inhibitors boosted with ritonavir can lead to drug-drug interactions, particularly with inhaled corticosteroids such as fluticasone, because of the potent inhibition of cytochrome P450-3A4 activity. Ritonavir 33-42 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 178-197 19194313-1 2009 BACKGROUND: Preliminary studies suggest that the new film-coated tablet formulation of lopinavir/ritonavir (LPV/r-fct) could cut down the rate of adverse gastrointestinal symptoms of the conventional lopinavir/ritonavir soft gelatine capsules (LPV/r-sgc). Ritonavir 97-106 sarcoglycan beta Homo sapiens 250-253 18855943-7 2009 Treatment of hCMEC/D3 cells for 72 hr with rifampin or SR12813 (two well-established hPXR ligands) or PIs (atazanavir or ritonavir) resulted in an increase in P-gp expression by 1.8-, 6-, and 2-fold, respectively, with no effect observed for MRP1 expression. Ritonavir 121-130 phosphoglycolate phosphatase Homo sapiens 159-163 18855943-7 2009 Treatment of hCMEC/D3 cells for 72 hr with rifampin or SR12813 (two well-established hPXR ligands) or PIs (atazanavir or ritonavir) resulted in an increase in P-gp expression by 1.8-, 6-, and 2-fold, respectively, with no effect observed for MRP1 expression. Ritonavir 121-130 ATP binding cassette subfamily B member 1 Homo sapiens 242-246 18855943-9 2009 Long-term exposure of atazanavir or ritonavir to brain microvessel endothelium may result in further limitations in brain drug permeability as a result of the up-regulation of P-gp expression and function. Ritonavir 36-45 phosphoglycolate phosphatase Homo sapiens 176-180 18815591-0 2009 Dose-response of ritonavir on hepatic CYP3A activity and elvitegravir oral exposure. Ritonavir 17-26 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 38-43 19197797-6 2009 Consequently, the coadministration of elvitegravir with the protease inhibitor ritonavir (a substantial CYP3A4 inhibitor) results in significantly enhanced bioavailability and a longer half-life than with elvitegravir alone, allowing for the once-daily dosing of elvitegravir. Ritonavir 79-88 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 104-110 19004566-0 2009 Ritonavir and disulfiram have potential to inhibit caspase-1 mediated inflammation and reduce neurological sequelae after minor blast exposure. Ritonavir 0-9 caspase 1 Homo sapiens 51-60 19004566-3 2009 Ritonavir, a generically available protease inhibitor with a benign short-term side-effect profile, has been shown to inhibit expression of caspase-1. Ritonavir 0-9 caspase 1 Homo sapiens 140-149 19207033-3 2009 We investigated the impact of three common exonic ABCB1 polymorphisms on the concentrations of lopinavir and ritonavir in blood, semen and saliva of HIV-infected men under stable HAART containing ritonavir-boosted lopinavir. Ritonavir 109-118 ATP binding cassette subfamily B member 1 Homo sapiens 50-55 19095956-4 2009 In addition, the HIV PI ritonavir abrogated the interferon-gamma-mediated degradation of the RANKL nuclear adapter protein TRAF6, a physiological block to RANKL activity. Ritonavir 24-33 TNF superfamily member 11 Homo sapiens 93-98 19095956-4 2009 In addition, the HIV PI ritonavir abrogated the interferon-gamma-mediated degradation of the RANKL nuclear adapter protein TRAF6, a physiological block to RANKL activity. Ritonavir 24-33 TNF superfamily member 11 Homo sapiens 155-160 18815591-1 2009 Ritonavir, a potent inhibitor of cytochrome P450 isoform 3A (CYP3A) activity, is frequently used to boost the effects of protease inhibitors at doses of 100-400 mg per day; however, human data regarding the optimal dose required for boosting are limited. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 33-59 18815591-1 2009 Ritonavir, a potent inhibitor of cytochrome P450 isoform 3A (CYP3A) activity, is frequently used to boost the effects of protease inhibitors at doses of 100-400 mg per day; however, human data regarding the optimal dose required for boosting are limited. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 61-66 18815591-2 2009 This study systematically evaluated the ritonavir dose-response relationship on presystemic and systemic CYP3A metabolism using the human immunodeficiency virus integrase inhibitor elvitegravir and midazolam as probe substrates. Ritonavir 40-49 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 105-110 18815591-4 2009 The 20-mg dose of ritonavir substantially reduced CYP3A-mediated clearance (CL), as evidenced by a 66% reduction in midazolam CL that plateaued to 17% of baseline activity at a 100-mg dose. Ritonavir 18-27 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 50-55 18815591-6 2009 These data provide a critical understanding of ritonavir"s dose-response relationship for inhibition of CYP3A activity in humans. Ritonavir 47-56 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 104-109 19323590-20 2009 The "boosting" dose of ritonavir acts an an inhibitor of CYP3A, thereby increasing darunavir bioavailability. Ritonavir 23-32 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 57-62 19960055-0 2009 Functional role of p-glycoprotein and binding protein effect on the placental transfer of lopinavir/ritonavir in the ex vivo human perfusion model. Ritonavir 100-109 ATP binding cassette subfamily B member 1 Homo sapiens 19-33 19834271-10 2009 EFV, ritonavir (RTV) and nelfinavir (NFV) inhibited the expression of adiponectin mRNA in mature 3T3-L1 and to a greater extent in pre-mature 3T3-L1. Ritonavir 5-14 adiponectin, C1Q and collagen domain containing Homo sapiens 70-81 19834271-10 2009 EFV, ritonavir (RTV) and nelfinavir (NFV) inhibited the expression of adiponectin mRNA in mature 3T3-L1 and to a greater extent in pre-mature 3T3-L1. Ritonavir 16-19 adiponectin, C1Q and collagen domain containing Homo sapiens 70-81 19202563-3 2009 Clotrimazole, rifampin, ritonavir, phenytoin, and phenobarbital induced CYP2C9 consistent with previous findings for CYP3A4. Ritonavir 24-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 19202563-3 2009 Clotrimazole, rifampin, ritonavir, phenytoin, and phenobarbital induced CYP2C9 consistent with previous findings for CYP3A4. Ritonavir 24-33 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 117-123 18854490-6 2009 Oral codosing of TC-1 with ritonavir, a potent CYP3A4 inhibitor, twice daily over 3.5 days in rhesus monkeys led to sustained plasma TC-1 exposure and a significant and sustained reduction in CSF sAPP beta, A beta 40, A beta 42, and plasma A beta 40 levels. Ritonavir 27-36 CYP3A4 Macaca mulatta 47-53 18854490-6 2009 Oral codosing of TC-1 with ritonavir, a potent CYP3A4 inhibitor, twice daily over 3.5 days in rhesus monkeys led to sustained plasma TC-1 exposure and a significant and sustained reduction in CSF sAPP beta, A beta 40, A beta 42, and plasma A beta 40 levels. Ritonavir 27-36 amyloid beta (A4) precursor protein Mus musculus 207-213 18854490-6 2009 Oral codosing of TC-1 with ritonavir, a potent CYP3A4 inhibitor, twice daily over 3.5 days in rhesus monkeys led to sustained plasma TC-1 exposure and a significant and sustained reduction in CSF sAPP beta, A beta 40, A beta 42, and plasma A beta 40 levels. Ritonavir 27-36 amyloid beta (A4) precursor protein Mus musculus 218-224 18854490-6 2009 Oral codosing of TC-1 with ritonavir, a potent CYP3A4 inhibitor, twice daily over 3.5 days in rhesus monkeys led to sustained plasma TC-1 exposure and a significant and sustained reduction in CSF sAPP beta, A beta 40, A beta 42, and plasma A beta 40 levels. Ritonavir 27-36 amyloid beta (A4) precursor protein Mus musculus 218-224 19114959-5 2009 In response to bradykinin (10(-6) M), ritonavir-treated rings showed a significant reduction of endothelium-dependent vasorelaxation by 32% compared with untreated control vessels (P<0.05, n=5, Student t-test). Ritonavir 38-47 kininogen 1 Homo sapiens 15-25 19960055-2 2009 To study the influence of P-glycoprotein (P-glycoprotein, ABCB1, MDR1) function on placental transfer of lopinavir with ritonavir at different albumin concentrations. Ritonavir 120-129 ATP binding cassette subfamily B member 1 Homo sapiens 26-40 18849545-0 2008 Ritonavir increases CD36, ABCA1 and CYP27 expression in THP-1 macrophages. Ritonavir 0-9 GLI family zinc finger 2 Homo sapiens 56-61 18528434-2 2008 Here we report the effect of ATV and ATV/ritonavir (RTV) on another UGT1A isoenzyme, UGT1A4. Ritonavir 41-50 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 68-73 18528434-2 2008 Here we report the effect of ATV and ATV/ritonavir (RTV) on another UGT1A isoenzyme, UGT1A4. Ritonavir 41-50 UDP glucuronosyltransferase family 1 member A4 Homo sapiens 85-91 18528434-2 2008 Here we report the effect of ATV and ATV/ritonavir (RTV) on another UGT1A isoenzyme, UGT1A4. Ritonavir 52-55 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 68-73 18528434-2 2008 Here we report the effect of ATV and ATV/ritonavir (RTV) on another UGT1A isoenzyme, UGT1A4. Ritonavir 52-55 UDP glucuronosyltransferase family 1 member A4 Homo sapiens 85-91 18849545-4 2008 Exposure to ritonavir (2.5 mug/ml) increased CD36 protein (28%, P < 0.05) and mRNA (38%, P < 0.05) in differentiated THP-1 macrophages, but not in undifferentiated monocytes. Ritonavir 12-21 GLI family zinc finger 2 Homo sapiens 123-128 18849545-8 2008 CONCLUSIONS: We propose that ritonavir induces ABCA1 expression in THP-1 macrophages through LXRalpha. Ritonavir 29-38 GLI family zinc finger 2 Homo sapiens 67-72 18692133-4 2008 Moreover, we have reported the impact of FTC combined with protease inhibitors (PIs) (ritonavir, atazanavir, lopinavir) on Pgp and MRP1 expression and function, and on PI accumulation. Ritonavir 86-95 phosphoglycolate phosphatase Homo sapiens 123-126 19186349-1 2008 PURPOSE: Ritonavir is a powerful inhibitor of cytochrome P450 3A (CYP3A) that metabolizes many antiretrovirals. Ritonavir 9-18 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 46-64 19186349-1 2008 PURPOSE: Ritonavir is a powerful inhibitor of cytochrome P450 3A (CYP3A) that metabolizes many antiretrovirals. Ritonavir 9-18 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 66-71 19186349-7 2008 CYP3A activity in subjects on ritonavir and with CVH was further reduced to 4% of controls (no CVH, R+ 2.1 +/- 0.8 vs. R+, CVH+ 1.0 +/- 0.4 mL/min/kg, P < 0.006). Ritonavir 30-39 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-5 19186349-8 2008 CONCLUSIONS: Ritonavir markedly decreases CYP3A activity. Ritonavir 13-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 42-47 19186349-9 2008 In the presence of CVH, ritonavir-based therapy further reduces CYP3A activity by half. Ritonavir 24-33 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 64-69 19186349-10 2008 Coinfection with CVH impairs CYP3A activity in the presence of the CYP3A inhibitor ritonavir. Ritonavir 83-92 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 67-72 18957053-3 2008 In this study, we investigated a role for Rit signaling in IFNgamma-induced dendritic retraction. Ritonavir 42-45 interferon gamma Rattus norvegicus 59-67 18957053-5 2008 In pheochromacytoma cells and hippocampal neurons, IFNgamma caused rapid Rit activation as indicated by increased GTP binding to Rit. Ritonavir 73-76 interferon gamma Rattus norvegicus 51-59 18957053-5 2008 In pheochromacytoma cells and hippocampal neurons, IFNgamma caused rapid Rit activation as indicated by increased GTP binding to Rit. Ritonavir 129-132 interferon gamma Rattus norvegicus 51-59 18957053-6 2008 Silencing of Rit by RNA interference suppressed IFNgamma-elicited activation of p38 MAPK in pheochromacytoma cells, and pharmacological inhibition of p38 MAPK significantly attenuated the dendrite-inhibiting effects of IFNgamma in cultured sympathetic and hippocampal neurons without altering signal transducer and activator of transcription 1 activation. Ritonavir 13-16 interferon gamma Rattus norvegicus 48-56 18957053-6 2008 Silencing of Rit by RNA interference suppressed IFNgamma-elicited activation of p38 MAPK in pheochromacytoma cells, and pharmacological inhibition of p38 MAPK significantly attenuated the dendrite-inhibiting effects of IFNgamma in cultured sympathetic and hippocampal neurons without altering signal transducer and activator of transcription 1 activation. Ritonavir 13-16 mitogen activated protein kinase 14 Rattus norvegicus 80-83 18957053-6 2008 Silencing of Rit by RNA interference suppressed IFNgamma-elicited activation of p38 MAPK in pheochromacytoma cells, and pharmacological inhibition of p38 MAPK significantly attenuated the dendrite-inhibiting effects of IFNgamma in cultured sympathetic and hippocampal neurons without altering signal transducer and activator of transcription 1 activation. Ritonavir 13-16 mitogen activated protein kinase 14 Rattus norvegicus 150-153 18957053-6 2008 Silencing of Rit by RNA interference suppressed IFNgamma-elicited activation of p38 MAPK in pheochromacytoma cells, and pharmacological inhibition of p38 MAPK significantly attenuated the dendrite-inhibiting effects of IFNgamma in cultured sympathetic and hippocampal neurons without altering signal transducer and activator of transcription 1 activation. Ritonavir 13-16 interferon gamma Rattus norvegicus 219-227 18957053-6 2008 Silencing of Rit by RNA interference suppressed IFNgamma-elicited activation of p38 MAPK in pheochromacytoma cells, and pharmacological inhibition of p38 MAPK significantly attenuated the dendrite-inhibiting effects of IFNgamma in cultured sympathetic and hippocampal neurons without altering signal transducer and activator of transcription 1 activation. Ritonavir 13-16 signal transducer and activator of transcription 1 Rattus norvegicus 293-343 18957053-7 2008 These observations identify Rit as a downstream target of IFNgamma and suggest that a novel IFNgamma-Rit-p38 signaling pathway contributes to dendritic retraction and may, therefore, represent a potential therapeutic target in diseases with a significant neuroinflammatory component. Ritonavir 28-31 interferon gamma Rattus norvegicus 58-66 18957053-7 2008 These observations identify Rit as a downstream target of IFNgamma and suggest that a novel IFNgamma-Rit-p38 signaling pathway contributes to dendritic retraction and may, therefore, represent a potential therapeutic target in diseases with a significant neuroinflammatory component. Ritonavir 28-31 mitogen activated protein kinase 14 Rattus norvegicus 105-108 18957053-7 2008 These observations identify Rit as a downstream target of IFNgamma and suggest that a novel IFNgamma-Rit-p38 signaling pathway contributes to dendritic retraction and may, therefore, represent a potential therapeutic target in diseases with a significant neuroinflammatory component. Ritonavir 101-104 interferon gamma Rattus norvegicus 92-100 18957053-7 2008 These observations identify Rit as a downstream target of IFNgamma and suggest that a novel IFNgamma-Rit-p38 signaling pathway contributes to dendritic retraction and may, therefore, represent a potential therapeutic target in diseases with a significant neuroinflammatory component. Ritonavir 101-104 mitogen activated protein kinase 14 Rattus norvegicus 105-108 18692133-4 2008 Moreover, we have reported the impact of FTC combined with protease inhibitors (PIs) (ritonavir, atazanavir, lopinavir) on Pgp and MRP1 expression and function, and on PI accumulation. Ritonavir 86-95 ATP binding cassette subfamily C member 1 Homo sapiens 131-135 19238656-6 2008 Acute ritonavir inhibits first-pass CYP3A > 96%. Ritonavir 6-15 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 36-41 19238656-1 2008 Ritonavir effects on CYP3A and P-glycoprotein activities. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 21-26 19238656-1 2008 Ritonavir effects on CYP3A and P-glycoprotein activities. Ritonavir 0-9 ATP binding cassette subfamily B member 1 Homo sapiens 31-45 19238656-2 2008 Ritonavir diminishes methadone plasma concentrations, an effect attributed to CYP3A induction, but the actual mechanisms are unknown. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 78-83 19008174-3 2008 In this retrospective analysis of 22 patients during therapy with tipranavir/ritonavir (TPV) 500 mg/200 mg bid, we found significantly decreased TPV-trough levels in combination with tenofovir (15.32+/-5.22 microg/ml) in comparison to TPV trough levels without tenofovir (20.21+/-14.87 microg/ml). Ritonavir 77-86 BH3 interacting domain death agonist Homo sapiens 107-110 19238656-3 2008 We determined short-term (2-day) and steady-state (2-week) ritonavir effects on intestinal and hepatic CYP3A4/5 (probed with intravenous (IV) and oral alfentanil (ALF) and with miosis) and P-glycoprotein (P-gp) (fexofenadine), and on methadone pharmacokinetics and pharmacodynamics in healthy volunteers. Ritonavir 59-68 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 103-109 19238656-7 2008 Chronic ritonavir inhibits hepatic CYP3A (> 70%) and first-pass CYP3A (> 90%). Ritonavir 8-17 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 35-40 19238656-7 2008 Chronic ritonavir inhibits hepatic CYP3A (> 70%) and first-pass CYP3A (> 90%). Ritonavir 8-17 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 67-72 19238656-8 2008 Acute and steady-state ritonavir increased the fexofenadine AUC(0-infinity) 2.8- and 1.4-fold, respectively, suggesting P-gp inhibition. Ritonavir 23-32 ATP binding cassette subfamily B member 1 Homo sapiens 120-124 19238656-9 2008 Steady-state compared with acute ritonavir caused mild apparent induction of P-gp and hepatic CYP3A, but net inhibition still predominated. Ritonavir 33-42 ATP binding cassette subfamily B member 1 Homo sapiens 77-81 19238656-9 2008 Steady-state compared with acute ritonavir caused mild apparent induction of P-gp and hepatic CYP3A, but net inhibition still predominated. Ritonavir 33-42 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 94-99 19238656-10 2008 Ritonavir inhibited both intestinal and hepatic CYP3A and drug transport. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 48-53 19238656-11 2008 ALF miosis noninvasively determined CYP3A inhibition by ritonavir. Ritonavir 56-65 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 36-41 18523872-6 2008 P-gp inhibitors ivermectin, LY335979, PSC833, and the P-gp/Bcrp inhibitor ritonavir did not influence Bcrp mediated topotecan transport, however, blocked ABZSO transport. Ritonavir 74-83 phosphoglycolate phosphatase Mus musculus 54-58 19195459-2 2008 This drug is primarily metabolized by the liver through the cytochrome P450 3A4 (CYP3A4) isozyme and consequently coadministration with low doses of ritonavir markedly increases exposure and allows both daily doses and pill burden to be reduced. Ritonavir 149-158 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 60-79 19195459-2 2008 This drug is primarily metabolized by the liver through the cytochrome P450 3A4 (CYP3A4) isozyme and consequently coadministration with low doses of ritonavir markedly increases exposure and allows both daily doses and pill burden to be reduced. Ritonavir 149-158 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 81-87 19195459-3 2008 Both darunavir and ritonavir act as inhibitors and substrates of CYP3A4 and may therefore present pharmacological interactions with a large number of drugs commonly used in HIV-infected patients. Ritonavir 19-28 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 65-71 18719645-4 2008 Microarray analysis also identified a novel gene downregulated by ritonavir, Cidea, whose expression levels may affect free-fatty acid metabolism. Ritonavir 66-75 cell death-inducing DNA fragmentation factor, alpha subunit-like effector A Mus musculus 77-82 18523872-6 2008 P-gp inhibitors ivermectin, LY335979, PSC833, and the P-gp/Bcrp inhibitor ritonavir did not influence Bcrp mediated topotecan transport, however, blocked ABZSO transport. Ritonavir 74-83 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 59-63 18676772-10 2008 GRN163L-induced cell death could also be expedited by addition of the chemotherapeutic agents doxorubicin and ritonavir. Ritonavir 110-119 granulin Mus musculus 0-3 18639527-4 2008 Here, we purified Ste24p, the yeast ortholog of ZMPSTE24, and showed that its enzymatic activity was blocked by three HIV-PIs (lopinavir, ritonavir, and tipranavir). Ritonavir 138-147 zinc metalloprotease Saccharomyces cerevisiae S288C 18-24 18620493-5 2008 Among the drugs tested, several inhibited PAF-induced platelet aggregation in a concentration-depended manner, with tenofovir, efavirenz, and ritonavir exhibiting the higher inhibitory effect. Ritonavir 142-151 PCNA clamp associated factor Homo sapiens 42-45 18580608-9 2008 In contrast, both atazanavir and ritonavir were associated with altered leptin secretion. Ritonavir 33-42 leptin Homo sapiens 72-78 18458892-3 2008 Efavirenz is an inducer of CYP3A, whereas the ritonavir-boosted regimens are net inhibitors of CYP3A. Ritonavir 46-55 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 95-100 18458892-7 2008 CONCLUSION: Changes in plasma 4beta-hydroxycholesterol following the initiation of efavirenz- or atazanavir/ritonavir-based antiretroviral therapy reflected the respective net increase and decrease of CYP3A activity of these regimens. Ritonavir 108-117 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 201-206 18577765-5 2008 DATA SYNTHESIS: Ritonavir is a well-known inhibitor of the metabolism of numerous medications that are substrates of the CYP3A and CYP2D6 pathways. Ritonavir 16-25 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 131-137 18576910-9 2008 The increased warfarin doses required in these two patients may have been caused by induction of CYP3A4 by nevirapine, CYP2C9 by nelfinavir, or CYP2C9 by lopinavir-ritonavir. Ritonavir 164-173 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 97-103 18183034-0 2008 Effect of an antiretroviral regimen containing ritonavir boosted lopinavir on intestinal and hepatic CYP3A, CYP2D6 and P-glycoprotein in HIV-infected patients. Ritonavir 47-56 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 101-106 18183034-0 2008 Effect of an antiretroviral regimen containing ritonavir boosted lopinavir on intestinal and hepatic CYP3A, CYP2D6 and P-glycoprotein in HIV-infected patients. Ritonavir 47-56 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 108-114 18183034-0 2008 Effect of an antiretroviral regimen containing ritonavir boosted lopinavir on intestinal and hepatic CYP3A, CYP2D6 and P-glycoprotein in HIV-infected patients. Ritonavir 47-56 ATP binding cassette subfamily B member 1 Homo sapiens 119-133 18183034-1 2008 This study aimed to quantify the inhibition of cytochrome P450 (CYP3A), CYP2D6, and P-glycoprotein in human immunodeficiency virus (HIV)-infected patients receiving an antiretroviral therapy (ART) containing ritonavir boosted lopinavir, and to identify factors influencing ritonavir and lopinavir pharmacokinetics. Ritonavir 208-217 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 64-69 18183034-1 2008 This study aimed to quantify the inhibition of cytochrome P450 (CYP3A), CYP2D6, and P-glycoprotein in human immunodeficiency virus (HIV)-infected patients receiving an antiretroviral therapy (ART) containing ritonavir boosted lopinavir, and to identify factors influencing ritonavir and lopinavir pharmacokinetics. Ritonavir 273-282 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 64-69 18183034-8 2008 In conclusion, CYP3A, CYP2D6, and P-glycoprotein are profoundly inhibited in patients receiving ritonavir boosted lopinavir. Ritonavir 96-105 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 15-20 18183034-8 2008 In conclusion, CYP3A, CYP2D6, and P-glycoprotein are profoundly inhibited in patients receiving ritonavir boosted lopinavir. Ritonavir 96-105 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 22-28 18183034-8 2008 In conclusion, CYP3A, CYP2D6, and P-glycoprotein are profoundly inhibited in patients receiving ritonavir boosted lopinavir. Ritonavir 96-105 ATP binding cassette subfamily B member 1 Homo sapiens 34-48 18576910-9 2008 The increased warfarin doses required in these two patients may have been caused by induction of CYP3A4 by nevirapine, CYP2C9 by nelfinavir, or CYP2C9 by lopinavir-ritonavir. Ritonavir 164-173 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 144-150 18356150-7 2008 At baseline, patients starting lopinavir/ritonavir had higher HIV-1 RNA and lower CD4+ cell counts. Ritonavir 41-50 CD4 molecule Homo sapiens 82-85 18520056-1 2008 Atazanavir (ATV) is clinically coadministered with low-dose ritonavir (RTV), which boosts the oral bioavailability (BA) of ATV by inhibiting cytochrome P450 (CYP) 3A, and P-glycoprotein (Pgp) via the same metabolic pathway; however, it is well known that in the chronic phase, the inhibition effect of RTV on Pgp and CYP3A becomes an induction effect. Ritonavir 60-69 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 141-165 18520056-1 2008 Atazanavir (ATV) is clinically coadministered with low-dose ritonavir (RTV), which boosts the oral bioavailability (BA) of ATV by inhibiting cytochrome P450 (CYP) 3A, and P-glycoprotein (Pgp) via the same metabolic pathway; however, it is well known that in the chronic phase, the inhibition effect of RTV on Pgp and CYP3A becomes an induction effect. Ritonavir 60-69 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 171-185 18520056-1 2008 Atazanavir (ATV) is clinically coadministered with low-dose ritonavir (RTV), which boosts the oral bioavailability (BA) of ATV by inhibiting cytochrome P450 (CYP) 3A, and P-glycoprotein (Pgp) via the same metabolic pathway; however, it is well known that in the chronic phase, the inhibition effect of RTV on Pgp and CYP3A becomes an induction effect. Ritonavir 60-69 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 187-190 18520056-1 2008 Atazanavir (ATV) is clinically coadministered with low-dose ritonavir (RTV), which boosts the oral bioavailability (BA) of ATV by inhibiting cytochrome P450 (CYP) 3A, and P-glycoprotein (Pgp) via the same metabolic pathway; however, it is well known that in the chronic phase, the inhibition effect of RTV on Pgp and CYP3A becomes an induction effect. Ritonavir 60-69 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 309-312 18520056-1 2008 Atazanavir (ATV) is clinically coadministered with low-dose ritonavir (RTV), which boosts the oral bioavailability (BA) of ATV by inhibiting cytochrome P450 (CYP) 3A, and P-glycoprotein (Pgp) via the same metabolic pathway; however, it is well known that in the chronic phase, the inhibition effect of RTV on Pgp and CYP3A becomes an induction effect. Ritonavir 60-69 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 317-322 18520056-1 2008 Atazanavir (ATV) is clinically coadministered with low-dose ritonavir (RTV), which boosts the oral bioavailability (BA) of ATV by inhibiting cytochrome P450 (CYP) 3A, and P-glycoprotein (Pgp) via the same metabolic pathway; however, it is well known that in the chronic phase, the inhibition effect of RTV on Pgp and CYP3A becomes an induction effect. Ritonavir 71-74 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 141-165 18520056-1 2008 Atazanavir (ATV) is clinically coadministered with low-dose ritonavir (RTV), which boosts the oral bioavailability (BA) of ATV by inhibiting cytochrome P450 (CYP) 3A, and P-glycoprotein (Pgp) via the same metabolic pathway; however, it is well known that in the chronic phase, the inhibition effect of RTV on Pgp and CYP3A becomes an induction effect. Ritonavir 71-74 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 171-185 18520056-1 2008 Atazanavir (ATV) is clinically coadministered with low-dose ritonavir (RTV), which boosts the oral bioavailability (BA) of ATV by inhibiting cytochrome P450 (CYP) 3A, and P-glycoprotein (Pgp) via the same metabolic pathway; however, it is well known that in the chronic phase, the inhibition effect of RTV on Pgp and CYP3A becomes an induction effect. Ritonavir 71-74 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 187-190 18520056-1 2008 Atazanavir (ATV) is clinically coadministered with low-dose ritonavir (RTV), which boosts the oral bioavailability (BA) of ATV by inhibiting cytochrome P450 (CYP) 3A, and P-glycoprotein (Pgp) via the same metabolic pathway; however, it is well known that in the chronic phase, the inhibition effect of RTV on Pgp and CYP3A becomes an induction effect. Ritonavir 71-74 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 309-312 18520056-1 2008 Atazanavir (ATV) is clinically coadministered with low-dose ritonavir (RTV), which boosts the oral bioavailability (BA) of ATV by inhibiting cytochrome P450 (CYP) 3A, and P-glycoprotein (Pgp) via the same metabolic pathway; however, it is well known that in the chronic phase, the inhibition effect of RTV on Pgp and CYP3A becomes an induction effect. Ritonavir 71-74 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 317-322 18356600-8 2008 Patients taking indinavir boosted with ritonavir had median fibrinogen levels 8% higher than those on indinavir alone (P = 0.049). Ritonavir 39-48 fibrinogen beta chain Homo sapiens 60-70 18469453-0 2008 Ritonavir and disulfiram may be synergistic in lowering active interleukin-18 levels in acute pancreatitis, and thereby hasten recovery. Ritonavir 0-9 interleukin 18 Homo sapiens 63-77 18469453-4 2008 The anti-retroviral drug ritonavir inhibits conversion of inactive pro-caspase-1 to active caspase-1. Ritonavir 25-34 caspase 1 Homo sapiens 71-80 18469453-4 2008 The anti-retroviral drug ritonavir inhibits conversion of inactive pro-caspase-1 to active caspase-1. Ritonavir 25-34 caspase 1 Homo sapiens 91-100 18469453-5 2008 Since ritonavir is well tolerated in short-term use it may therefore prove useful in treating acute pancreatitis by lowering caspase-1 mediated IL-18 formation and the many inflammatory mediators downstream from that. Ritonavir 6-15 caspase 1 Homo sapiens 125-134 18469453-5 2008 Since ritonavir is well tolerated in short-term use it may therefore prove useful in treating acute pancreatitis by lowering caspase-1 mediated IL-18 formation and the many inflammatory mediators downstream from that. Ritonavir 6-15 interleukin 18 Homo sapiens 144-149 18469453-9 2008 Given the current morbidity and mortality of pancreatitis, research should be directed to ritonavir and disulfiram as treatment options for illnesses like pancreatitis where excessive IL-18 contributes to pathology. Ritonavir 90-99 interleukin 18 Homo sapiens 184-189 18285471-1 2008 Ritonavir is the most potent and efficacious inhibitor of cytochrome P4503A (CYP3A), and it is used accordingly for the pharmacoenhancement of other antiretrovirals. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 58-64 18285471-1 2008 Ritonavir is the most potent and efficacious inhibitor of cytochrome P4503A (CYP3A), and it is used accordingly for the pharmacoenhancement of other antiretrovirals. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 77-82 18285471-4 2008 Ritonavir induces CYP2B6 in human hepatocytes. Ritonavir 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 18-24 18285471-5 2008 This investigation tested the hypothesis that ritonavir induces human CYP2B6 in vivo. Ritonavir 46-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-76 18285471-13 2008 These results show that ritonavir induces human CYP2B6 activity. Ritonavir 24-33 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-54 18285471-15 2008 The ritonavir induction of CYP2B6 activity may have significant implications for drug interactions and clarify previously unexplained interactions. Ritonavir 4-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 18285477-0 2008 Effect of low-dose ritonavir on the pharmacokinetics of the CXCR4 antagonist AMD070 in healthy volunteers. Ritonavir 19-28 C-X-C motif chemokine receptor 4 Homo sapiens 60-65 18285477-2 2008 Since AMD070 is a substrate of cytochrome P450 3A4 and P-glycoprotein, both of which may be affected by ritonavir, we tested for a ritonavir effect on AMD070 pharmacokinetics. Ritonavir 104-113 ATP binding cassette subfamily B member 1 Homo sapiens 55-69 18285477-2 2008 Since AMD070 is a substrate of cytochrome P450 3A4 and P-glycoprotein, both of which may be affected by ritonavir, we tested for a ritonavir effect on AMD070 pharmacokinetics. Ritonavir 131-140 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 31-50 18285477-2 2008 Since AMD070 is a substrate of cytochrome P450 3A4 and P-glycoprotein, both of which may be affected by ritonavir, we tested for a ritonavir effect on AMD070 pharmacokinetics. Ritonavir 131-140 ATP binding cassette subfamily B member 1 Homo sapiens 55-69 18473749-9 2008 A small number of drugs such as rifampin, phenytoin and ritonavir are identified as inducers of CYP3A4. Ritonavir 56-65 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 96-102 18230615-10 2008 Ritonavir, like lopinavir, inhibits ZMPSTE24 and leads to an accumulation of prelamin A. Ritonavir 0-9 zinc metallopeptidase STE24 Homo sapiens 36-44 18646322-9 2008 Ritonavir 10 microg/mL caused a similar P-gp increment as did 20 microg/ mL saquinavir. Ritonavir 0-9 ATP binding cassette subfamily B member 1 Homo sapiens 40-44 18497877-5 2008 Oral administration of the PIs nelfinavir and ritonavir significantly inhibited photoreceptor apoptosis, while preventing the translocation of AIF from mitochondria to the nucleus as well as the activation of caspase-9. Ritonavir 46-55 apoptosis-inducing factor, mitochondrion-associated 1 Mus musculus 143-146 18497877-5 2008 Oral administration of the PIs nelfinavir and ritonavir significantly inhibited photoreceptor apoptosis, while preventing the translocation of AIF from mitochondria to the nucleus as well as the activation of caspase-9. Ritonavir 46-55 caspase 9 Mus musculus 209-218 18093255-4 2008 WHAT THIS STUDY ADDS: The present study also utilizes the human perfused human isolated placenta to investigate the possible inhibitory effects of the P-gp inhibitor PSC833 and the P-gp substrate/inhibitor ritonavir on the maternal to foetal transfer clearance of indinavir. Ritonavir 206-215 phosphoglycolate phosphatase Homo sapiens 181-185 17713471-5 2008 The LPV/RTV treatment also inhibited the formation of SN38 glucuronide (SN38G), as shown by the 36% decrease in the SN38G/SN38 AUCs ratio (5.9+/-1.6 vs 9.2+/-2.6, P=0.002) consistent with UGT1A1 inhibition by LPV/RTV. Ritonavir 8-11 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 188-194 18356600-10 2008 The associations of ritonavir, indinavir, efavirenz and nevirapine with fibrinogen levels persisted after multivariable analysis and were independent of other antiretroviral use. Ritonavir 20-29 fibrinogen beta chain Homo sapiens 72-82 18096673-5 2008 A-792611 also attenuated the rifampin and ritonavir-mediated activation of the human pregnane X receptor (PXR) in luciferase reporter assays. Ritonavir 42-51 nuclear receptor subfamily 1 group I member 2 Homo sapiens 85-104 18474494-0 2008 Randomized study of dual versus single ritonavir-enhanced protease inhibitors for protease inhibitor-experienced patients with HIV. Ritonavir 39-48 serpin family A member 13, pseudogene Homo sapiens 58-76 18096673-5 2008 A-792611 also attenuated the rifampin and ritonavir-mediated activation of the human pregnane X receptor (PXR) in luciferase reporter assays. Ritonavir 42-51 nuclear receptor subfamily 1 group I member 2 Homo sapiens 106-109 17981343-12 2008 BALF IL-4 concentrations were significantly higher at week 1 in adjuvanted RIT cats compared with baseline and month 2, and also with placebo RIT cats at week 1. Ritonavir 75-78 interleukin 4 Felis catus 5-9 17981343-12 2008 BALF IL-4 concentrations were significantly higher at week 1 in adjuvanted RIT cats compared with baseline and month 2, and also with placebo RIT cats at week 1. Ritonavir 142-145 interleukin 4 Felis catus 5-9 18090045-0 2007 Effects of ritonavir and amprenavir on insulin sensitivity in healthy volunteers. Ritonavir 11-20 insulin Homo sapiens 39-46 18317438-0 2008 Improvement in insulin sensitivity and dyslipidemia in protease inhibitor-treated adult male patients after switch to atazanavir/ritonavir. Ritonavir 129-138 insulin Homo sapiens 15-22 18317438-10 2008 CONCLUSIONS: Using the gold-standard euglycemic clamp, ritonavir-boosted ATV therapy improved PI-induced insulin resistance among dyslipidemic HIV-infected men on PI-based antiretroviral therapy. Ritonavir 55-64 insulin Homo sapiens 105-112 18389894-2 2008 Darunavir inhibits and is primarily metabolized by cytochrome P450 3A (CYP3A) isoenzymes and is coadministered with low-dose ritonavir (darunavir/r); ritonavir is an inhibitor of CYP3A isoenzymes and pharmacologically enhances darunavir, resulting in increased plasma concentrations and allowing for a lower daily dose. Ritonavir 150-159 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 51-69 18389894-2 2008 Darunavir inhibits and is primarily metabolized by cytochrome P450 3A (CYP3A) isoenzymes and is coadministered with low-dose ritonavir (darunavir/r); ritonavir is an inhibitor of CYP3A isoenzymes and pharmacologically enhances darunavir, resulting in increased plasma concentrations and allowing for a lower daily dose. Ritonavir 150-159 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 71-76 17581594-0 2008 Ritonavir 100 mg does not cause QTc prolongation in healthy subjects: a possible role as CYP3A inhibitor in thorough QTc studies. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 89-94 18504407-4 2008 OBJECTIVE: We sought to investigate the changes in allergen-induced CD203c expression in patients with Japanese cedar pollen (JCP) pollinosis who received RIT. Ritonavir 155-158 ectonucleotide pyrophosphatase/phosphodiesterase 3 Homo sapiens 68-74 18504407-15 2008 Significant reductions in the responses were observed in 4 subjects after RIT (reduction in CD203c expression, RCE) whereas no changes were seen in 3 subjects (non-RCE). Ritonavir 74-77 ectonucleotide pyrophosphatase/phosphodiesterase 3 Homo sapiens 92-98 18094422-1 2007 PURPOSE: To evaluate the effects of ritonavir, a potent inhibitor of CYP3A4, on the steady-state pharmacokinetics of imatinib. Ritonavir 36-45 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 69-75 18094422-8 2007 Ritonavir (1 micromol/L) completely inhibited CYP3A4-mediated metabolism of imatinib to CGP74588 but inhibited metabolism in microsomes by only 50%. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 46-52 17976838-5 2007 We find that Rit-mediated neuritogenesis is dependent upon MEK/ERK MAP kinase signaling but independent of RalGEF activation. Ritonavir 13-16 mitogen-activated protein kinase kinase 7 Homo sapiens 59-62 17976838-5 2007 We find that Rit-mediated neuritogenesis is dependent upon MEK/ERK MAP kinase signaling but independent of RalGEF activation. Ritonavir 13-16 mitogen-activated protein kinase 1 Homo sapiens 63-66 18085477-1 2007 In THP-1 monocytes, cellular proteasome inhibition by ritonavir or ALLN is associated with increased production of oxidative stress. Ritonavir 54-63 GLI family zinc finger 2 Homo sapiens 3-8 18090297-0 2007 Effects of atazanavir/ritonavir and lopinavir/ritonavir on glucose uptake and insulin sensitivity. Ritonavir 46-55 insulin Homo sapiens 78-85 18090045-2 2007 METHODS: In two separate double-blind, randomized, cross-over studies, we assessed the effects of a single dose of ritonavir (800 mg) and amprenavir (1200 mg) on insulin sensitivity (euglycemic hyperglycemic clamp) in healthy normal volunteers. Ritonavir 115-124 insulin Homo sapiens 162-169 18090045-4 2007 CONCLUSION: Compared to previously performed studies of identical design using single doses of indinavir and lopinavir/ritonavir, a hierarchy of insulin resistance was observed with the greatest effect seen with indinavir followed by ritonavir and lopinavir/ritonavir, with little effect of amprenavir. Ritonavir 234-243 insulin Homo sapiens 145-152 18090045-4 2007 CONCLUSION: Compared to previously performed studies of identical design using single doses of indinavir and lopinavir/ritonavir, a hierarchy of insulin resistance was observed with the greatest effect seen with indinavir followed by ritonavir and lopinavir/ritonavir, with little effect of amprenavir. Ritonavir 234-243 insulin Homo sapiens 145-152 17700364-5 2007 RESULTS: Variants of ABCA1, APOA5, APOC3, APOE, and CETP contributed to plasma triglyceride levels, particularly in the setting of ritonavir-containing antiretroviral therapy. Ritonavir 131-140 ATP binding cassette subfamily A member 1 Homo sapiens 21-26 17639026-3 2007 At 10 microM, ritonavir and nelfinavir suppressed CYP3A4 activity but induced its transcripts and protein expression (19- and 12- and 12- and 6-fold, respectively; a >2-fold change over control was interpreted as induction). Ritonavir 14-23 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 50-56 17517050-10 2007 Through its inhibition of CYP3A and P-gp, ritonavir could attenuate the pharmacokinetic variability linked to genetic differences, reducing significantly the interindividual variability of indinavir. Ritonavir 42-51 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 26-31 17517050-10 2007 Through its inhibition of CYP3A and P-gp, ritonavir could attenuate the pharmacokinetic variability linked to genetic differences, reducing significantly the interindividual variability of indinavir. Ritonavir 42-51 ATP binding cassette subfamily B member 1 Homo sapiens 36-40 17646413-7 2007 High-dose (400 mg BID) ritonavir substantially reduced the steady-state mean voriconazole exposure (area under the concentration-time curve from 0 to 12 h [AUC(0-12)], -82%; maximum concentration [C(max)], -66%). Ritonavir 23-32 BH3 interacting domain death agonist Homo sapiens 18-21 17646413-8 2007 However, the effect of low-dose (100 mg BID) ritonavir was less pronounced (AUC(0-12), -39%; C(max), -24%). Ritonavir 45-54 BH3 interacting domain death agonist Homo sapiens 40-43 17646413-9 2007 The decrease in voriconazole exposure was probably due to the induction of CYP2C19 and CYP2C9 by ritonavir. Ritonavir 97-106 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 75-82 17646413-9 2007 The decrease in voriconazole exposure was probably due to the induction of CYP2C19 and CYP2C9 by ritonavir. Ritonavir 97-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 17646413-13 2007 Due to the significant effect of ritonavir on voriconazole exposure, coadministration of voriconazole with 400 mg BID ritonavir is contraindicated; coadministration with 100 mg BID ritonavir should be avoided, unless an assessment of the benefit/risk to the patient justifies the use. Ritonavir 118-127 BH3 interacting domain death agonist Homo sapiens 114-117 17646413-13 2007 Due to the significant effect of ritonavir on voriconazole exposure, coadministration of voriconazole with 400 mg BID ritonavir is contraindicated; coadministration with 100 mg BID ritonavir should be avoided, unless an assessment of the benefit/risk to the patient justifies the use. Ritonavir 118-127 BH3 interacting domain death agonist Homo sapiens 114-117 17719200-4 2007 We identified genes involved in cholesterol and fatty acid biosynthesis, as well as genes involved in fatty acid and cholesterol breakdown, whose expressions were regulated in opposite manners by ritonavir and bezafibrate, a hypolipidemic agonist of the peroxisome proliferator-activated receptor alpha. Ritonavir 196-205 peroxisome proliferator activated receptor alpha Rattus norvegicus 254-302 17700364-5 2007 RESULTS: Variants of ABCA1, APOA5, APOC3, APOE, and CETP contributed to plasma triglyceride levels, particularly in the setting of ritonavir-containing antiretroviral therapy. Ritonavir 131-140 cholesteryl ester transfer protein Homo sapiens 52-56 17683706-0 2007 [Insulin resistance in HIV-infected patients receiving long-term therapy with efavirenz, lopinavir/ritonavir and atazanavir]. Ritonavir 99-108 insulin Homo sapiens 1-8 17683706-7 2007 Insulin resistance was found in 5 (10.6%) patients, 4 among those receiving lopinavir/ritonavir, one among those treated with efavirenz and none among subjects receiving atazanavir (p = 0.065). Ritonavir 86-95 insulin Homo sapiens 0-7 17591975-4 2007 A clinically relevant concentration of ritonavir (15 mumol/L) significantly reduced cholesterol efflux from THP-1 and peripheral blood mononuclear cells to apolipoprotein A-I by 30 and 29%, respectively, as compared with controls. Ritonavir 39-48 GLI family zinc finger 2 Homo sapiens 108-113 17591975-4 2007 A clinically relevant concentration of ritonavir (15 mumol/L) significantly reduced cholesterol efflux from THP-1 and peripheral blood mononuclear cells to apolipoprotein A-I by 30 and 29%, respectively, as compared with controls. Ritonavir 39-48 apolipoprotein A1 Homo sapiens 156-174 17591975-5 2007 In addition, ritonavir significantly decreased the expression of scavenger receptor B1 and caveolin-1, whereas it significantly increased superoxide anion production and activated extracellular signal-regulated kinase (ERK) 1/2 in macrophages. Ritonavir 13-22 caveolin 1 Homo sapiens 91-101 17591975-5 2007 In addition, ritonavir significantly decreased the expression of scavenger receptor B1 and caveolin-1, whereas it significantly increased superoxide anion production and activated extracellular signal-regulated kinase (ERK) 1/2 in macrophages. Ritonavir 13-22 mitogen-activated protein kinase 3 Homo sapiens 180-227 17591975-7 2007 Consequently, the antioxidant seleno-l-methionine, the specific ERK1/2 inhibitor PD98059, or infection of a recombinant adenovirus encoding the dominant-negative form of ERK2 effectively blocked ritonavir-induced decrease of cholesterol efflux. Ritonavir 195-204 mitogen-activated protein kinase 3 Homo sapiens 64-70 17351982-0 2007 Doxorubicin cardiomyopathy via TLR-2 stimulation: potential for prevention using current anti-retroviral inhibitors such as ritonavir and nelfinavir. Ritonavir 124-133 toll like receptor 2 Homo sapiens 31-36 17591975-7 2007 Consequently, the antioxidant seleno-l-methionine, the specific ERK1/2 inhibitor PD98059, or infection of a recombinant adenovirus encoding the dominant-negative form of ERK2 effectively blocked ritonavir-induced decrease of cholesterol efflux. Ritonavir 195-204 mitogen-activated protein kinase 1 Homo sapiens 170-174 17591975-8 2007 Therefore, human immunodeficiency virus protease inhibitor ritonavir significantly inhibits cholesterol efflux from macrophages, which may be mediated by mitochondrial dysfunction, oxidative stress, ERK1/2 activation, and down-regulation of scavenger receptor B1 and caveolin-1. Ritonavir 59-68 mitogen-activated protein kinase 3 Homo sapiens 199-205 17591975-8 2007 Therefore, human immunodeficiency virus protease inhibitor ritonavir significantly inhibits cholesterol efflux from macrophages, which may be mediated by mitochondrial dysfunction, oxidative stress, ERK1/2 activation, and down-regulation of scavenger receptor B1 and caveolin-1. Ritonavir 59-68 caveolin 1 Homo sapiens 267-277 17594245-1 2007 A 14-year-old female with perinatally acquired HIV on boosted protease inhibitor (PI) therapy with atazanavir and ritonavir rapidly developed cushingoid features with excessive weight gain and moon facies within 2 weeks of receiving inhaled fluticasone/salmeterol for asthma treatment. Ritonavir 114-123 serpin family A member 13, pseudogene Homo sapiens 62-80 17409512-1 2007 Atazanavir (ATV) is a low oral bioavailability (BA) compound and, clinically, is generally coadministrated with ritonavir (RTV), which boosts the oral BA of ATV by inhibiting cytochrome P450 (CYP) 3A, and P-glycoprotein (Pgp) via the same metabolic pathway. Ritonavir 112-121 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 175-199 17472747-7 2007 The percent of total body weight gained during the treatment period was measured and confirmed that female LDL-R gained significantly less weight with ritonavir treatment than males. Ritonavir 151-160 low density lipoprotein receptor Mus musculus 107-112 17472747-11 2007 Ritonavir further suppressed leptin levels in (p < 0.05). Ritonavir 0-9 leptin Mus musculus 29-35 17461851-0 2007 Prediction of clinical benefits of ritonavir-boosted TMC114 from treatment effects on CD4 counts and HIV RNA. Ritonavir 35-44 CD4 molecule Homo sapiens 86-89 17409512-1 2007 Atazanavir (ATV) is a low oral bioavailability (BA) compound and, clinically, is generally coadministrated with ritonavir (RTV), which boosts the oral BA of ATV by inhibiting cytochrome P450 (CYP) 3A, and P-glycoprotein (Pgp) via the same metabolic pathway. Ritonavir 112-121 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 205-219 17409512-1 2007 Atazanavir (ATV) is a low oral bioavailability (BA) compound and, clinically, is generally coadministrated with ritonavir (RTV), which boosts the oral BA of ATV by inhibiting cytochrome P450 (CYP) 3A, and P-glycoprotein (Pgp) via the same metabolic pathway. Ritonavir 112-121 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 221-224 17409512-1 2007 Atazanavir (ATV) is a low oral bioavailability (BA) compound and, clinically, is generally coadministrated with ritonavir (RTV), which boosts the oral BA of ATV by inhibiting cytochrome P450 (CYP) 3A, and P-glycoprotein (Pgp) via the same metabolic pathway. Ritonavir 123-126 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 175-199 17409512-1 2007 Atazanavir (ATV) is a low oral bioavailability (BA) compound and, clinically, is generally coadministrated with ritonavir (RTV), which boosts the oral BA of ATV by inhibiting cytochrome P450 (CYP) 3A, and P-glycoprotein (Pgp) via the same metabolic pathway. Ritonavir 123-126 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 205-219 17409512-1 2007 Atazanavir (ATV) is a low oral bioavailability (BA) compound and, clinically, is generally coadministrated with ritonavir (RTV), which boosts the oral BA of ATV by inhibiting cytochrome P450 (CYP) 3A, and P-glycoprotein (Pgp) via the same metabolic pathway. Ritonavir 123-126 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 221-224 17713972-9 2007 Coadministration with small doses of the strong CYP3A4 inhibitor ritonavir results in an increase in darunavir bioavailability from 37% to 82%. Ritonavir 65-74 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 48-54 17240460-6 2007 Analysis of gene and protein expression determined a selectively increase of the pro-inflammatory cytokines monocyte chemoattractant protein (MCP)-1 and interleukin-8 (IL-8) following exposure to a pharmacological concentration of NFV and RTV. Ritonavir 239-242 C-C motif chemokine ligand 2 Homo sapiens 108-148 17240460-6 2007 Analysis of gene and protein expression determined a selectively increase of the pro-inflammatory cytokines monocyte chemoattractant protein (MCP)-1 and interleukin-8 (IL-8) following exposure to a pharmacological concentration of NFV and RTV. Ritonavir 239-242 C-X-C motif chemokine ligand 8 Homo sapiens 153-166 17240460-6 2007 Analysis of gene and protein expression determined a selectively increase of the pro-inflammatory cytokines monocyte chemoattractant protein (MCP)-1 and interleukin-8 (IL-8) following exposure to a pharmacological concentration of NFV and RTV. Ritonavir 239-242 C-X-C motif chemokine ligand 8 Homo sapiens 168-172 17148966-13 2007 CONCLUSIONS: Polymorphisms at MDR1-3435 significantly influence ATV plasma concentrations, as does being Caucasian patients with CT/TT genotypes, having lower ATV levels, even using ritonavir boosting. Ritonavir 182-191 ATP binding cassette subfamily B member 1 Homo sapiens 30-34 18997280-0 2007 Alpha-Tocopherol counteracts ritonavir-induced proinflammatory cytokines expression in differentiated THP-1 cells. Ritonavir 29-38 GLI family zinc finger 2 Homo sapiens 102-107 18997280-5 2007 Here, we report that after differentiating THP-1 cells to macrophages, ritonavir treatment (10 microg/mL) significantly increases expression of proinflammatory cytokines, IL-6, MCP-1 and IL-8, at both mRNA and protein levels. Ritonavir 71-80 GLI family zinc finger 2 Homo sapiens 43-48 18997280-5 2007 Here, we report that after differentiating THP-1 cells to macrophages, ritonavir treatment (10 microg/mL) significantly increases expression of proinflammatory cytokines, IL-6, MCP-1 and IL-8, at both mRNA and protein levels. Ritonavir 71-80 interleukin 6 Homo sapiens 171-175 18997280-5 2007 Here, we report that after differentiating THP-1 cells to macrophages, ritonavir treatment (10 microg/mL) significantly increases expression of proinflammatory cytokines, IL-6, MCP-1 and IL-8, at both mRNA and protein levels. Ritonavir 71-80 C-C motif chemokine ligand 2 Homo sapiens 177-182 18997280-5 2007 Here, we report that after differentiating THP-1 cells to macrophages, ritonavir treatment (10 microg/mL) significantly increases expression of proinflammatory cytokines, IL-6, MCP-1 and IL-8, at both mRNA and protein levels. Ritonavir 71-80 C-X-C motif chemokine ligand 8 Homo sapiens 187-191 18997280-6 2007 This ritonavir-induced effect is significantly suppressed by treatment of THP-1/macrophages with 50 muM alpha-T. Ritonavir 5-14 GLI family zinc finger 2 Homo sapiens 74-79 18997280-7 2007 We conclude that ritonavir can induce proinflammatory cytokines synthesis in THP-1/macrophages, which might be associated with the development of premature atherosclerosis in ritonavir-treated patients and that this effect is prevented by alpha-T. Ritonavir 17-26 GLI family zinc finger 2 Homo sapiens 77-82 18997280-7 2007 We conclude that ritonavir can induce proinflammatory cytokines synthesis in THP-1/macrophages, which might be associated with the development of premature atherosclerosis in ritonavir-treated patients and that this effect is prevented by alpha-T. Ritonavir 175-184 GLI family zinc finger 2 Homo sapiens 77-82 17201460-10 2007 The CLss/F of ritonavir was 40% lower in HCV+/FIB+patients than in HCV-patients (p=0.005) and 44% lower than in HCV+/FIB-patients (p=0.040). Ritonavir 14-23 LDL receptor related protein 4 Homo sapiens 4-8 17106066-1 2007 The protease inhibitor (PI) ritonavir (RTV) has been associated with elevated resting lipolytic rate, hyperlipidemia, and insulin resistance/glucose intolerance. Ritonavir 28-37 insulin Homo sapiens 122-129 17106066-1 2007 The protease inhibitor (PI) ritonavir (RTV) has been associated with elevated resting lipolytic rate, hyperlipidemia, and insulin resistance/glucose intolerance. Ritonavir 39-42 insulin Homo sapiens 122-129 17503669-5 2007 hOAT3 was more sensitive to Pls with ritonavir (RTV) and lopinavir being the most potent inhibitors of TFV transport (62% and 37% inhibition, respectively, at their Cmax). Ritonavir 37-46 solute carrier family 22 member 8 Homo sapiens 0-5 17503669-5 2007 hOAT3 was more sensitive to Pls with ritonavir (RTV) and lopinavir being the most potent inhibitors of TFV transport (62% and 37% inhibition, respectively, at their Cmax). Ritonavir 48-51 solute carrier family 22 member 8 Homo sapiens 0-5 17713972-11 2007 With regard to the necessary coadministration with low-dose ritonavir as a potent CYP3A4 inhibitor, coadministration of other substrates of CYP3A4 with darunavir/ritonavir requires caution or is even contraindicated. Ritonavir 60-69 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 82-88 17713972-11 2007 With regard to the necessary coadministration with low-dose ritonavir as a potent CYP3A4 inhibitor, coadministration of other substrates of CYP3A4 with darunavir/ritonavir requires caution or is even contraindicated. Ritonavir 60-69 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 140-146 17713972-11 2007 With regard to the necessary coadministration with low-dose ritonavir as a potent CYP3A4 inhibitor, coadministration of other substrates of CYP3A4 with darunavir/ritonavir requires caution or is even contraindicated. Ritonavir 162-171 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 140-146 17069100-10 2006 The percentage of IFN-gamma+ NK cells increased after 3 weeks (P = .03) and 3 months (P = .01) of RIT. Ritonavir 98-101 interferon gamma Homo sapiens 18-27 17375980-15 2007 However, ritonavir, a potent inhibitor of CYP3A4, does not affect the pharmacokinetics of escitalopram. Ritonavir 9-18 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 42-48 16861219-8 2006 Atazanavir and ritonavir also decreased CYP7A1 protein levels and bile acid biosynthesis, while amprenavir had no significant effect. Ritonavir 15-24 cytochrome P450 family 7 subfamily A member 1 Rattus norvegicus 40-46 16630656-4 2006 The treatment significantly modified expression of several apoptosis-regulating proteins, including upregulation of Bax or downregulation of Bcl-2 and Mcl-1 by BOR+RIT, as well as downregulation of Bcl-2 and XIAP by BOR+CAM. Ritonavir 164-167 BCL2 apoptosis regulator Homo sapiens 141-146 17048292-3 2006 DMP 323 proved to be a P-gp substrate in competition studies with P-gp inhibitor ritonavir and H17 as a representative of another class of non-peptidic HIV-1 protease inhibitors. Ritonavir 81-90 ATP binding cassette subfamily B member 1 Homo sapiens 23-27 17178259-4 2006 Simvastatin, lovastatin, and atorvastatin are metabolized by cytochrome P450 (CYP) 3A4 (simvastatin acid is also metabolized by CYP2C8); their plasma concentrations and risk of myotoxicity are greatly increased by strong inhibitors of CYP3A4 (eg, itraconazole and ritonavir). Ritonavir 264-273 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 61-86 17178259-4 2006 Simvastatin, lovastatin, and atorvastatin are metabolized by cytochrome P450 (CYP) 3A4 (simvastatin acid is also metabolized by CYP2C8); their plasma concentrations and risk of myotoxicity are greatly increased by strong inhibitors of CYP3A4 (eg, itraconazole and ritonavir). Ritonavir 264-273 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 128-134 16565735-12 2006 We confirm that RIT in chronic B-LPD can result in high and durable CR rates but with significant incidences of acute and chronic GVHD. Ritonavir 16-19 acyl-CoA synthetase bubblegum family member 1 Homo sapiens 33-36 16954722-7 2006 During euglycemic clamp, there was no significant change from baseline insulin sensitivity with atazanavir/ritonavir (P = 0.132), while insulin sensitivity significantly decreased with lopinavir/ritonavir from the baseline (-25%; P < 0.001) and from that seen with atazanavir/ritonavir (-18%; P = 0.023). Ritonavir 195-204 insulin Homo sapiens 136-143 16954722-8 2006 During OGTT, the HOMA insulin resistance index significantly increased from baseline at 120 min with atazanavir/ritonavir and at 150 min with lopinavir/ritonavir. Ritonavir 112-121 insulin Homo sapiens 22-29 16954722-8 2006 During OGTT, the HOMA insulin resistance index significantly increased from baseline at 120 min with atazanavir/ritonavir and at 150 min with lopinavir/ritonavir. Ritonavir 152-161 insulin Homo sapiens 22-29 16457885-0 2006 HIV-1 protease inhibitor ritonavir potentiates the effect of 1,25-dihydroxyvitamin D3 to induce growth arrest and differentiation of human myeloid leukemia cells via down-regulation of CYP24. Ritonavir 25-34 cytochrome P450 family 24 subfamily A member 1 Homo sapiens 185-190 16861604-8 2006 Treatment of mature adipocytes with ritonavir, but not atazanavir, also selectively decreased insulin signaling. Ritonavir 36-45 insulin Homo sapiens 94-101 16861604-9 2006 Moreover, ritonavir also selectively decreased expression of adiponectin, an insulin-sensitizing adipocytokine, while inducing interleukin-6, a proinflammatory cytokine implicated in insulin resistance. Ritonavir 10-19 insulin Homo sapiens 77-84 16861604-9 2006 Moreover, ritonavir also selectively decreased expression of adiponectin, an insulin-sensitizing adipocytokine, while inducing interleukin-6, a proinflammatory cytokine implicated in insulin resistance. Ritonavir 10-19 insulin Homo sapiens 183-190 16954722-0 2006 Effects of atazanavir/ritonavir and lopinavir/ritonavir on glucose uptake and insulin sensitivity: demonstrable differences in vitro and clinically. Ritonavir 46-55 insulin Homo sapiens 78-85 16954722-7 2006 During euglycemic clamp, there was no significant change from baseline insulin sensitivity with atazanavir/ritonavir (P = 0.132), while insulin sensitivity significantly decreased with lopinavir/ritonavir from the baseline (-25%; P < 0.001) and from that seen with atazanavir/ritonavir (-18%; P = 0.023). Ritonavir 195-204 insulin Homo sapiens 136-143 16720753-8 2006 [(3)H]Taurocholate transport by recombinant NTCP and Ntcp was inhibited by ritonavir (IC(50) = 2.1 and 6.4 microM in human and rat, respectively), saquinavir (IC(50) = 6.7 and 20 microM, respectively), and efavirenz (IC(50) = 43 and 97 microM, respectively). Ritonavir 75-84 solute carrier family 10 member 1 Rattus norvegicus 44-48 16720753-8 2006 [(3)H]Taurocholate transport by recombinant NTCP and Ntcp was inhibited by ritonavir (IC(50) = 2.1 and 6.4 microM in human and rat, respectively), saquinavir (IC(50) = 6.7 and 20 microM, respectively), and efavirenz (IC(50) = 43 and 97 microM, respectively). Ritonavir 75-84 solute carrier family 10 member 1 Rattus norvegicus 53-57 16890574-0 2006 Potent cytochrome P450 2C19 genotype-related interaction between voriconazole and the cytochrome P450 3A4 inhibitor ritonavir. Ritonavir 116-125 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 7-27 16890574-0 2006 Potent cytochrome P450 2C19 genotype-related interaction between voriconazole and the cytochrome P450 3A4 inhibitor ritonavir. Ritonavir 116-125 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 86-105 16545584-12 2006 The cells demonstrated P-glycoprotein-mediated function by directional transport of dexamethasone, ritonavir, and vinblastine in a transwell assay that was inhibited in the presence of cyclosporin A, verapamil, or quinidine. Ritonavir 99-108 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 23-37 16639344-0 2006 Lopinavir/ritonavir induces the hepatic activity of cytochrome P450 enzymes CYP2C9, CYP2C19, and CYP1A2 but inhibits the hepatic and intestinal activity of CYP3A as measured by a phenotyping drug cocktail in healthy volunteers. Ritonavir 10-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 16639344-0 2006 Lopinavir/ritonavir induces the hepatic activity of cytochrome P450 enzymes CYP2C9, CYP2C19, and CYP1A2 but inhibits the hepatic and intestinal activity of CYP3A as measured by a phenotyping drug cocktail in healthy volunteers. Ritonavir 10-19 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 84-91 16639344-0 2006 Lopinavir/ritonavir induces the hepatic activity of cytochrome P450 enzymes CYP2C9, CYP2C19, and CYP1A2 but inhibits the hepatic and intestinal activity of CYP3A as measured by a phenotyping drug cocktail in healthy volunteers. Ritonavir 10-19 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 97-103 16639344-0 2006 Lopinavir/ritonavir induces the hepatic activity of cytochrome P450 enzymes CYP2C9, CYP2C19, and CYP1A2 but inhibits the hepatic and intestinal activity of CYP3A as measured by a phenotyping drug cocktail in healthy volunteers. Ritonavir 10-19 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 156-161 16507617-13 2006 QT interval increases were observed with buprenorphine/naloxone in combination with either delavirdine or ritonavir, which inhibit CYP3A4. Ritonavir 106-115 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 131-137 16551874-0 2006 Effects of HIV protease inhibitor ritonavir on Akt-regulated cell proliferation in breast cancer. Ritonavir 34-43 AKT serine/threonine kinase 1 Homo sapiens 47-50 16551874-6 2006 Ritonavir depletes ER-alpha levels notably in ER-positive lines. Ritonavir 0-9 estrogen receptor 1 Homo sapiens 19-27 16551874-7 2006 Ritonavir causes G1 arrest, depletes cyclin-dependent kinases 2, 4, and 6 and cyclin D1 but not cyclin E, and depletes phosphorylated Rb and Ser473 Akt. Ritonavir 0-9 cyclin D1 Homo sapiens 78-87 16551874-9 2006 Myristoyl-Akt, but not activated K-Ras, rescues ritonavir inhibition. Ritonavir 48-57 AKT serine/threonine kinase 1 Homo sapiens 10-13 16551874-10 2006 Ritonavir inhibited a MDA-MB-231 xenograft and intratumoral Akt activity at a clinically attainable serum Cmax of 22 +/- 8 micromol/L. Ritonavir 0-9 AKT serine/threonine kinase 1 Homo sapiens 60-63 16551874-11 2006 Because heat shock protein 90 (Hsp90) substrates are depleted by ritonavir, ritonavir effects on Hsp90 were tested. Ritonavir 76-85 heat shock protein 90 alpha family class A member 1 Homo sapiens 97-102 16551874-12 2006 Ritonavir binds Hsp90 (K(D), 7.8 micromol/L) and partially inhibits its chaperone function. Ritonavir 0-9 heat shock protein 90 alpha family class A member 1 Homo sapiens 16-21 16551874-13 2006 Ritonavir blocks association of Hsp90 with Akt and, with sustained exposure, notably depletes Hsp90. Ritonavir 0-9 heat shock protein 90 alpha family class A member 1 Homo sapiens 32-37 16551874-13 2006 Ritonavir blocks association of Hsp90 with Akt and, with sustained exposure, notably depletes Hsp90. Ritonavir 0-9 AKT serine/threonine kinase 1 Homo sapiens 43-46 16551874-13 2006 Ritonavir blocks association of Hsp90 with Akt and, with sustained exposure, notably depletes Hsp90. Ritonavir 0-9 heat shock protein 90 alpha family class A member 1 Homo sapiens 94-99 16551874-14 2006 Stably expressed Hsp90alpha short hairpin RNA also depletes Hsp90, inhibiting proliferation and sensitizing breast cancer cells to low ritonavir concentrations. Ritonavir 135-144 heat shock protein 90 alpha family class A member 1 Homo sapiens 17-27 16551874-14 2006 Stably expressed Hsp90alpha short hairpin RNA also depletes Hsp90, inhibiting proliferation and sensitizing breast cancer cells to low ritonavir concentrations. Ritonavir 135-144 heat shock protein 90 alpha family class A member 1 Homo sapiens 17-22 16551874-15 2006 CONCLUSIONS: Ritonavir inhibits breast cancer growth in part by inhibiting Hsp90 substrates, including Akt. Ritonavir 13-22 heat shock protein 90 alpha family class A member 1 Homo sapiens 75-80 16551874-15 2006 CONCLUSIONS: Ritonavir inhibits breast cancer growth in part by inhibiting Hsp90 substrates, including Akt. Ritonavir 13-22 AKT serine/threonine kinase 1 Homo sapiens 103-106 16551874-16 2006 Ritonavir may be of interest for breast cancer therapeutics and its efficacy may be increased by sustained exposure or Hsp90 RNA interference. Ritonavir 0-9 heat shock protein 90 alpha family class A member 1 Homo sapiens 119-124 16757288-2 2006 We describe the effect of the coadministration of Amprenavir/Ritonavir (APV/r) and FosAmprenavir (FosAPV) on cyclosporine (CsA) concentrations in two patients receiving OLT for end-stage liver disease due to hepatitis C Virus. Ritonavir 61-70 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 123-126 16495253-6 2006 For FPV 1,400 mg BID plus RTV 200 mg BID, the values for plasma APV AUC(0-tau), C(max), and C(tau) were 26, 48, and 32% higher, respectively, and the values for plasma RTV AUC(0-tau), C(max), and C(tau) increased 4.15-fold, 4.17-fold, and 3.99-fold, respectively, compared to those for FPV 700 mg BID plus RTV 100 mg BID. Ritonavir 168-171 BH3 interacting domain death agonist Homo sapiens 17-20 16485915-11 2006 Furthermore, fosamprenavir is commonly administered in combination with low-dose ritonavir, which is also extensively metabolised by CYP3A4, and is a more potent CYP3A4 inhibitor than amprenavir. Ritonavir 81-90 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 133-139 16495253-6 2006 For FPV 1,400 mg BID plus RTV 200 mg BID, the values for plasma APV AUC(0-tau), C(max), and C(tau) were 26, 48, and 32% higher, respectively, and the values for plasma RTV AUC(0-tau), C(max), and C(tau) increased 4.15-fold, 4.17-fold, and 3.99-fold, respectively, compared to those for FPV 700 mg BID plus RTV 100 mg BID. Ritonavir 168-171 BH3 interacting domain death agonist Homo sapiens 37-40 16495253-6 2006 For FPV 1,400 mg BID plus RTV 200 mg BID, the values for plasma APV AUC(0-tau), C(max), and C(tau) were 26, 48, and 32% higher, respectively, and the values for plasma RTV AUC(0-tau), C(max), and C(tau) increased 4.15-fold, 4.17-fold, and 3.99-fold, respectively, compared to those for FPV 700 mg BID plus RTV 100 mg BID. Ritonavir 168-171 BH3 interacting domain death agonist Homo sapiens 37-40 16495253-6 2006 For FPV 1,400 mg BID plus RTV 200 mg BID, the values for plasma APV AUC(0-tau), C(max), and C(tau) were 26, 48, and 32% higher, respectively, and the values for plasma RTV AUC(0-tau), C(max), and C(tau) increased 4.15-fold, 4.17-fold, and 3.99-fold, respectively, compared to those for FPV 700 mg BID plus RTV 100 mg BID. Ritonavir 168-171 BH3 interacting domain death agonist Homo sapiens 37-40 16174765-2 2006 We examined the effect of ritonavir, an HIV protease inhibitor, on HTLV-I-infected T-cell lines and primary ATL cells and found that it induced apoptosis and inhibited transcriptional activation of NF-kappaB in these cells. Ritonavir 26-35 nuclear factor kappa B subunit 1 Homo sapiens 198-207 16174765-3 2006 Furthermore, ritonavir inhibited expression of Bcl-xL, survivin, c-Myc, and cyclin D2, the targets of NF-kappaB. Ritonavir 13-22 BCL2 like 1 Homo sapiens 47-53 16174765-3 2006 Furthermore, ritonavir inhibited expression of Bcl-xL, survivin, c-Myc, and cyclin D2, the targets of NF-kappaB. Ritonavir 13-22 MYC proto-oncogene, bHLH transcription factor Homo sapiens 65-70 16174765-3 2006 Furthermore, ritonavir inhibited expression of Bcl-xL, survivin, c-Myc, and cyclin D2, the targets of NF-kappaB. Ritonavir 13-22 cyclin D2 Homo sapiens 76-85 16174765-3 2006 Furthermore, ritonavir inhibited expression of Bcl-xL, survivin, c-Myc, and cyclin D2, the targets of NF-kappaB. Ritonavir 13-22 nuclear factor kappa B subunit 1 Homo sapiens 102-111 16174765-4 2006 In nonobese diabetic/severe combined immunodeficient (NOD/SCID)/gammacnull (NOG) mice, ritonavir very efficiently prevented tumor growth and leukemic infiltration in various organs of NOG mice at the same dose used for treatment of patients with AIDS. Ritonavir 87-96 noggin Mus musculus 76-79 16174765-4 2006 In nonobese diabetic/severe combined immunodeficient (NOD/SCID)/gammacnull (NOG) mice, ritonavir very efficiently prevented tumor growth and leukemic infiltration in various organs of NOG mice at the same dose used for treatment of patients with AIDS. Ritonavir 87-96 noggin Mus musculus 184-187 16174765-5 2006 Our data indicate that ritonavir has potent anti-NF-kappaB and antitumor effects and might be clinically applicable for treatment of ATL. Ritonavir 23-32 nuclear factor kappa B subunit 1 Homo sapiens 49-58 16269669-0 2006 Ritonavir impairs lipoprotein lipase-mediated lipolysis and decreases uptake of fatty acids in adipose tissue. Ritonavir 0-9 lipase, endothelial Mus musculus 30-36 16269669-8 2006 CONCLUSIONS: We conclude that RTV causes hypertriglyceridemia via decreased lipoprotein lipase-mediated clearance of VLDL-TG. Ritonavir 30-33 lipase, endothelial Mus musculus 88-94 16432264-4 2006 Moreover, medications prescribed to HIV-positive patients may also be CYP3A inhibitors and inducers: Tipranavir, in the absence of ritonavir, is a CYP3A inducer, and ritonavir is a CYP3A inhibitor. Ritonavir 166-175 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 70-75 16338282-0 2005 Effect of low-dose ritonavir (100 mg twice daily) on the activity of cytochrome P450 2D6 in healthy volunteers. Ritonavir 19-28 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 69-88 16322314-11 2005 to mice in the presence and absence of the CYP3A and ABCG2 inhibitor, ritonavir, there was an increase in BPU plasma exposure and decrease in metabolite exposure but no overall change in cumulative exposure to BPU and the cytotoxic metabolites. Ritonavir 70-79 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 43-48 16940746-11 2006 Histamine release and production of IL-4 and IL-13 by basophils before the treatment correlated with the severity of adverse events during the incremental phase of RIT. Ritonavir 164-167 interleukin 4 Homo sapiens 36-40 16940746-11 2006 Histamine release and production of IL-4 and IL-13 by basophils before the treatment correlated with the severity of adverse events during the incremental phase of RIT. Ritonavir 164-167 interleukin 13 Homo sapiens 45-50 16940746-12 2006 CONCLUSION: We report the decrease in blood basophil numbers, their lower activation status and the reduced production of IL-4 and IL-13 early in the course of RIT. Ritonavir 160-163 interleukin 4 Homo sapiens 122-126 16940746-12 2006 CONCLUSION: We report the decrease in blood basophil numbers, their lower activation status and the reduced production of IL-4 and IL-13 early in the course of RIT. Ritonavir 160-163 interleukin 13 Homo sapiens 131-136 16387596-3 2006 OBJECTIVES: We hypothesized that omalizumab, a humanized monoclonal anti-IgE antibody, would be effective in enhancing both safety and efficacy of RIT. Ritonavir 147-150 immunoglobulin heavy constant epsilon Homo sapiens 73-76 16322314-11 2005 to mice in the presence and absence of the CYP3A and ABCG2 inhibitor, ritonavir, there was an increase in BPU plasma exposure and decrease in metabolite exposure but no overall change in cumulative exposure to BPU and the cytotoxic metabolites. Ritonavir 70-79 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 53-58 16322314-12 2005 Thus, we conclude that (a) CYP3A4 and CYP1A1 are the predominant cytochrome P450 enzymes that catalyze BPU metabolism, (b) BPU is metabolized to two cytotoxic and four noncytotoxic metabolites, and (c) ritonavir inhibits BPU metabolism to improve the systemic exposure to BPU without altering cumulative exposure to BPU and the cytotoxic metabolites. Ritonavir 202-211 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 27-33 16338282-2 2005 When applied in a therapeutic dose (600 mg twice daily), ritonavir also inhibits CYP2D6. Ritonavir 57-66 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 81-87 16322314-12 2005 Thus, we conclude that (a) CYP3A4 and CYP1A1 are the predominant cytochrome P450 enzymes that catalyze BPU metabolism, (b) BPU is metabolized to two cytotoxic and four noncytotoxic metabolites, and (c) ritonavir inhibits BPU metabolism to improve the systemic exposure to BPU without altering cumulative exposure to BPU and the cytotoxic metabolites. Ritonavir 202-211 cytochrome P450 family 1 subfamily A member 1 Homo sapiens 38-44 16322314-12 2005 Thus, we conclude that (a) CYP3A4 and CYP1A1 are the predominant cytochrome P450 enzymes that catalyze BPU metabolism, (b) BPU is metabolized to two cytotoxic and four noncytotoxic metabolites, and (c) ritonavir inhibits BPU metabolism to improve the systemic exposure to BPU without altering cumulative exposure to BPU and the cytotoxic metabolites. Ritonavir 202-211 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-80 16338282-3 2005 The effect of low-dose ritonavir on CYP2D6 is unknown and was investigated in this study. Ritonavir 23-32 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 36-42 16338282-9 2005 CONCLUSIONS: Low-dose ritonavir (100 mg twice daily) exerts a modest inhibitory effect on the activity of CYP2D6 in extensive metabolizers, as assessed with desipramine as the index substrate. Ritonavir 22-31 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 106-112 16338282-11 2005 It is expected that the effect of low-dose ritonavir on CYP2D6 will not require standard dose reductions for CYP2D6 substrates. Ritonavir 43-52 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 56-62 16309240-8 2005 Western blot analysis showed that apoptosis was induced by an increased expression of Bax and decreased expression of Bcl-2 after treatment with Ritonavir and ionizing radiation. Ritonavir 145-154 BCL2-associated X protein Mus musculus 86-89 16309240-8 2005 Western blot analysis showed that apoptosis was induced by an increased expression of Bax and decreased expression of Bcl-2 after treatment with Ritonavir and ionizing radiation. Ritonavir 145-154 B cell leukemia/lymphoma 2 Mus musculus 118-123 16278782-6 2005 In human adipocytes, ritonavir at therapeutic concentrations inhibited insulin-stimulated lipogenesis, reduced GLUT4 mRNA, fatty acid synthase and adiponectin expression, while increasing IL-6 mRNA expression. Ritonavir 21-30 insulin Homo sapiens 71-78 16174444-11 2005 CONCLUSION: The results indicate that [(131)I]-labeled anti-CEA MoAbs can be effective in RIT on colonic cancers. Ritonavir 90-93 carcinoembryonic antigen gene family Mus musculus 60-63 16163639-9 2005 The mean ALT level increased by 45 U/L at 24 weeks and 18 U/L at 48 weeks in the nelfinavir group but decreased by 18 U/L at 24 weeks and 7 U/L at 48 weeks in the lopinavir-ritonavir group. Ritonavir 173-182 glutamic pyruvic transaminase, soluble Mus musculus 9-12 16162970-8 2005 Potent cytotoxic activity of ET-743 after 120 h treatment was observed, which could be increased in combination with the CYP inhibitors metyrapone (3A4), phenanthrene (substrate for 2E1, 3A4), piperonyl butoxide (3A), proadifen (2C9, 2E1, 3A4), ritonavir (3A4), and warfarin (2C9, 2C19). Ritonavir 245-254 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-124 16278782-6 2005 In human adipocytes, ritonavir at therapeutic concentrations inhibited insulin-stimulated lipogenesis, reduced GLUT4 mRNA, fatty acid synthase and adiponectin expression, while increasing IL-6 mRNA expression. Ritonavir 21-30 solute carrier family 2 member 4 Homo sapiens 111-116 16278782-6 2005 In human adipocytes, ritonavir at therapeutic concentrations inhibited insulin-stimulated lipogenesis, reduced GLUT4 mRNA, fatty acid synthase and adiponectin expression, while increasing IL-6 mRNA expression. Ritonavir 21-30 fatty acid synthase Homo sapiens 123-142 16278782-6 2005 In human adipocytes, ritonavir at therapeutic concentrations inhibited insulin-stimulated lipogenesis, reduced GLUT4 mRNA, fatty acid synthase and adiponectin expression, while increasing IL-6 mRNA expression. Ritonavir 21-30 adiponectin, C1Q and collagen domain containing Homo sapiens 147-158 16278782-6 2005 In human adipocytes, ritonavir at therapeutic concentrations inhibited insulin-stimulated lipogenesis, reduced GLUT4 mRNA, fatty acid synthase and adiponectin expression, while increasing IL-6 mRNA expression. Ritonavir 21-30 interleukin 6 Homo sapiens 188-192 16278782-8 2005 All together, these data show effects of ritonavir on human preadipocytes and adipocytes aiming at reducing adipose tissue mass and increasing insulin resistance. Ritonavir 41-50 insulin Homo sapiens 143-150 16388722-11 2005 Lopinavir/ritonavir-treated children displayed higher CD4+ T-lymphocyte counts than saquinavir-treated children since the first month of therapy (week 4: P=0.042; week 24: P= 0.029) while nelfinavir-treated children took 24 weeks to reach such an outcome (P=0.034). Ritonavir 10-19 CD4 molecule Homo sapiens 54-57 16078135-6 2005 The rank order potency for MRP-related drug efflux transporters, was nelfinavir>ritonavir>saquinavir>amprenavir>indinavir. Ritonavir 83-92 ATP binding cassette subfamily C member 1 Homo sapiens 27-30 16388722-12 2005 Since lopinavir/ritonavir-based regimen controls viral replication more efficiently and restores CD4+ T-lymphocyte count more quickly than saquinavir- or nelfinavir-based HAART, it may be considered when a salvage therapy or a rapid increase in CD4+ T-lymphocytes is necessary. Ritonavir 16-25 CD4 molecule Homo sapiens 97-100 16388722-12 2005 Since lopinavir/ritonavir-based regimen controls viral replication more efficiently and restores CD4+ T-lymphocyte count more quickly than saquinavir- or nelfinavir-based HAART, it may be considered when a salvage therapy or a rapid increase in CD4+ T-lymphocytes is necessary. Ritonavir 16-25 CD4 molecule Homo sapiens 245-248 16105660-4 2005 Ritonavir significantly reduced the mRNA levels of tight junction proteins zonula occluden-1, occludin, and claudin-1 by 40-60% as compared to controls (P<0.05) by real-time PCR analysis. Ritonavir 0-9 occludin Homo sapiens 94-102 16105660-4 2005 Ritonavir significantly reduced the mRNA levels of tight junction proteins zonula occluden-1, occludin, and claudin-1 by 40-60% as compared to controls (P<0.05) by real-time PCR analysis. Ritonavir 0-9 claudin 1 Homo sapiens 108-117 16105660-8 2005 Furthermore, ritonavir activated ERK1/2 (phosphorylation), but not P38 and JNK. Ritonavir 13-22 mitogen-activated protein kinase 3 Homo sapiens 33-39 16105660-9 2005 Specific ERK1/2 inhibitor, PD89059, significantly abolished ritonavir-induced endothelial permeability by 92%. Ritonavir 60-69 mitogen-activated protein kinase 3 Homo sapiens 9-15 16105660-11 2005 ERK1/2 activation is involved in the signal transduction pathway of ritonavir-induced endothelial permeability. Ritonavir 68-77 mitogen-activated protein kinase 3 Homo sapiens 0-6 15860655-8 2005 Direct comparison of known mechanism-based inactivators and quasi-irreversible inhibitors, based on our screening of apparent partition ratios, has identified ritonavir, mibefradil, and azamulin as highly effective mechanism-based inactivators; e.g., 1 mol of CYP3A4 was inactivated on turnover of about 2 mol of compound. Ritonavir 159-168 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 260-266 15755851-1 2005 Ritonavir, a protease inhibitor (PI), is a potent inhibitor of cytochrome P450 3A4. Ritonavir 0-9 serpin family A member 13, pseudogene Homo sapiens 13-36 15755851-1 2005 Ritonavir, a protease inhibitor (PI), is a potent inhibitor of cytochrome P450 3A4. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 63-82 15755851-3 2005 Six patients with preexisting HIV-lipodystrophy developed symptomatic Cushing"s syndrome when treated with inhaled fluticasone at varying doses for asthma while concurrently treated with low-dose ritonavir-boosted PI antiretroviral therapy (ART) regimens for HIV infection. Ritonavir 196-205 serpin family A member 13, pseudogene Homo sapiens 214-216 15629450-2 2005 We have investigated these claims with an in vitro study of the effect of ritonavir on the m-calpain and mu-calpain isoforms. Ritonavir 74-83 calpain 2 Homo sapiens 91-100 15764714-9 2005 At 30 microM and above, acute atazanavir exposure reversed P-gp induction caused by 3-day pretreatment with 10 microM ritonavir. Ritonavir 118-127 ATP binding cassette subfamily B member 1 Homo sapiens 59-63 15910662-12 2005 This population analysis in patients with sustained virological response, quantified the effect of ritonavir on the absorption rate constant and on the clearance of indinavir, showed an increase of Cl/F in men and can be used to draw reference curve for therapeutic drug monitoring. Ritonavir 99-108 crooked neck pre-mRNA splicing factor 1 Homo sapiens 198-202 15905730-6 2005 In response to bradykinin at 10 M, ritonavir (15 and 30 microM) reduced the relaxation by 27% and 78%, respectively, as compared with controls (P < 0.05). Ritonavir 35-44 kininogen 1 Homo sapiens 15-25 15905730-9 2005 The eNOS mRNA showed a 54% and 65% reduction for 15- and 30-microM ritonavir-treated rings, respectively (P < 0.05). Ritonavir 67-76 nitric oxide synthase 3 Homo sapiens 4-8 15905730-12 2005 HCAECs treated with ritonavir showed significant reductions in mRNA and protein levels of eNOS (P < 0.05). Ritonavir 20-29 nitric oxide synthase 3 Homo sapiens 90-94 15905730-13 2005 Thus, ritonavir significantly impairs vasomotor function and reduces eNOS expression in porcine coronary artery endothelial cells as well as HCAECs. Ritonavir 6-15 nitric oxide synthase 3 Homo sapiens 69-73 15809899-4 2005 However, when treated with ritonavir, individuals with unfavorable genotypes of APOC3 and [corrected] APOE were at risk of extreme hypertriglyceridemia. Ritonavir 27-36 apolipoprotein C3 Homo sapiens 80-85 15809899-4 2005 However, when treated with ritonavir, individuals with unfavorable genotypes of APOC3 and [corrected] APOE were at risk of extreme hypertriglyceridemia. Ritonavir 27-36 apolipoprotein E Homo sapiens 102-106 15809899-6 2005 The net effect of the APOE*APOC3*ritonavir interaction was an increase in plasma triglyceride levels of 2.23 mmol/L. Ritonavir 33-42 apolipoprotein C3 Homo sapiens 27-32 15780763-0 2005 CD36 overexpression in ritonavir-treated THP-1 cells is reversed by alpha-tocopherol. Ritonavir 23-32 GLI family zinc finger 2 Homo sapiens 41-46 15780763-3 2005 We show here that treatment of THP-1 monocytes with ritonavir increases total protein and surface expression of CD36; however, only weak changes are observed at the mRNA level, suggesting that CD36 overexpression occurs mainly at the posttranscriptional level. Ritonavir 52-61 GLI family zinc finger 2 Homo sapiens 31-36 15780763-4 2005 Concentrations of ritonavir that upregulate CD36 expression inhibit proteasome activity in THP-1 cells, indicating a possible regulatory role of the proteasome in CD36 overexpression. Ritonavir 18-27 GLI family zinc finger 2 Homo sapiens 91-96 15780763-7 2005 Furthermore, in THP-1 monocytes, alpha-tocopherol reverses the proteasome activity inhibited by ritonavir. Ritonavir 96-105 GLI family zinc finger 2 Homo sapiens 16-21 15780763-8 2005 This study indicates that an increased CD36 protein expression in THP-1 monocytes induced by ritonavir can be normalized by alpha-tocopherol. Ritonavir 93-102 GLI family zinc finger 2 Homo sapiens 66-71 15983895-0 2005 Pilot study of saquinavir and lopinavir/ritonavir twice daily in protease inhibitor-naive HIV-positive patients. Ritonavir 40-49 serpin family A member 13, pseudogene Homo sapiens 65-83 15983895-13 2005 CONCLUSION: Our study demonstrates that saquinavir/lopinavir/ritonavir 1000/400/100 mg bid with tenofovir intensification is a potent nucleoside-sparing regimen for PI-naive patients, associated with durable HIV suppression and improved CD4 cell counts. Ritonavir 61-70 CD4 molecule Homo sapiens 237-240 15721475-5 2005 Drug-induced QT prolongation is usually caused by block of human ether-a-go-go-related gene (HERG) potassium channels, and we showed that lopinavir, nelfinavir, ritonavir, and saquinavir caused dose-dependent block of HERG channels heterologously expressed in HEK293 cells in vitro. Ritonavir 161-170 potassium voltage-gated channel subfamily H member 2 Homo sapiens 93-97 15721475-5 2005 Drug-induced QT prolongation is usually caused by block of human ether-a-go-go-related gene (HERG) potassium channels, and we showed that lopinavir, nelfinavir, ritonavir, and saquinavir caused dose-dependent block of HERG channels heterologously expressed in HEK293 cells in vitro. Ritonavir 161-170 potassium voltage-gated channel subfamily H member 2 Homo sapiens 218-222 15525648-12 2005 Because ritonavir can alter the expression of insulin resistance-related cytokines in human adipocytes in a way parallel to the situation observed in vivo upon treatment of HIV-infected patients, we propose that protease inhibitors participate in insulin resistance through a direct effect on adipocytes. Ritonavir 8-17 insulin Homo sapiens 46-53 15657782-7 2005 CYP3A activity was lower in the efavirenz + ritonavir group (P = 0.01) and in the ritonavir group (P = 0.04) than in the nelfinavir group, although already strongly inhibited in the latter. Ritonavir 44-53 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-5 15657782-7 2005 CYP3A activity was lower in the efavirenz + ritonavir group (P = 0.01) and in the ritonavir group (P = 0.04) than in the nelfinavir group, although already strongly inhibited in the latter. Ritonavir 82-91 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-5 15657782-11 2005 The inhibition of CYP3A by ritonavir or nelfinavir offsets the inductive effects of efavirenz or nevirapine administered concomitantly. Ritonavir 27-36 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 18-23 15668539-0 2005 Combining fosamprenavir with lopinavir/ritonavir substantially reduces amprenavir and lopinavir exposure: ACTG protocol A5143 results. Ritonavir 39-48 actin gamma 1 Homo sapiens 106-110 15668549-3 2005 There was a median 2.5 log10 reduction in HIV RNA and 115 cell increase in CD4 cell counts after 4-8 weeks of saquinavir/ritonavir monotherapy. Ritonavir 121-130 CD4 molecule Homo sapiens 75-78 15523003-4 2005 Ritonavir was the most potent (K(I) = 0.10 and 0.17 microM) and demonstrated high k(inact) values (0.32 and 0.40 min(-1)) with both CYP3A4(+b(5)) and HLMs. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 132-138 15523003-6 2005 For CYP3A5, nelfinavir exhibited the highest k(inact) (0.47 min(-1)), but ritonavir was the most potent (K(I) = 0.12 microM). Ritonavir 74-83 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 4-10 15525648-12 2005 Because ritonavir can alter the expression of insulin resistance-related cytokines in human adipocytes in a way parallel to the situation observed in vivo upon treatment of HIV-infected patients, we propose that protease inhibitors participate in insulin resistance through a direct effect on adipocytes. Ritonavir 8-17 insulin Homo sapiens 247-254 16152760-8 2005 In the PI group, patients on ritonavir (RTV)-containing regimens had a lower LDL apoB ASR (P=0.009) and a trend to a lower LDL apoB FCR and increased RT compared with non-RTV-containing PI regimens (P=0.05). Ritonavir 40-43 apolipoprotein B Homo sapiens 127-131 15865214-6 2005 RESULTS: Nelfinavir, ritonavir and saquinavir inhibited adipogenesis and up-regulated the expression of TNF-alpha and IL-6, but this effect was not seen with indinavir, zidovudine and stavudine. Ritonavir 21-30 tumor necrosis factor Mus musculus 104-113 15865214-6 2005 RESULTS: Nelfinavir, ritonavir and saquinavir inhibited adipogenesis and up-regulated the expression of TNF-alpha and IL-6, but this effect was not seen with indinavir, zidovudine and stavudine. Ritonavir 21-30 interleukin 6 Mus musculus 118-122 15865214-7 2005 Adiponectin expression was significantly reduced in both NRTI- and PI-treated cells, although the most profound reductions were found with ritonavir and saquinavir. Ritonavir 139-148 adiponectin, C1Q and collagen domain containing Mus musculus 0-11 15865214-9 2005 CONCLUSIONS: Our data suggest that the PIs nelfinavir, ritonavir and saquinavir have potent effects in inhibiting adipocyte differentiation whilst up-regulating TNF-alpha and IL-6 mRNA levels and decreasing adiponectin levels. Ritonavir 55-64 tumor necrosis factor Mus musculus 161-170 15865214-9 2005 CONCLUSIONS: Our data suggest that the PIs nelfinavir, ritonavir and saquinavir have potent effects in inhibiting adipocyte differentiation whilst up-regulating TNF-alpha and IL-6 mRNA levels and decreasing adiponectin levels. Ritonavir 55-64 interleukin 6 Mus musculus 175-179 15865214-9 2005 CONCLUSIONS: Our data suggest that the PIs nelfinavir, ritonavir and saquinavir have potent effects in inhibiting adipocyte differentiation whilst up-regulating TNF-alpha and IL-6 mRNA levels and decreasing adiponectin levels. Ritonavir 55-64 adiponectin, C1Q and collagen domain containing Mus musculus 207-218 16430196-13 2005 CONCLUSION: Our study demonstrates that indinavir/ritonavir 400/100 mg plus stavudine and lamivudine twice daily, the least expensive boosted protease inhibitor, appears to be effective and safe up to 96 weeks despite high baseline viraemia and low CD4+ cell count in antiretroviral-naive patients. Ritonavir 50-59 CD4 molecule Homo sapiens 249-252 16152760-8 2005 In the PI group, patients on ritonavir (RTV)-containing regimens had a lower LDL apoB ASR (P=0.009) and a trend to a lower LDL apoB FCR and increased RT compared with non-RTV-containing PI regimens (P=0.05). Ritonavir 29-38 apolipoprotein B Homo sapiens 81-85 16152760-8 2005 In the PI group, patients on ritonavir (RTV)-containing regimens had a lower LDL apoB ASR (P=0.009) and a trend to a lower LDL apoB FCR and increased RT compared with non-RTV-containing PI regimens (P=0.05). Ritonavir 29-38 apolipoprotein B Homo sapiens 127-131 16152760-8 2005 In the PI group, patients on ritonavir (RTV)-containing regimens had a lower LDL apoB ASR (P=0.009) and a trend to a lower LDL apoB FCR and increased RT compared with non-RTV-containing PI regimens (P=0.05). Ritonavir 40-43 apolipoprotein B Homo sapiens 81-85 15635177-0 2005 Effect of chronic administration of ritonavir on function of cytochrome P450 3A and P-glycoprotein in rats. Ritonavir 36-45 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 84-98 15635177-1 2005 Ritonavir (RTV) is well known as an inhibitor of many drugs that are metabolized by cytochrome P450 (CYP) 3A or fluxed via P-glycoprotein (Pgp), although it is also reported that RTV is a potent inducer for them. Ritonavir 0-9 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 84-108 15635177-1 2005 Ritonavir (RTV) is well known as an inhibitor of many drugs that are metabolized by cytochrome P450 (CYP) 3A or fluxed via P-glycoprotein (Pgp), although it is also reported that RTV is a potent inducer for them. Ritonavir 0-9 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 123-137 15635177-1 2005 Ritonavir (RTV) is well known as an inhibitor of many drugs that are metabolized by cytochrome P450 (CYP) 3A or fluxed via P-glycoprotein (Pgp), although it is also reported that RTV is a potent inducer for them. Ritonavir 0-9 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 139-142 15635177-1 2005 Ritonavir (RTV) is well known as an inhibitor of many drugs that are metabolized by cytochrome P450 (CYP) 3A or fluxed via P-glycoprotein (Pgp), although it is also reported that RTV is a potent inducer for them. Ritonavir 11-14 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 84-108 15635177-1 2005 Ritonavir (RTV) is well known as an inhibitor of many drugs that are metabolized by cytochrome P450 (CYP) 3A or fluxed via P-glycoprotein (Pgp), although it is also reported that RTV is a potent inducer for them. Ritonavir 11-14 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 123-137 15635177-1 2005 Ritonavir (RTV) is well known as an inhibitor of many drugs that are metabolized by cytochrome P450 (CYP) 3A or fluxed via P-glycoprotein (Pgp), although it is also reported that RTV is a potent inducer for them. Ritonavir 11-14 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 139-142 15635177-1 2005 Ritonavir (RTV) is well known as an inhibitor of many drugs that are metabolized by cytochrome P450 (CYP) 3A or fluxed via P-glycoprotein (Pgp), although it is also reported that RTV is a potent inducer for them. Ritonavir 179-182 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 84-108 15635177-1 2005 Ritonavir (RTV) is well known as an inhibitor of many drugs that are metabolized by cytochrome P450 (CYP) 3A or fluxed via P-glycoprotein (Pgp), although it is also reported that RTV is a potent inducer for them. Ritonavir 179-182 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 123-137 15635177-1 2005 Ritonavir (RTV) is well known as an inhibitor of many drugs that are metabolized by cytochrome P450 (CYP) 3A or fluxed via P-glycoprotein (Pgp), although it is also reported that RTV is a potent inducer for them. Ritonavir 179-182 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 139-142 15635177-2 2005 In this study, to elucidate these contradictory phenomena, functional changes of CYP3A or Pgp during chronic administration of RTV were examined in rats. Ritonavir 127-130 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 81-86 15635177-2 2005 In this study, to elucidate these contradictory phenomena, functional changes of CYP3A or Pgp during chronic administration of RTV were examined in rats. Ritonavir 127-130 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 90-93 15635177-12 2005 It has been confirmed that RTV is not only a potent inhibitor but also a potent inducer of CYP3A, and that RTV is a potent inducer of intestinal Pgp. Ritonavir 27-30 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 91-96 15635177-12 2005 It has been confirmed that RTV is not only a potent inhibitor but also a potent inducer of CYP3A, and that RTV is a potent inducer of intestinal Pgp. Ritonavir 107-110 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 145-148 15504750-0 2004 The HIV protease inhibitor ritonavir synergizes with butyrate for induction of apoptotic cell death and mediates expression of heme oxygenase-1 in DLD-1 colon carcinoma cells. Ritonavir 27-36 heme oxygenase 1 Homo sapiens 127-143 15849716-7 2005 UGT1A1 was most responsive to the pregnane-X-receptor-agonists rifampicin, ritonavir, and clotrimazole at the mRNA level and, to a lesser extent, the constitutive androstane receptor-activators, phenobarbital and phenytoin. Ritonavir 75-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 15849716-7 2005 UGT1A1 was most responsive to the pregnane-X-receptor-agonists rifampicin, ritonavir, and clotrimazole at the mRNA level and, to a lesser extent, the constitutive androstane receptor-activators, phenobarbital and phenytoin. Ritonavir 75-84 nuclear receptor subfamily 1 group I member 2 Homo sapiens 34-53 15504750-3 2004 Moreover, ritonavir can suppress activation of the transcription factor nuclear factor-kappaB and is an inhibitor of interleukin-1beta and tumor necrosis factor-alpha production in peripheral blood mononuclear cells. Ritonavir 10-19 interleukin 1 beta Homo sapiens 117-166 15504750-7 2004 Notably, ritonavir potently synergized with the short-chain fatty acid butyrate for induction of caspase-3-dependent apoptosis in DLD-1 cells. Ritonavir 9-18 caspase 3 Homo sapiens 97-106 15504750-9 2004 Ritonavir-induced HO-1 protein was suppressed by SB203580 or SB202190 and preceded by immediate upregulation of cellular c-Fos and c-Jun protein levels. Ritonavir 0-9 heme oxygenase 1 Homo sapiens 18-22 15504750-9 2004 Ritonavir-induced HO-1 protein was suppressed by SB203580 or SB202190 and preceded by immediate upregulation of cellular c-Fos and c-Jun protein levels. Ritonavir 0-9 Fos proto-oncogene, AP-1 transcription factor subunit Homo sapiens 121-126 15504750-9 2004 Ritonavir-induced HO-1 protein was suppressed by SB203580 or SB202190 and preceded by immediate upregulation of cellular c-Fos and c-Jun protein levels. Ritonavir 0-9 Jun proto-oncogene, AP-1 transcription factor subunit Homo sapiens 131-136 15504750-11 2004 The present data suggest that ritonavir has the potential to curb colon carcinogenesis by reducing cell growth via mechanisms that include apoptosis and by simultaneously modulating colonic inflammation via induction of anti-inflammatory HO-1. Ritonavir 30-39 heme oxygenase 1 Homo sapiens 238-242 15590971-6 2004 The impaired insulin secretion by ritonavir, nelfinavir and saquinavir was associated with decreased insulin-stimulated IRS-2 phosphorylation, and, for nelfinavir and saquinavir, with decreased insulin-stimulated IRS-1 and Thr308-Akt phosphorylation. Ritonavir 34-43 insulin Homo sapiens 13-20 15590971-8 2004 In conclusion, certain HIV-1 protease inhibitors, such as ritonavir, nelfinavir and saquinavir, not only induce peripheral insulin resistance, but also impair glucose-stimulated insulin secretion from beta cells. Ritonavir 58-67 insulin Homo sapiens 178-185 15590971-6 2004 The impaired insulin secretion by ritonavir, nelfinavir and saquinavir was associated with decreased insulin-stimulated IRS-2 phosphorylation, and, for nelfinavir and saquinavir, with decreased insulin-stimulated IRS-1 and Thr308-Akt phosphorylation. Ritonavir 34-43 insulin receptor substrate 2 Homo sapiens 120-125 15590971-6 2004 The impaired insulin secretion by ritonavir, nelfinavir and saquinavir was associated with decreased insulin-stimulated IRS-2 phosphorylation, and, for nelfinavir and saquinavir, with decreased insulin-stimulated IRS-1 and Thr308-Akt phosphorylation. Ritonavir 34-43 insulin Homo sapiens 101-108 15590971-8 2004 In conclusion, certain HIV-1 protease inhibitors, such as ritonavir, nelfinavir and saquinavir, not only induce peripheral insulin resistance, but also impair glucose-stimulated insulin secretion from beta cells. Ritonavir 58-67 insulin Homo sapiens 123-130 15492266-0 2004 HIV-1 protease inhibitor, ritonavir: a potent inhibitor of CYP3A4, enhanced the anticancer effects of docetaxel in androgen-independent prostate cancer cells in vitro and in vivo. Ritonavir 26-35 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 59-65 15483467-8 2004 This was compensated for by an increased ritonavir dose of 200 mg bid, which resulted in a statistically nonsignificant increase in saquinavir exposure compared with baseline. Ritonavir 41-50 BH3 interacting domain death agonist Homo sapiens 66-69 15483467-13 2004 However, the addition of a further 100 mg ritonavir bid restored the small and insignificant decrease. Ritonavir 42-51 BH3 interacting domain death agonist Homo sapiens 52-55 15577646-0 2004 The effects of HIV protease inhibitors atazanavir and lopinavir/ritonavir on insulin sensitivity in HIV-seronegative healthy adults. Ritonavir 64-73 insulin Homo sapiens 77-84 15577646-3 2004 METHODS: Randomized, double-blind, crossover study of the effect of 5 days of administering ATV, lopinavir/ritonavir (LPV/r) or placebo on insulin-stimulated glucose disposal in 30 healthy HIV-negative subjects. Ritonavir 107-116 insulin Homo sapiens 139-146 15492266-3 2004 Recent studies showed that ritonavir inhibited cytochrome P450 3A4 enzyme (CYP3A4) in liver microsomes. Ritonavir 27-36 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 47-73 15492266-3 2004 Recent studies showed that ritonavir inhibited cytochrome P450 3A4 enzyme (CYP3A4) in liver microsomes. Ritonavir 27-36 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 75-81 15492266-6 2004 Ritonavir enhanced the antiproliferative and proapoptotic effects of docetaxel in the hormonally independent DU145 prostate cancer cells in vitro as measured by the clonogenic soft agar assay and detection of the activated form of caspase-3 and cleavage of poly(ADP-ribose) polymerase using Western blot analysis. Ritonavir 0-9 caspase 3 Homo sapiens 231-240 15492266-6 2004 Ritonavir enhanced the antiproliferative and proapoptotic effects of docetaxel in the hormonally independent DU145 prostate cancer cells in vitro as measured by the clonogenic soft agar assay and detection of the activated form of caspase-3 and cleavage of poly(ADP-ribose) polymerase using Western blot analysis. Ritonavir 0-9 poly(ADP-ribose) polymerase 1 Homo sapiens 257-284 15492266-8 2004 An ELISA-based assay also showed that ritonavir inhibited DNA binding activity of nuclear factor kappaB (NFkappaB) in DU145 cells, which is a contributor to drug resistance in cancer cells. Ritonavir 38-47 nuclear factor kappa B subunit 1 Homo sapiens 82-103 15492266-8 2004 An ELISA-based assay also showed that ritonavir inhibited DNA binding activity of nuclear factor kappaB (NFkappaB) in DU145 cells, which is a contributor to drug resistance in cancer cells. Ritonavir 38-47 nuclear factor kappa B subunit 1 Homo sapiens 105-113 15492266-11 2004 Ritonavir also inhibited NFkappaB DNA binding activity in DU145 xenografts. Ritonavir 0-9 nuclear factor kappa B subunit 1 Homo sapiens 25-33 15553233-5 2004 RESULTS: Increase of calculated permeabilities of P-gp-specific substrate talinolol was found under co-administration of both PIs, ritonavir and H17, with highest absorption rates in the ileal and colon segment. Ritonavir 131-140 ATP binding cassette subfamily B member 1 Homo sapiens 50-54 15221291-4 2004 Following the introduction of a more sensitive assay, using the human TSH receptor as an antigen, it has been expected that the incidence of radiation-induced GD after RIT for functional thyroid autonomy will be reduced. Ritonavir 168-171 thyroid stimulating hormone receptor Homo sapiens 70-82 15553233-6 2004 H17 proved to be a better P-gp inhibitor than ritonavir by resulting IC50 values and also in the cellular uptake of rhodamine. Ritonavir 46-55 H1.7 linker histone Homo sapiens 0-3 15205383-10 2004 In summary, coadministration of low-dose ritonavir (a potent CYP3A4 inhibitor) drastically decreased the levels of sequential oxygenated metabolites and markedly increased the levels of the parent drug and primary oxygenated metabolites overall in plasma, urine, and feces. Ritonavir 41-50 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 61-67 15220698-12 2004 These data suggest that amprenavir/saquinavir/ritonavir may be a viable salvage regimen in heavily PI-experienced individuals. Ritonavir 46-55 serpin family A member 13, pseudogene Homo sapiens 99-101 15266218-0 2004 In vitro interaction of the HIV protease inhibitor ritonavir with herbal constituents: changes in P-gp and CYP3A4 activity. Ritonavir 51-60 phosphoglycolate phosphatase Homo sapiens 98-102 15266218-0 2004 In vitro interaction of the HIV protease inhibitor ritonavir with herbal constituents: changes in P-gp and CYP3A4 activity. Ritonavir 51-60 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 107-113 15266218-6 2004 Quercetin, hypericin, and kaempferol exhibited a remarkable inhibition of P-gp-mediated efflux of ritonavir by increasing its cellular uptake in these models. Ritonavir 98-107 phosphoglycolate phosphatase Homo sapiens 74-78 15266218-7 2004 These values were also comparable with the inhibitory effect of quinidine in Caco-2 cells, a well-known inhibitor of P-gp, on ritonavir efflux from Caco-2 cells. Ritonavir 126-135 phosphoglycolate phosphatase Homo sapiens 117-121 15266218-8 2004 Allicin exhibited a concentration-dependent inhibition of ritonavir efflux when tested on MDR1-MDCK cells. Ritonavir 58-67 ATP binding cassette subfamily B member 1 Homo sapiens 90-94 15229466-1 2004 BACKGROUND: Ritonavir is a potent in vitro inhibitor of several cytochrome P450 isozymes and ABC transporters including the efflux pump P-glycoprotein (P-gp). Ritonavir 12-21 ATP binding cassette subfamily B member 1 Homo sapiens 136-150 15229466-1 2004 BACKGROUND: Ritonavir is a potent in vitro inhibitor of several cytochrome P450 isozymes and ABC transporters including the efflux pump P-glycoprotein (P-gp). Ritonavir 12-21 ATP binding cassette subfamily B member 1 Homo sapiens 152-156 15229466-9 2004 CONCLUSION: This inhibition of renal digoxin clearance is likely caused by ritonavir inhibition of P-gp. Ritonavir 75-84 ATP binding cassette subfamily B member 1 Homo sapiens 99-103 15229466-11 2004 These findings may, therefore, indicate that the pharmacokinetics of P-gp substrates sharing the renal tubular elimination pathway will be affected when combined with therapeutic doses of ritonavir in antiretroviral treatment regimens. Ritonavir 188-197 ATP binding cassette subfamily B member 1 Homo sapiens 69-73 15247556-0 2004 Coadministration of lopinavir/ritonavir and phenytoin results in two-way drug interaction through cytochrome P-450 induction. Ritonavir 30-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-114 15247556-1 2004 Lopinavir/ritonavir (LPV/RTV) is a CYP3A4 inhibitor and substrate; it also may induce cytochrome P-450 (CYP) isozymes. Ritonavir 10-19 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 35-41 15247556-1 2004 Lopinavir/ritonavir (LPV/RTV) is a CYP3A4 inhibitor and substrate; it also may induce cytochrome P-450 (CYP) isozymes. Ritonavir 10-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 15247556-1 2004 Lopinavir/ritonavir (LPV/RTV) is a CYP3A4 inhibitor and substrate; it also may induce cytochrome P-450 (CYP) isozymes. Ritonavir 10-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-38 15059841-7 2004 Additional ABC transporter substrate inhibitors like quinidine, diltiazem, and ritonavir also enhanced transduction 2- to 3-fold, although ABC transporter inhibitors that are not substrates did not. Ritonavir 79-88 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 11-26 15254587-5 2004 In keeping with its antiresorptive properties, ritonavir impairs receptor activator of nuclear factor kappaB ligand-induced (RANKL-induced) activation of NF-kappaB and Akt signaling pathways, both critical to osteoclast formation and function. Ritonavir 47-56 TNF superfamily member 11 Homo sapiens 125-130 15199084-13 2004 The pharmacokinetics of enfuvirtide are affected to a small extent when coadministered with ritonavir at a dose of 200 mg bid but not when coadministered with a saquinavir-ritonavir combination (1000/100 mg bid). Ritonavir 92-101 BH3 interacting domain death agonist Homo sapiens 122-125 15254587-5 2004 In keeping with its antiresorptive properties, ritonavir impairs receptor activator of nuclear factor kappaB ligand-induced (RANKL-induced) activation of NF-kappaB and Akt signaling pathways, both critical to osteoclast formation and function. Ritonavir 47-56 nuclear factor kappa B subunit 1 Homo sapiens 154-163 15254587-5 2004 In keeping with its antiresorptive properties, ritonavir impairs receptor activator of nuclear factor kappaB ligand-induced (RANKL-induced) activation of NF-kappaB and Akt signaling pathways, both critical to osteoclast formation and function. Ritonavir 47-56 AKT serine/threonine kinase 1 Homo sapiens 168-171 15254587-6 2004 In particular, ritonavir is found to inhibit RANKL-induced Akt signaling by disrupting the recruitment of TNF receptor-associated factor 6/c-Src complex to lipid rafts. Ritonavir 15-24 TNF superfamily member 11 Homo sapiens 45-50 15254587-6 2004 In particular, ritonavir is found to inhibit RANKL-induced Akt signaling by disrupting the recruitment of TNF receptor-associated factor 6/c-Src complex to lipid rafts. Ritonavir 15-24 AKT serine/threonine kinase 1 Homo sapiens 59-62 15254587-6 2004 In particular, ritonavir is found to inhibit RANKL-induced Akt signaling by disrupting the recruitment of TNF receptor-associated factor 6/c-Src complex to lipid rafts. Ritonavir 15-24 LOC222344 Homo sapiens 106-138 15254587-6 2004 In particular, ritonavir is found to inhibit RANKL-induced Akt signaling by disrupting the recruitment of TNF receptor-associated factor 6/c-Src complex to lipid rafts. Ritonavir 15-24 SRC proto-oncogene, non-receptor tyrosine kinase Homo sapiens 139-144 15167636-0 2004 Ritonavir decreases the nonrenal clearance of digoxin in healthy volunteers with known MDR1 genotypes. Ritonavir 0-9 ATP binding cassette subfamily B member 1 Homo sapiens 87-91 15007102-5 2004 We found that ritonavir, saquinavir, and nelfinavir were effective inhibitors of wild-type BCRP (482R) with IC50 values of 19.5 +/- 0.8 microM, 19.5 +/- 7.6 microM, and 12.5 +/- 4.1 microM, respectively. Ritonavir 14-23 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 91-95 15222662-8 2004 Patients in the lopinavir-ritonavir group were more likely to have received highly active antiretroviral therapy and azithromycin than patients receiving nelfinavir, and they had lower baseline CD4+ cell counts (p < or = 0.01 for each comparison). Ritonavir 26-35 CD4 molecule Homo sapiens 194-197 15167636-1 2004 Our objective was to examine the influence of ritonavir on P-glycoprotein (P-gp) activity in humans by characterizing the effect of ritonavir on the pharmacokinetics of the P-gp substrate digoxin in individuals with known MDR1 genotypes. Ritonavir 46-55 ATP binding cassette subfamily B member 1 Homo sapiens 59-73 15167636-1 2004 Our objective was to examine the influence of ritonavir on P-glycoprotein (P-gp) activity in humans by characterizing the effect of ritonavir on the pharmacokinetics of the P-gp substrate digoxin in individuals with known MDR1 genotypes. Ritonavir 46-55 ATP binding cassette subfamily B member 1 Homo sapiens 75-79 15167636-1 2004 Our objective was to examine the influence of ritonavir on P-glycoprotein (P-gp) activity in humans by characterizing the effect of ritonavir on the pharmacokinetics of the P-gp substrate digoxin in individuals with known MDR1 genotypes. Ritonavir 132-141 ATP binding cassette subfamily B member 1 Homo sapiens 173-177 15167636-12 2004 The most likely mechanism for this interaction is ritonavir-associated inhibition of P-gp. Ritonavir 50-59 ATP binding cassette subfamily B member 1 Homo sapiens 85-89 15167636-13 2004 Thus, ritonavir can alter the pharmacokinetics of coadministered medications that are P-gp substrates. Ritonavir 6-15 ATP binding cassette subfamily B member 1 Homo sapiens 86-90 15078991-2 2004 In this study, we found that protease inhibitors, including ritonavir, saquinavir, and nelfinavir, but not indinavir, induced growth arrest and apoptosis of U266, RPMI8226, and ARH77 human multiple myeloma (MM) cells in association with down-regulation of antiapoptotic protein Mcl-1. Ritonavir 60-69 low density lipoprotein receptor adaptor protein 1 Homo sapiens 177-180 15063133-7 2004 After radioiodine therapy (RIT), TCR-Mf increases within about half a year to a maximum. Ritonavir 27-30 T cell receptor beta variable 20/OR9-2 (non-functional) Homo sapiens 33-36 15078991-2 2004 In this study, we found that protease inhibitors, including ritonavir, saquinavir, and nelfinavir, but not indinavir, induced growth arrest and apoptosis of U266, RPMI8226, and ARH77 human multiple myeloma (MM) cells in association with down-regulation of antiapoptotic protein Mcl-1. Ritonavir 60-69 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 278-283 15078991-7 2004 Furthermore, ritonavir inhibited production of vascular endothelial growth factor one of the targets of STAT 3, in U266 and RPMI8226 cells as measured by ELISA. Ritonavir 13-22 vascular endothelial growth factor A Homo sapiens 47-81 15078991-7 2004 Furthermore, ritonavir inhibited production of vascular endothelial growth factor one of the targets of STAT 3, in U266 and RPMI8226 cells as measured by ELISA. Ritonavir 13-22 signal transducer and activator of transcription 3 Homo sapiens 104-110 15040543-16 2004 The addition of ritonavir further reduced P-gp function. Ritonavir 16-25 ATP binding cassette subfamily B member 1 Homo sapiens 42-46 15203316-4 2004 Production by PBMCs of IL-12 and IFN-gamma was significantly higher and production of IL-4 was significantly lower after stimulation with Der f allergen in RIT-treated patients than in control patients. Ritonavir 156-159 interferon gamma Homo sapiens 33-42 15203316-5 2004 Significant increases in the expression of IL-12R beta2 chain before and after stimulation of CD4(+) T cells with IL-12 or IFN-gamma were observed in RIT-treated patients compared with that in control patients. Ritonavir 150-153 CD4 molecule Homo sapiens 94-97 15203316-5 2004 Significant increases in the expression of IL-12R beta2 chain before and after stimulation of CD4(+) T cells with IL-12 or IFN-gamma were observed in RIT-treated patients compared with that in control patients. Ritonavir 150-153 interferon gamma Homo sapiens 123-132 14985144-0 2004 Ritonavir and dexamethasone induce expression of CYP3A and P-glycoprotein in rats. Ritonavir 0-9 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 49-54 14592951-0 2004 Adiponectin ameliorates dyslipidemia induced by the human immunodeficiency virus protease inhibitor ritonavir in mice. Ritonavir 100-109 adiponectin, C1Q and collagen domain containing Mus musculus 0-11 14592951-4 2004 The steady-state mRNA levels of the adiponectin gene and secretion of this protein from 3T3-L1 adipocytes are significantly decreased after treatment with several PIs (indinavir, nelfinavir, and ritonavir), with ritonavir having the greatest effect. Ritonavir 195-204 adiponectin, C1Q and collagen domain containing Mus musculus 36-47 14592951-4 2004 The steady-state mRNA levels of the adiponectin gene and secretion of this protein from 3T3-L1 adipocytes are significantly decreased after treatment with several PIs (indinavir, nelfinavir, and ritonavir), with ritonavir having the greatest effect. Ritonavir 212-221 adiponectin, C1Q and collagen domain containing Mus musculus 36-47 14592951-5 2004 Intragastric administration of ritonavir into mice decreases plasma concentrations of adiponectin and concurrently increases the plasma levels of triglyceride, free fatty acids, and cholesterol. Ritonavir 31-40 adiponectin, C1Q and collagen domain containing Mus musculus 86-97 14592951-6 2004 Adiponectin replacement therapy markedly ameliorates ritonavir-induced elevations of triglyceride and free fatty acids. Ritonavir 53-62 adiponectin, C1Q and collagen domain containing Mus musculus 0-11 14592951-7 2004 These beneficial effects of adiponectin are partly due to its ability to decrease ritonavir-induced synthesis of fatty acids and triglyceride, and to increase fatty acid combustion in the liver tissue. Ritonavir 82-91 adiponectin, C1Q and collagen domain containing Mus musculus 28-39 14985144-0 2004 Ritonavir and dexamethasone induce expression of CYP3A and P-glycoprotein in rats. Ritonavir 0-9 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 59-73 14985144-2 2004 The consequences of extended exposure to the human immunodeficiency viral protease inhibitor ritonavir (RIT) on the expression and function of CYP3A isoforms in the liver and in enteric mucosal cells, and on the expression of the efflux transport protein P-glycoprotein (P-gp) in enteric mucosa and in brain microvessel endothelial cells, were evaluated in rat. Ritonavir 93-102 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 143-148 14985144-2 2004 The consequences of extended exposure to the human immunodeficiency viral protease inhibitor ritonavir (RIT) on the expression and function of CYP3A isoforms in the liver and in enteric mucosal cells, and on the expression of the efflux transport protein P-glycoprotein (P-gp) in enteric mucosa and in brain microvessel endothelial cells, were evaluated in rat. Ritonavir 93-102 ATP binding cassette subfamily B member 1 Homo sapiens 255-269 14985144-2 2004 The consequences of extended exposure to the human immunodeficiency viral protease inhibitor ritonavir (RIT) on the expression and function of CYP3A isoforms in the liver and in enteric mucosal cells, and on the expression of the efflux transport protein P-glycoprotein (P-gp) in enteric mucosa and in brain microvessel endothelial cells, were evaluated in rat. Ritonavir 93-102 ATP binding cassette subfamily B member 1 Homo sapiens 271-275 14985144-2 2004 The consequences of extended exposure to the human immunodeficiency viral protease inhibitor ritonavir (RIT) on the expression and function of CYP3A isoforms in the liver and in enteric mucosal cells, and on the expression of the efflux transport protein P-glycoprotein (P-gp) in enteric mucosa and in brain microvessel endothelial cells, were evaluated in rat. Ritonavir 104-107 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 143-148 14985144-2 2004 The consequences of extended exposure to the human immunodeficiency viral protease inhibitor ritonavir (RIT) on the expression and function of CYP3A isoforms in the liver and in enteric mucosal cells, and on the expression of the efflux transport protein P-glycoprotein (P-gp) in enteric mucosa and in brain microvessel endothelial cells, were evaluated in rat. Ritonavir 104-107 ATP binding cassette subfamily B member 1 Homo sapiens 255-269 14985144-2 2004 The consequences of extended exposure to the human immunodeficiency viral protease inhibitor ritonavir (RIT) on the expression and function of CYP3A isoforms in the liver and in enteric mucosal cells, and on the expression of the efflux transport protein P-glycoprotein (P-gp) in enteric mucosa and in brain microvessel endothelial cells, were evaluated in rat. Ritonavir 104-107 ATP binding cassette subfamily B member 1 Homo sapiens 271-275 14985144-7 2004 Compared with vehicle control, CYP3A activity in liver microsomes (intrinsic clearance for triazolam hydroxylation in vitro) was increased by a factor of 2-4 by RIT, and by 10-14-fold by DEX. Ritonavir 161-164 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 31-36 14985144-10 2004 Both RIT and DEX also increased function and expression of enteric CYP3A, although to a more modest extent (about 1.7-fold for RIT, about 3.3-fold for DEX). Ritonavir 5-8 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 67-72 14985144-10 2004 Both RIT and DEX also increased function and expression of enteric CYP3A, although to a more modest extent (about 1.7-fold for RIT, about 3.3-fold for DEX). Ritonavir 127-130 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 67-72 14985144-12 2004 Enteric P-gp expression was equally induced (by 2.8-fold) by both RIT and DEX. Ritonavir 66-69 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 8-12 14985144-15 2004 Thus, increased expression of CYP3A isoforms and of P-gp occurs with 3 days of exposure to RIT in rats. Ritonavir 91-94 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 30-35 14985144-15 2004 Thus, increased expression of CYP3A isoforms and of P-gp occurs with 3 days of exposure to RIT in rats. Ritonavir 91-94 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 52-56 14525546-11 2003 Conversely, ritonavir and indinavir induced transient P-gp expression in a small percentage of the CEMrev cells. Ritonavir 12-21 ATP binding cassette subfamily B member 1 Homo sapiens 54-58 15491071-8 2004 The mortality rate was significantly affected by ritonavir (2/10 maternal deaths in E2 and 4/10 in E3). Ritonavir 49-58 dihydrolipoamide S-succinyltransferase Rattus norvegicus 84-92 15002082-1 2004 PURPOSE: To compare the long-term clinical efficacy and toxicity of initial strategies of nelfinavir (NFV) or ritonavir (RTV) in patients with CD4+ cells below 200/mm3. Ritonavir 110-119 CD4 molecule Homo sapiens 143-146 15002082-1 2004 PURPOSE: To compare the long-term clinical efficacy and toxicity of initial strategies of nelfinavir (NFV) or ritonavir (RTV) in patients with CD4+ cells below 200/mm3. Ritonavir 121-124 CD4 molecule Homo sapiens 143-146 15764168-10 2004 We found that antiretroviral combination therapy including lopinavir/ritonavir substantially decreases the viral load, both in CSF and plasma, as well as the intrathecal immunoactivation, measured by beta2-microglobulin and neopterin. Ritonavir 69-78 beta-2-microglobulin Homo sapiens 200-219 14709251-6 2003 Ritonavir was coadministered with the peroxisome proliferator-activated receptor alpha (PPARalpha) agonist gemfibrozil and the PPARgamma agonist rosiglitazone for 8 weeks. Ritonavir 0-9 peroxisome proliferator activated receptor alpha Mus musculus 38-86 14709251-6 2003 Ritonavir was coadministered with the peroxisome proliferator-activated receptor alpha (PPARalpha) agonist gemfibrozil and the PPARgamma agonist rosiglitazone for 8 weeks. Ritonavir 0-9 peroxisome proliferator activated receptor alpha Mus musculus 88-97 14709251-6 2003 Ritonavir was coadministered with the peroxisome proliferator-activated receptor alpha (PPARalpha) agonist gemfibrozil and the PPARgamma agonist rosiglitazone for 8 weeks. Ritonavir 0-9 peroxisome proliferator activated receptor gamma Mus musculus 127-136 14709251-9 2003 Ritonavir reduced basal expression of two PPARalpha target genes in liver, as well as the PPARgamma target gene phosphoenolpyruvate carboxykinase (PEPCK) in adipose tissues. Ritonavir 0-9 peroxisome proliferator activated receptor alpha Mus musculus 42-51 14709251-9 2003 Ritonavir reduced basal expression of two PPARalpha target genes in liver, as well as the PPARgamma target gene phosphoenolpyruvate carboxykinase (PEPCK) in adipose tissues. Ritonavir 0-9 phosphoenolpyruvate carboxykinase 1, cytosolic Mus musculus 112-145 14709251-9 2003 Ritonavir reduced basal expression of two PPARalpha target genes in liver, as well as the PPARgamma target gene phosphoenolpyruvate carboxykinase (PEPCK) in adipose tissues. Ritonavir 0-9 phosphoenolpyruvate carboxykinase 1, cytosolic Mus musculus 147-152 14709251-10 2003 Ritonavir partially inhibited induction of PPAR target genes by gemfibrozil and rosiglitazone. Ritonavir 0-9 peroxisome proliferator activated receptor alpha Mus musculus 43-47 14709251-12 2003 Similarly, rosiglitazone induced expression of uncoupling protein-1, uncoupling protein-2, and PEPCK in adipose tissues, and this effect was partially inhibited by ritonavir. Ritonavir 164-173 phosphoenolpyruvate carboxykinase 1, cytosolic Mus musculus 95-100 14709251-13 2003 Thus, the effects of ritonavir on serum triglycerides and body composition may be due, at least in part, to an inhibition of PPAR function. Ritonavir 21-30 peroxisome proliferator activated receptor alpha Mus musculus 125-129 15090838-0 2004 Resistance to amprenavir before and after treatment with lopinavir/ritonavir in highly protease inhibitor-experienced HIV patients. Ritonavir 67-76 serpin family A member 13, pseudogene Homo sapiens 87-105 14731164-1 2004 In a controlled, prospective study, the efficacy of ritonavir 200 mg twice daily (bid) in inhibiting the decrease of amprenavir plasma concentrations caused by co-administration of lopinavir was assessed. Ritonavir 52-61 BH3 interacting domain death agonist Homo sapiens 82-85 14731164-7 2004 In conclusion, gastrointestinal tolerance of a regimen containing an increased dose of ritonavir 200 mg bid was low, while the regimen did not prevent a decrease of amprenavir and possibly lopinavir plasma concentrations. Ritonavir 87-96 BH3 interacting domain death agonist Homo sapiens 104-107 14690877-7 2003 However, it cannot be excluded that co-administration of Pgp inhibitors such as ritonavir or paroxetine could increase MDMA, MDE and PMA bioavailability and also enhance brain entry leading to severe side effects. Ritonavir 80-89 ATP binding cassette subfamily B member 1 Homo sapiens 57-60 12837856-5 2003 In this present study we investigated the effect of several HIV protease inhibitors (ABT-378, Amprenavir, Indinavir, Nelfinavir, Ritonavir, and Saquinavir) on mRNA, protein, and functional levels of LDLR family members. Ritonavir 129-138 low density lipoprotein receptor Homo sapiens 199-203 14525542-1 2003 OBJECTIVES: To examine the relationship between levels of the T-cell regulatory cytokine interleukin-7 (IL-7) and CD4 cell counts during immune reconstitution and to assess its prognostic value in advanced HIV-1-infected patients receiving lopinavir/ritonavir-based therapy. Ritonavir 250-259 interleukin 7 Homo sapiens 104-108 14525546-7 2003 A similar strategy was adopted to determine whether other PIs, such as ritonavir and indinavir, were able to induce P-gp expression in CEMrev cells. Ritonavir 71-80 ATP binding cassette subfamily B member 1 Homo sapiens 116-120 12714803-4 2003 In fact, ritonavir, saquinavir and indinavir act differently to the P-glycoprotein blocker in CEM-VBL10 cells. Ritonavir 9-18 ATP binding cassette subfamily B member 1 Homo sapiens 68-82 14562864-4 2003 Since CYP3A is the major cytochrome P450 isoform for the phase I metabolism of TPV, its exposure is markedly enhanced in the presence of ritonavir (RTV). Ritonavir 137-146 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 6-11 14562864-4 2003 Since CYP3A is the major cytochrome P450 isoform for the phase I metabolism of TPV, its exposure is markedly enhanced in the presence of ritonavir (RTV). Ritonavir 148-151 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 6-11 12902797-9 2003 However, nelfinavir, ritonavir, and lopinavir caused marked toxicity, indicating that at higher concentrations, the increase in P-gp may be at least partially related to a stress response. Ritonavir 21-30 ATP binding cassette subfamily B member 1 Homo sapiens 128-132 12902799-6 2003 In contrast, daily administration of leptin significantly reversed the elevated plasma cholesterol level induced by ritonavir. Ritonavir 116-125 leptin Mus musculus 37-43 12902799-7 2003 Leptin replacement therapy also significantly reduced the ritonavir-induced interscapular fat mass and improved liver steatosis. Ritonavir 58-67 leptin Mus musculus 0-6 12967347-6 2003 Also, ritonavir (10 microM), which leads to a 2-fold increase in 2-DG transport, demonstrated increased GLUT (i.e. 1, 3 or 4) presence in the plasma membrane fraction, in the presence or absence of insulin. Ritonavir 6-15 solute carrier family 2 member 1 Homo sapiens 104-108 12948013-5 2003 RESULTS: In situ experimental results revealed that the extent to which the intestinal absorption is affected by P-gp was in the following order: quinidine > ritonavir > loperamide, verapamil, daunomycin > digoxin, cyclosporin A > dexamethasone, and vinblastine. Ritonavir 161-170 ATP binding cassette subfamily B member 1 Homo sapiens 113-117 12948015-9 2003 The rank order in P-gp inhibitory potency for terfenadine, verapamil, ritonavir. Ritonavir 70-79 ATP binding cassette subfamily B member 1 Homo sapiens 18-22 12948015-11 2003 Ritonavir and St. John"s wort extract showed a concentration-dependent P-gp induction, with good correlation between western blot analysis and RH123 accumulation. Ritonavir 0-9 ATP binding cassette subfamily B member 1 Homo sapiens 71-75 12824804-0 2003 Digoxin toxicity and ritonavir: a drug interaction mediated through p-glycoprotein? Ritonavir 21-30 ATP binding cassette subfamily B member 1 Homo sapiens 68-82 12714803-6 2003 In CEM-VBL100 cells, expressing a very high number of P-glycoprotein molecules, only ritonavir acts as an efficient drug efflux inhibitor and MDR-reversing agent. Ritonavir 85-94 ATP binding cassette subfamily B member 1 Homo sapiens 54-68 12809966-0 2003 An evaluation of the potential for pharmacokinetic interaction between escitalopram and the cytochrome P450 3A4 inhibitor ritonavir. Ritonavir 122-131 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 92-111 12723462-2 2003 The viral protease inhibitor, ritonavir, may cause drug interactions by inhibiting the activity of cytochrome P450-3A (CYP3A) isoforms. Ritonavir 30-39 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 99-117 12723462-2 2003 The viral protease inhibitor, ritonavir, may cause drug interactions by inhibiting the activity of cytochrome P450-3A (CYP3A) isoforms. Ritonavir 30-39 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 119-124 12723462-8 2003 The findings are consistent with impairment of CYP3A-mediated trazodone metabolism by ritonavir. Ritonavir 86-95 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 47-52 12584153-8 2003 This assumption was confirmed by inhibition with ketoconazole and ritonavir (two potent inhibitors of CYP3A) whereas sulfaphenazole (2C9 inhibitor) and quinidine (2D6 inhibitor) were inefficient. Ritonavir 66-75 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 102-107 12605563-4 2003 Coadministration of ritonavir can enhance exposures of a primary PI by inhibiting CYP3A4 metabolism, P-glycoprotein activity and multi-drug resistance protein-1-mediated efflux. Ritonavir 20-29 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 82-88 12719666-10 2003 However, the treatment with LPV/RTV was not interrupted for these patients, because in the follow-up they showed an increase in CD4+ values. Ritonavir 32-35 CD4 molecule Homo sapiens 128-131 12724045-8 2003 Although lopinavir is less potent than ritonavir as an inhibitor of CYP3A, lopinavir is nonetheless likely to contribute to net CYP3A inhibition in-vivo during treatment with the lopinavir-ritonavir combination. Ritonavir 39-48 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 68-73 12578750-6 2003 Saquinavir and ritonavir, but not nelfinavir also inhibited TNF-alpha induced cell death. Ritonavir 15-24 tumor necrosis factor Homo sapiens 60-69 12604693-0 2003 Pharmacokinetics and interactions of a novel antagonist of chemokine receptor 5 (CCR5) with ritonavir in rats and monkeys: role of CYP3A and P-glycoprotein. Ritonavir 92-101 C-C motif chemokine receptor 5 Rattus norvegicus 59-79 12604693-0 2003 Pharmacokinetics and interactions of a novel antagonist of chemokine receptor 5 (CCR5) with ritonavir in rats and monkeys: role of CYP3A and P-glycoprotein. Ritonavir 92-101 C-C motif chemokine receptor 5 Rattus norvegicus 81-85 12604693-3 2003 Both the in vitro MDR1-mediated transport and oxidative metabolism of MRK-1 were inhibited by ritonavir. Ritonavir 94-103 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 18-22 12604693-6 2003 Further pharmacokinetic studies in rats indicated that P-glycoprotein (P-gp) inhibition by ritonavir increased the intestinal absorption of 2 mg/kg MRK-1 maximally by approximately 30 to 40%, and a major component of the interaction likely resulted from its reduced systemic clearance via the inhibition of CYP3A isozymes. Ritonavir 91-100 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 55-69 12604693-6 2003 Further pharmacokinetic studies in rats indicated that P-glycoprotein (P-gp) inhibition by ritonavir increased the intestinal absorption of 2 mg/kg MRK-1 maximally by approximately 30 to 40%, and a major component of the interaction likely resulted from its reduced systemic clearance via the inhibition of CYP3A isozymes. Ritonavir 91-100 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 71-75 12604693-6 2003 Further pharmacokinetic studies in rats indicated that P-glycoprotein (P-gp) inhibition by ritonavir increased the intestinal absorption of 2 mg/kg MRK-1 maximally by approximately 30 to 40%, and a major component of the interaction likely resulted from its reduced systemic clearance via the inhibition of CYP3A isozymes. Ritonavir 91-100 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 307-312 12790697-1 2003 UNLABELLED: Protease inhibitor boosting involves concurrent administration of a protease inhibitor, such as saquinavir, plus a potent inhibitor of cytochrome P450 (CYP) 3A4, usually ritonavir in subtherapeutic doses. Ritonavir 182-191 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 147-172 12499178-1 2003 The protease inhibitor (PI) ritonavir is used as a strong inhibitor of cytochrome P450 3A4, which boosts the activities of coadministered PIs, resulting in augmented plasma PI levels, simplification of the dosage regimen, and better efficacy against resistant viruses. Ritonavir 28-37 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 71-90 12949438-6 2003 Serious cases by coadministration of CYP3A inhibitors resulting in acute hepatitis, hypotension, rhabdomyolyis, torsade de pointes, sedation, or ergotism are presented: interactions with azole antifungals (ketoconazole, itraconazole, fluconazole), HIV protease inhibitors (ritonavir, indinavir, saquinavir, nelfinavir), macrolide antibiotics (clarithromycin, erythromycin), and grapefruit juice. Ritonavir 273-282 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 37-42 12790697-2 2003 Since protease inhibitors are extensively metabolised by CYP3A4, this results in a marked increase in systemic exposure of saquinavir or other protease inhibitors boosted by ritonavir. Ritonavir 174-183 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 57-63 12460905-9 2002 The net effect of low micromolar concentrations of ritonavir on the chymotrypsin-like activity in cells and cell lysates was a weak inhibition, consistent with marginal alterations of polyubiquitinated proteins, marginal alterations in acid-soluble proteolytic peptide levels, and a small accumulation of the tumor suppressor protein p53, in cells treated with ritonavir. Ritonavir 51-60 transformation related protein 53, pseudogene Mus musculus 334-337 12518028-1 2003 The objective of our research was to investigate the effects of the protease inhibitors ritonavir, saquinavir, and indinavir on triglyceride synthesis, lipolysis, insulin binding, and signaling in differentiating 3T3 L1 pre-adipocytes. Ritonavir 88-97 insulin Homo sapiens 163-170 12518028-7 2003 During differentiation with ritonavir (i.e., 1-11 days post addition of differentiating cocktail), insulin receptor substrate-1 (IRS-1) tyrosine phosphorylation was ascertained (day 11) and found to be decreased in the ritonavir exposed cells when compared with control cells. Ritonavir 219-228 insulin receptor substrate 1 Homo sapiens 99-127 12518028-7 2003 During differentiation with ritonavir (i.e., 1-11 days post addition of differentiating cocktail), insulin receptor substrate-1 (IRS-1) tyrosine phosphorylation was ascertained (day 11) and found to be decreased in the ritonavir exposed cells when compared with control cells. Ritonavir 219-228 insulin receptor substrate 1 Homo sapiens 129-134 12662125-40 2003 Coadministration of lopinavir/ritonavir is contraindicated with certain drugs (i.e. flecainide, propafenone, astemizole, terfenadine, ergot derivatives, cisapride, pimozide, midazolam and triazolam) that are highly dependent on CYP3A or CYP2D6 for clearance and for which elevated plasma concentrations are associated with serious and/or life-threatening events. Ritonavir 30-39 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 237-243 12460905-11 2002 We demonstrate that p21 accumulation in the presence of ritonavir is attributable to the inhibition of proteolytic degradation. Ritonavir 56-65 cyclin-dependent kinase inhibitor 1A (P21) Mus musculus 20-23 12444650-1 2002 Stereodivergent synthesis of the diamino alcohol core of ritonavir and its C-2 epimer. Ritonavir 57-66 complement C2 Homo sapiens 75-78 12441801-3 2002 OBJECTIVES: To investigate whether the ABC transporters MRP1, MRP2, MRP3, MRP5 and breast cancer resistance protein 1 (Bcrp1) are efficient transporters of the HPI saquinavir, ritonavir and indinavir. Ritonavir 176-185 ATP binding cassette subfamily C member 3 Homo sapiens 68-72 12441801-3 2002 OBJECTIVES: To investigate whether the ABC transporters MRP1, MRP2, MRP3, MRP5 and breast cancer resistance protein 1 (Bcrp1) are efficient transporters of the HPI saquinavir, ritonavir and indinavir. Ritonavir 176-185 ATP binding cassette subfamily C member 5 Homo sapiens 74-78 12441801-3 2002 OBJECTIVES: To investigate whether the ABC transporters MRP1, MRP2, MRP3, MRP5 and breast cancer resistance protein 1 (Bcrp1) are efficient transporters of the HPI saquinavir, ritonavir and indinavir. Ritonavir 176-185 ATP binding cassette subfamily C member 1 Homo sapiens 56-60 12441801-5 2002 RESULTS: MRP2 efficiently transported saquinavir, ritonavir and indinavir and this transport could be enhanced by probenecid. Ritonavir 50-59 ATP binding cassette subfamily C member 2 Homo sapiens 9-13 12441801-3 2002 OBJECTIVES: To investigate whether the ABC transporters MRP1, MRP2, MRP3, MRP5 and breast cancer resistance protein 1 (Bcrp1) are efficient transporters of the HPI saquinavir, ritonavir and indinavir. Ritonavir 176-185 ATP binding cassette subfamily C member 2 Homo sapiens 62-66 12444942-0 2002 Ritonavir plus saquinavir versus single protease inhibitor therapy in protease inhibitor-naive HIV-infected patients: the Swiss HIV Cohort Study. Ritonavir 0-9 serpin family A member 13, pseudogene Homo sapiens 70-88 12386120-6 2002 The increased tissue exposure to DPC in rats could largely be attributed to inhibition of CYP3A1/2 by RTV. Ritonavir 102-105 cytochrome P450, family 3, subfamily a, polypeptide 23-polypeptide 1 Rattus norvegicus 90-96 12386120-8 2002 The Pgp-mediated transport of DPC across Caco-2 cells was readily saturated at >or=10 microM and was inhibited significantly by RTV at 5 to 10 microM. Ritonavir 131-134 ATP binding cassette subfamily B member 1 Homo sapiens 4-7 12386120-9 2002 The data above and the reported RTV concentrations suggested that both the Pgp and CYP3A4 inhibition by RTV may play a significant role in enhancing the systemic and tissue exposure to DPC in humans. Ritonavir 32-35 ATP binding cassette subfamily B member 1 Homo sapiens 75-78 12386120-9 2002 The data above and the reported RTV concentrations suggested that both the Pgp and CYP3A4 inhibition by RTV may play a significant role in enhancing the systemic and tissue exposure to DPC in humans. Ritonavir 32-35 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 83-89 12386120-9 2002 The data above and the reported RTV concentrations suggested that both the Pgp and CYP3A4 inhibition by RTV may play a significant role in enhancing the systemic and tissue exposure to DPC in humans. Ritonavir 104-107 ATP binding cassette subfamily B member 1 Homo sapiens 75-78 12386120-9 2002 The data above and the reported RTV concentrations suggested that both the Pgp and CYP3A4 inhibition by RTV may play a significant role in enhancing the systemic and tissue exposure to DPC in humans. Ritonavir 104-107 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 83-89 12490421-1 2002 Low doses of ritonavir, a strong inhibitor of cytochrome P450 3A4, enhances the pharmacokinetic profile of indinavir with increased serum levels. Ritonavir 13-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 46-65 12218384-0 2002 The protease inhibitor ritonavir inhibits the functional activity of the multidrug resistance related-protein 1 (MRP-1). Ritonavir 23-32 ATP binding cassette subfamily C member 1 Homo sapiens 113-118 12203188-2 2002 Further clinical evaluation revealed, however, that the patient had developed exogenous Cushing syndrome, which presumably was caused by the inhibition of CYP3A4"s metabolism of inhaled fluticasone by the protease inhibitor ritonavir. Ritonavir 224-233 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 155-161 12218384-0 2002 The protease inhibitor ritonavir inhibits the functional activity of the multidrug resistance related-protein 1 (MRP-1). Ritonavir 23-32 ATP binding cassette subfamily C member 1 Homo sapiens 73-111 12356805-0 2002 P-glycoprotein and MRP1 expression and reduced ritonavir and saquinavir accumulation in HIV-infected individuals. Ritonavir 47-56 ATP binding cassette subfamily C member 1 Homo sapiens 19-23 12356805-2 2002 As the HIV protease inhibitors are substrates for the efflux transporters P-glycoprotein and MRP, we wished to investigate whether differences in expression of these transporters on human lymphocytes correlated with intracellular concentrations of ritonavir and saquinavir. Ritonavir 248-257 ATP binding cassette subfamily B member 1 Homo sapiens 74-88 12356805-2 2002 As the HIV protease inhibitors are substrates for the efflux transporters P-glycoprotein and MRP, we wished to investigate whether differences in expression of these transporters on human lymphocytes correlated with intracellular concentrations of ritonavir and saquinavir. Ritonavir 248-257 ATP binding cassette subfamily C member 1 Homo sapiens 93-96 12356805-6 2002 RESULTS: Patients with lower MRP1 expression (<median) had a significantly higher accumulation of both ritonavir and saquinavir than those with higher MRP1 expression (P = 0.035, CI = -1.70 to -0.06 and P = 0.043, CI = -12.79 to -0.11, respectively). Ritonavir 106-115 ATP binding cassette subfamily C member 1 Homo sapiens 29-33 12356805-7 2002 Ritonavir accumulation was significantly greater in patients with lower P-glycoprotein expression (<median) than in patients with higher expression (P = 0.014, CI = -1.56 to -0.14). Ritonavir 0-9 ATP binding cassette subfamily B member 1 Homo sapiens 72-86 12356805-9 2002 Combining expression of P-glycoprotein and MRP1 (expression index, EI = [(P-glycoprotein - 1) + (MRP1 - 1) x 100]) resulted in a statistically significant relationship between transporter expression and intracellular accumulation of both saquinavir (r(2) = 0.195, P = 0.035) and ritonavir (r(2) = 0.220, P = 0.049). Ritonavir 279-288 ATP binding cassette subfamily B member 1 Homo sapiens 24-38 12356805-9 2002 Combining expression of P-glycoprotein and MRP1 (expression index, EI = [(P-glycoprotein - 1) + (MRP1 - 1) x 100]) resulted in a statistically significant relationship between transporter expression and intracellular accumulation of both saquinavir (r(2) = 0.195, P = 0.035) and ritonavir (r(2) = 0.220, P = 0.049). Ritonavir 279-288 ATP binding cassette subfamily C member 1 Homo sapiens 43-47 12356805-9 2002 Combining expression of P-glycoprotein and MRP1 (expression index, EI = [(P-glycoprotein - 1) + (MRP1 - 1) x 100]) resulted in a statistically significant relationship between transporter expression and intracellular accumulation of both saquinavir (r(2) = 0.195, P = 0.035) and ritonavir (r(2) = 0.220, P = 0.049). Ritonavir 279-288 ATP binding cassette subfamily B member 1 Homo sapiens 74-92 12356805-9 2002 Combining expression of P-glycoprotein and MRP1 (expression index, EI = [(P-glycoprotein - 1) + (MRP1 - 1) x 100]) resulted in a statistically significant relationship between transporter expression and intracellular accumulation of both saquinavir (r(2) = 0.195, P = 0.035) and ritonavir (r(2) = 0.220, P = 0.049). Ritonavir 279-288 ATP binding cassette subfamily C member 1 Homo sapiens 97-101 12356805-10 2002 CONCLUSION: Increased expression of P-glycoprotein and MRP1 on lymphocytes is associated with lower intracellular accumulation of saquinavir and ritonavir. Ritonavir 145-154 ATP binding cassette subfamily B member 1 Homo sapiens 36-50 12356805-10 2002 CONCLUSION: Increased expression of P-glycoprotein and MRP1 on lymphocytes is associated with lower intracellular accumulation of saquinavir and ritonavir. Ritonavir 145-154 ATP binding cassette subfamily C member 1 Homo sapiens 55-59 12218384-4 2002 OBJECTIVE: To analyze the inhibitory potential of the anti-retroviral drugs indinavir, amprenavir, ritonavir, lamivudine or zidovudine to modulate MRP-1 function. Ritonavir 99-108 ATP binding cassette subfamily C member 1 Homo sapiens 147-152 12218384-8 2002 RESULTS: Ritonavir inhibited the functional activity of MRP-1 similarly to probenecid, as demonstrated by re-sensitization of MRP-1 over-expressing cells to cytotoxic effects of etoposide. Ritonavir 9-18 ATP binding cassette subfamily C member 1 Homo sapiens 56-61 12218384-8 2002 RESULTS: Ritonavir inhibited the functional activity of MRP-1 similarly to probenecid, as demonstrated by re-sensitization of MRP-1 over-expressing cells to cytotoxic effects of etoposide. Ritonavir 9-18 ATP binding cassette subfamily C member 1 Homo sapiens 126-131 12218384-11 2002 CONCLUSIONS: These data may be exploitable to further improve sanctuary site concentrations of anti-HIV or anti-cancer drugs by using ritonavir as a lead compound to develop more potent MRP-1 inhibitors. Ritonavir 134-143 ATP binding cassette subfamily C member 1 Homo sapiens 186-191 12235177-4 2002 Results showed that ritonavir increased VLDL triglyceride production by 30% over a 4 h period when mice were fed the low-fat basal diet. Ritonavir 20-29 CD320 antigen Mus musculus 40-44 12186895-5 2002 Processing of Vif required an active viral Pr and was sensitive to Pr inhibitors such as ritonavir. Ritonavir 89-98 vif protein Simian immunodeficiency virus 14-17 12235177-5 2002 The ritonavir effect was more pronounced under high-fat feeding conditions, with a 2-fold increase in VLDL triglyceride production rate. Ritonavir 4-13 CD320 antigen Mus musculus 102-106 12065438-8 2002 Ritonavir and troleandomycin showed marked PXR activation but no increase (in the case of troleandomycin) or a significant decrease (in the case of ritonavir) in microsomal CYP3A4 activity. Ritonavir 0-9 nuclear receptor subfamily 1 group I member 2 Homo sapiens 43-46 12086554-9 2002 Ritonavir potently inhibits CYP3A4 and is used to enhance the systemic exposure of lopinavir. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 28-34 12172335-2 2002 The atypical antipsychotic olanzapine is metabolized primarily by CYP1A2 and glucuronosyl transferases, both of which are induced by the HIV protease inhibitor ritonavir. Ritonavir 160-169 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 66-72 11914372-6 2002 We also find that Rit-mediated effects on neurite outgrowth can be blocked by co-expression of dominant-negative mutants of C-Raf1 or mitogen-activated protein kinase kinase 1 (MEK1). Ritonavir 18-21 v-raf-leukemia viral oncogene 1 Mus musculus 124-130 11914372-6 2002 We also find that Rit-mediated effects on neurite outgrowth can be blocked by co-expression of dominant-negative mutants of C-Raf1 or mitogen-activated protein kinase kinase 1 (MEK1). Ritonavir 18-21 mitogen-activated protein kinase kinase 1 Mus musculus 134-175 11914372-6 2002 We also find that Rit-mediated effects on neurite outgrowth can be blocked by co-expression of dominant-negative mutants of C-Raf1 or mitogen-activated protein kinase kinase 1 (MEK1). Ritonavir 18-21 mitogen-activated protein kinase kinase 1 Mus musculus 177-181 11914372-9 2002 Instead, pharmacological inhibitors of MEK block Rit-stimulated cell survival. Ritonavir 49-52 midkine Mus musculus 39-42 11918509-5 2002 DISCUSSION: RTV is an inducer of the hepatic isoenzymes CYP1A2, CYP1A4, and CYP2C9/19 and leads to extensive metabolism of acenocoumarol that cannot be balanced by dose increases. Ritonavir 12-15 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 56-62 12079557-7 2002 Addition of AZT or ritonavir before or after establishment of productive HIV infection dramatically reduces virus replication and M-CSF production by human MDMs. Ritonavir 19-28 colony stimulating factor 1 Homo sapiens 130-135 12112443-9 2002 In addition, results from functional studies show that the accumulation of the P-glycoprotein substrate digoxin by RBE4 monolayer cells is significantly enhanced in the presence of standard P-glycoprotein inhibitors (verapamil, cyclosporin A, PSC 833), protease inhibitors (saquinavir, ritonavir, indinavir), and the metabolic inhibitor, sodium azide. Ritonavir 286-295 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 79-93 11978161-12 2002 Based on the pharmacokinetics of these medications, we hypothesize that inhibition of CYP2D6 and CYP3A4 by ritonavir and indinavir may have resulted in an accumulation of the active moiety of risperidone, which may explain the occurrence of EPS in this patient. Ritonavir 107-116 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 86-92 11978161-12 2002 Based on the pharmacokinetics of these medications, we hypothesize that inhibition of CYP2D6 and CYP3A4 by ritonavir and indinavir may have resulted in an accumulation of the active moiety of risperidone, which may explain the occurrence of EPS in this patient. Ritonavir 107-116 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 97-103 11986235-6 2002 Transcriptional activation of nuclear factor-kappaB, as induced by the KS-promoting factor TNF-alpha, the HIV-1 Tat protein, or the human herpesvirus 8 protein ORF74, was inhibited by ritonavir. Ritonavir 184-193 ORF74 Human gammaherpesvirus 8 160-165 11972482-7 2002 New IgE sensitizations developed in 65% of the birch RIT-treated patients when studied by immunoblotting. Ritonavir 53-56 immunoglobulin heavy constant epsilon Homo sapiens 4-7 11918509-5 2002 DISCUSSION: RTV is an inducer of the hepatic isoenzymes CYP1A2, CYP1A4, and CYP2C9/19 and leads to extensive metabolism of acenocoumarol that cannot be balanced by dose increases. Ritonavir 12-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 12206135-3 2002 In addition, MRP2 levels are altered in hepatocytes in response to hormones such as glucocorticoids and to structurally unrelated drugs such as rifampicin, phenobarbital, ritonavir, and cisplatin. Ritonavir 171-180 ATP binding cassette subfamily C member 2 Homo sapiens 13-17 12405866-6 2002 Ritonavir is the most potent inhibitor of CYP3A4. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 42-48 12405866-18 2002 Losartan may have increased effect when coadministered with ritonavir and nelfinavir because of the induction of CYP2C9 and the expected increase in formation of the active metabolite, E-3174. Ritonavir 60-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-119 11849198-12 2002 P-glycoprotein mediated drug efflux was determined as a function of rhodamine efflux in the absence and presence of ritonavir. Ritonavir 116-125 ATP binding cassette subfamily B member 1 Homo sapiens 0-14 11849198-19 2002 Ritonavir (5 microm) inhibited P-glycoprotein mediated efflux in both groups producing greater intracellular accumulation of rhodamine. Ritonavir 0-9 ATP binding cassette subfamily B member 1 Homo sapiens 31-45 11878403-11 2001 Many combinations contain ritonavir because this PI has the most pronounced inhibiting effects on CYP3A4. Ritonavir 26-35 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 98-104 12593790-0 2002 Ritonavir and Saquinavir directly stimulate anterior pituitary prolactin secretion, in vitro. Ritonavir 0-9 prolactin Homo sapiens 63-72 12593790-2 2002 This could be explained by a direct effect of ritonavir and saquinavir on anterior pituitary prolactin (PRL) release, and/or an indirect effect of PIs on the secretion of hypothalamic dopamine, which is the main PRL inhibitory factor. Ritonavir 46-55 prolactin Homo sapiens 93-102 11719726-2 2001 The absence of marked central opioid effects has been attributed to its low bioavailability and its poor penetration of the blood-brain barrier, both of which might be altered by ritonavir, a potent P-glycoprotein and cytochrome P4503A inhibitor. Ritonavir 179-188 ATP binding cassette subfamily B member 1 Homo sapiens 199-213 11745741-0 2001 Ritonavir induces P-glycoprotein expression, multidrug resistance-associated protein (MRP1) expression, and drug transporter-mediated activity in a human intestinal cell line. Ritonavir 0-9 ATP binding cassette subfamily B member 1 Homo sapiens 18-32 11745741-0 2001 Ritonavir induces P-glycoprotein expression, multidrug resistance-associated protein (MRP1) expression, and drug transporter-mediated activity in a human intestinal cell line. Ritonavir 0-9 ATP binding cassette subfamily C member 1 Homo sapiens 45-90 11745741-1 2001 The present study characterized the response of P-glycoprotein (P-gp) and multidrug resistance-associated protein (MRP1) to chronic ritonavir (RIT) exposure by assessing increases in P-gp and MRP1 protein expression and activity. Ritonavir 132-141 ATP binding cassette subfamily B member 1 Homo sapiens 48-62 11745741-1 2001 The present study characterized the response of P-glycoprotein (P-gp) and multidrug resistance-associated protein (MRP1) to chronic ritonavir (RIT) exposure by assessing increases in P-gp and MRP1 protein expression and activity. Ritonavir 132-141 ATP binding cassette subfamily C member 3 Homo sapiens 64-119 11745741-1 2001 The present study characterized the response of P-glycoprotein (P-gp) and multidrug resistance-associated protein (MRP1) to chronic ritonavir (RIT) exposure by assessing increases in P-gp and MRP1 protein expression and activity. Ritonavir 132-141 ATP binding cassette subfamily B member 1 Homo sapiens 64-68 11745741-1 2001 The present study characterized the response of P-glycoprotein (P-gp) and multidrug resistance-associated protein (MRP1) to chronic ritonavir (RIT) exposure by assessing increases in P-gp and MRP1 protein expression and activity. Ritonavir 132-141 ATP binding cassette subfamily C member 1 Homo sapiens 115-119 11745741-1 2001 The present study characterized the response of P-glycoprotein (P-gp) and multidrug resistance-associated protein (MRP1) to chronic ritonavir (RIT) exposure by assessing increases in P-gp and MRP1 protein expression and activity. Ritonavir 143-146 ATP binding cassette subfamily B member 1 Homo sapiens 48-62 11745741-1 2001 The present study characterized the response of P-glycoprotein (P-gp) and multidrug resistance-associated protein (MRP1) to chronic ritonavir (RIT) exposure by assessing increases in P-gp and MRP1 protein expression and activity. Ritonavir 143-146 ATP binding cassette subfamily C member 3 Homo sapiens 64-119 11745741-1 2001 The present study characterized the response of P-glycoprotein (P-gp) and multidrug resistance-associated protein (MRP1) to chronic ritonavir (RIT) exposure by assessing increases in P-gp and MRP1 protein expression and activity. Ritonavir 143-146 ATP binding cassette subfamily B member 1 Homo sapiens 64-68 11745741-1 2001 The present study characterized the response of P-glycoprotein (P-gp) and multidrug resistance-associated protein (MRP1) to chronic ritonavir (RIT) exposure by assessing increases in P-gp and MRP1 protein expression and activity. Ritonavir 143-146 ATP binding cassette subfamily C member 1 Homo sapiens 115-119 11745741-5 2001 RIT strongly induced P-gp and MRP1 expression (maximum 6-fold and 3-fold increases, respectively) in a concentration-dependent fashion. Ritonavir 0-3 ATP binding cassette subfamily B member 1 Homo sapiens 21-25 11745741-5 2001 RIT strongly induced P-gp and MRP1 expression (maximum 6-fold and 3-fold increases, respectively) in a concentration-dependent fashion. Ritonavir 0-3 ATP binding cassette subfamily C member 1 Homo sapiens 30-34 11468419-4 2001 We found that HIV-PIs (i.e., ritonavir, saquinavir, nelfinavir and indinavir) interfere with P-gp function in normal PBLs as demonstrated by the reduced efflux of rhodamine 123 (Rh123). Ritonavir 29-38 ATP binding cassette subfamily B member 1 Homo sapiens 93-97 11603001-3 2001 Human recombinant alpha interferon (IFN) was administered as 4 subcutaneous injections of 3 x 10(6) U on alternate days beginning 5 days before RIT to increase the expression of the tumor-associated antigen, TAG-72. Ritonavir 144-147 interferon alpha 1 Homo sapiens 18-40 11590527-1 2001 PURPOSE: Ritonavir (RTV) and delavirdine (DLV) are inhibitors of cytochrome P450 (CYP) 3A4, the specific CYP that metabolizes indinavir (IDV). Ritonavir 9-18 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 65-90 11590527-1 2001 PURPOSE: Ritonavir (RTV) and delavirdine (DLV) are inhibitors of cytochrome P450 (CYP) 3A4, the specific CYP that metabolizes indinavir (IDV). Ritonavir 20-23 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 65-90 11259625-0 2001 P-glycoprotein limits oral availability, brain, and fetal penetration of saquinavir even with high doses of ritonavir. Ritonavir 108-117 phosphoglycolate phosphatase Mus musculus 0-14 11475806-5 2001 Ritonavir, a potent CYP3A4 inhibitor, is coadministered with saquinavir, indinavir and amprenavir to enhance their plasma concentrations and their virological efficacy. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 20-26 11259625-2 2001 Because saquinavir and ritonavir are substrates and inhibitors of both the drug transporter P-glycoprotein (P-gp) and of the metabolizing enzyme CYP3A4, we wanted to sort out whether the ritonavir effect is primarily mediated by inhibition of CYP3A4 or P-gp or both. Ritonavir 23-32 phosphoglycolate phosphatase Mus musculus 92-106 11259625-2 2001 Because saquinavir and ritonavir are substrates and inhibitors of both the drug transporter P-glycoprotein (P-gp) and of the metabolizing enzyme CYP3A4, we wanted to sort out whether the ritonavir effect is primarily mediated by inhibition of CYP3A4 or P-gp or both. Ritonavir 23-32 phosphoglycolate phosphatase Mus musculus 108-112 11259625-2 2001 Because saquinavir and ritonavir are substrates and inhibitors of both the drug transporter P-glycoprotein (P-gp) and of the metabolizing enzyme CYP3A4, we wanted to sort out whether the ritonavir effect is primarily mediated by inhibition of CYP3A4 or P-gp or both. Ritonavir 23-32 phosphoglycolate phosphatase Mus musculus 253-257 11259625-2 2001 Because saquinavir and ritonavir are substrates and inhibitors of both the drug transporter P-glycoprotein (P-gp) and of the metabolizing enzyme CYP3A4, we wanted to sort out whether the ritonavir effect is primarily mediated by inhibition of CYP3A4 or P-gp or both. Ritonavir 187-196 phosphoglycolate phosphatase Mus musculus 92-106 11259625-2 2001 Because saquinavir and ritonavir are substrates and inhibitors of both the drug transporter P-glycoprotein (P-gp) and of the metabolizing enzyme CYP3A4, we wanted to sort out whether the ritonavir effect is primarily mediated by inhibition of CYP3A4 or P-gp or both. Ritonavir 187-196 phosphoglycolate phosphatase Mus musculus 108-112 11259625-3 2001 P-gp is known to limit the bioavailability, brain, testis, and fetal penetration of its substrates, so effective inhibition of P-gp by ritonavir in vivo might open up pharmacological sanctuary sites for saquinavir, with the potential of beneficial effects on therapy, but also of increased toxicity. Ritonavir 135-144 phosphoglycolate phosphatase Mus musculus 0-4 11259625-3 2001 P-gp is known to limit the bioavailability, brain, testis, and fetal penetration of its substrates, so effective inhibition of P-gp by ritonavir in vivo might open up pharmacological sanctuary sites for saquinavir, with the potential of beneficial effects on therapy, but also of increased toxicity. Ritonavir 135-144 phosphoglycolate phosphatase Mus musculus 127-131 11093774-7 2000 Finally, the HIV protease inhibitors saquinavir and ritonavir were potent inhibitors of transport mediated by both P-glycoprotein and Mrp. Ritonavir 52-61 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 115-129 11259625-4 2001 In vitro, P-gp-mediated transport of saquinavir and ritonavir was only moderately inhibited by both HPIs compared with the potent P-gp inhibitor PSC833. Ritonavir 52-61 phosphoglycolate phosphatase Mus musculus 10-14 11259625-4 2001 In vitro, P-gp-mediated transport of saquinavir and ritonavir was only moderately inhibited by both HPIs compared with the potent P-gp inhibitor PSC833. Ritonavir 52-61 phosphoglycolate phosphatase Mus musculus 130-134 11259625-6 2001 These data indicate that in vivo, ritonavir is a relatively poor P-gp inhibitor. Ritonavir 34-43 phosphoglycolate phosphatase Mus musculus 65-69 11242145-5 2001 Ritonavir and indinavir also showed polarized transport in the LLC-PK1 and LLC-PK1:MRP1 cell line, which could be inhibited by probenecid. Ritonavir 0-9 LOW QUALITY PROTEIN: multidrug resistance-associated protein 6 Sus scrofa 83-87 11159797-0 2001 Ritonavir, efavirenz, and nelfinavir inhibit CYP2B6 activity in vitro: potential drug interactions with bupropion. Ritonavir 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 45-51 11596909-3 2001 Ritonavir reduces the metabolization of lopinavir by the cytochrome P450 3A4 isoenzyme which leads to markedly increased plasma levels of lopinavir(4). Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 57-76 11134271-0 2001 Human immunodeficiency virus type 1 protease cleavage site mutations associated with protease inhibitor cross-resistance selected by indinavir, ritonavir, and/or saquinavir. Ritonavir 144-153 serpin family A member 13, pseudogene Homo sapiens 85-103 11134271-1 2001 We examined the prevalence of cleavage site mutations, both within and outside the gag region, in 28 protease inhibitor (PI) cross-resistant patients treated with indinavir, ritonavir, and/or saquinavir compared to control patients treated with reverse transcriptase inhibitors. Ritonavir 174-183 serpin family A member 13, pseudogene Homo sapiens 101-119 11155727-4 2000 Ritonavir and verapamil reduce the impact of PGP efflux. Ritonavir 0-9 phosphoglycolate phosphatase Homo sapiens 45-48 11155727-6 2000 IN VIVO: In vitro studies demonstrated that ritonavir can inverse the effect resulting from the expression of the MDR gene and the production of PGP, with restoration of significant intracellular levels of protease inhibitors, at least higher than obtained in the absence of ritonavir. Ritonavir 44-53 phosphoglycolate phosphatase Homo sapiens 145-148 11093774-7 2000 Finally, the HIV protease inhibitors saquinavir and ritonavir were potent inhibitors of transport mediated by both P-glycoprotein and Mrp. Ritonavir 52-61 ATP binding cassette subfamily C member 2 Rattus norvegicus 134-137 11101056-0 2000 Ritonavir increases the level of active ADD-1/SREBP-1 protein during adipogenesis. Ritonavir 0-9 adducin 1 Homo sapiens 40-45 11101056-0 2000 Ritonavir increases the level of active ADD-1/SREBP-1 protein during adipogenesis. Ritonavir 0-9 sterol regulatory element binding transcription factor 1 Homo sapiens 46-53 11020127-9 2000 Ritonavir is a potent inhibitor of hepatic cytochrome P450, mainly the CYP3A4 isoform. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 71-77 11101056-8 2000 RESULTS: Ritonavir (10 microg/ml) enhanced 3T3-L1 preadipocyte differentiation (30% increase in TG mass; 50% increase in GPDH activity), and transiently raised levels of the 68 kDa active mature form of ADD-1/SREBP-1 during adipogenesis by threefold, compared with standard differentiation. Ritonavir 9-18 adducin 1 Homo sapiens 203-208 11101056-8 2000 RESULTS: Ritonavir (10 microg/ml) enhanced 3T3-L1 preadipocyte differentiation (30% increase in TG mass; 50% increase in GPDH activity), and transiently raised levels of the 68 kDa active mature form of ADD-1/SREBP-1 during adipogenesis by threefold, compared with standard differentiation. Ritonavir 9-18 sterol regulatory element binding transcription factor 1 Homo sapiens 209-216 11101056-9 2000 In contrast, ritonavir attenuated the differentiation-induced increase in CEBPalpha and PPARgamma. Ritonavir 13-22 CCAAT enhancer binding protein alpha Homo sapiens 74-83 11101056-9 2000 In contrast, ritonavir attenuated the differentiation-induced increase in CEBPalpha and PPARgamma. Ritonavir 13-22 peroxisome proliferator activated receptor gamma Homo sapiens 88-97 11101056-10 2000 CONCLUSIONS: Our data suggest that ritonavir enhances 3T3-L1 adipogenesis by increasing the level of active mature ADD-1/SREBP-1. Ritonavir 35-44 adducin 1 Homo sapiens 115-120 11101056-10 2000 CONCLUSIONS: Our data suggest that ritonavir enhances 3T3-L1 adipogenesis by increasing the level of active mature ADD-1/SREBP-1. Ritonavir 35-44 sterol regulatory element binding transcription factor 1 Homo sapiens 121-128 11101056-11 2000 This effect may be due to reduced proteolysis of ADD-1/SREBP-1, as ritonavir inhibits an N-acetyl-leucyl-leucyl-norleucinal (ALLN)-sensitive proteosomal degradation pathway in lymphocytes, and ALLN itself inhibits the breakdown of mature ADD-1/SREBP-1. Ritonavir 67-76 adducin 1 Homo sapiens 49-54 11101056-11 2000 This effect may be due to reduced proteolysis of ADD-1/SREBP-1, as ritonavir inhibits an N-acetyl-leucyl-leucyl-norleucinal (ALLN)-sensitive proteosomal degradation pathway in lymphocytes, and ALLN itself inhibits the breakdown of mature ADD-1/SREBP-1. Ritonavir 67-76 sterol regulatory element binding transcription factor 1 Homo sapiens 55-62 11101056-11 2000 This effect may be due to reduced proteolysis of ADD-1/SREBP-1, as ritonavir inhibits an N-acetyl-leucyl-leucyl-norleucinal (ALLN)-sensitive proteosomal degradation pathway in lymphocytes, and ALLN itself inhibits the breakdown of mature ADD-1/SREBP-1. Ritonavir 67-76 adducin 1 Homo sapiens 238-243 11101056-11 2000 This effect may be due to reduced proteolysis of ADD-1/SREBP-1, as ritonavir inhibits an N-acetyl-leucyl-leucyl-norleucinal (ALLN)-sensitive proteosomal degradation pathway in lymphocytes, and ALLN itself inhibits the breakdown of mature ADD-1/SREBP-1. Ritonavir 67-76 sterol regulatory element binding transcription factor 1 Homo sapiens 244-251 11101056-12 2000 As mature ADD-1/SREBP-1 regulates several lipogenic enzymes, higher levels may explain the effect of ritonavir on TG accumulation and GPDH activity. Ritonavir 101-110 adducin 1 Homo sapiens 10-15 11101056-12 2000 As mature ADD-1/SREBP-1 regulates several lipogenic enzymes, higher levels may explain the effect of ritonavir on TG accumulation and GPDH activity. Ritonavir 101-110 sterol regulatory element binding transcription factor 1 Homo sapiens 16-23 10996400-4 2000 In these cells the PIs, nelfinavir, saquinavir, and ritonavir, reduced triglyceride accumulation, lipogenesis, and expression of the adipose markers, aP2 and LPL. Ritonavir 52-61 transcription factor AP-2, alpha Mus musculus 150-153 10996400-4 2000 In these cells the PIs, nelfinavir, saquinavir, and ritonavir, reduced triglyceride accumulation, lipogenesis, and expression of the adipose markers, aP2 and LPL. Ritonavir 52-61 lipoprotein lipase Mus musculus 158-161 10930961-14 2000 CONCLUSIONS: These results indicate that both saquinavir and ritonavir modify the pharmacokinetics of sildenafil presumably through inhibition of CYP3A4. Ritonavir 61-70 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 146-152 10930961-15 2000 The more pronounced effect of ritonavir may be attributed to its additional potent inhibition of CYP2C9. Ritonavir 30-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 10926350-13 2000 The HIV protease inhibitors amprenavir, indinavir, nelfinavir, ritonavir and saquinavir inhibit CYP3A4. Ritonavir 63-72 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 96-102 11020127-11 2000 Ritonavir acted as a CYP3A4 inhibitor, diminishing carbamazepine metabolism and provoking an increase in serum levels and clinical toxicity. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 21-27 10952482-0 2000 Potent mechanism-based inhibition of human CYP3A in vitro by amprenavir and ritonavir: comparison with ketoconazole. Ritonavir 76-85 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 43-48 10845927-9 2000 This new rIT is a promising candidate for further clinical evaluation in patients with Hodgkin lymphoma or other CD30(+) malignancies. Ritonavir 9-12 TNF receptor superfamily member 8 Homo sapiens 113-117 10952482-5 2000 CONCLUSIONS: Thus, ritonavir and amprenavir are highly potent mechanism-based inhibitors of human CYP3A isoforms. Ritonavir 19-28 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 98-103 10745059-9 2000 CONCLUSIONS: CYP3A-mediated clearance of trazodone is inhibited by ketoconazole, ritonavir and indinavir, and indicates the likelihood of pharmacokinetic interactions in vivo. Ritonavir 81-90 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 13-18 10935688-1 2000 BACKGROUND: The viral protease inhibitor ritonavir has the capacity to inhibit and induce the activity of cytochrome P450-3A (CYP3A) isoforms, leading to drug interactions that may influence the efficacy and toxicity of other antiretroviral therapies, as well as pharmacologic treatments of coincident or complicating diseases. Ritonavir 41-50 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 106-124 10935688-1 2000 BACKGROUND: The viral protease inhibitor ritonavir has the capacity to inhibit and induce the activity of cytochrome P450-3A (CYP3A) isoforms, leading to drug interactions that may influence the efficacy and toxicity of other antiretroviral therapies, as well as pharmacologic treatments of coincident or complicating diseases. Ritonavir 41-50 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 126-131 10545207-9 1999 Although Rit, Rin, and Ras have highly related effector domain sequences, Rit and Rin were found to interact with the known Ras binding proteins RalGDS, Rlf, and AF-6/Canoe but not with the Raf kinases, RIN1, or the p110 subunit of phosphatidylinositol 3-kinase. Ritonavir 9-12 Ras like without CAAX 2 Homo sapiens 82-85 10801241-2 2000 The viral protease inhibitor ritonavir is of particular concern since it both inhibits and induces the activity of cytochrome P450 3A (CYP3A) isoforms. Ritonavir 29-38 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 115-133 10801241-2 2000 The viral protease inhibitor ritonavir is of particular concern since it both inhibits and induces the activity of cytochrome P450 3A (CYP3A) isoforms. Ritonavir 29-38 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 135-140 10801241-3 2000 METHODS: The inhibitory effect of ritonavir on the metabolism of alprazolam, a CYP3A-mediated reaction in humans, was tested in vitro using human liver microsomes. Ritonavir 34-43 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 79-84 10725306-5 2000 Sildenafil biotransformation (36 microM) was inhibited by increasing concentrations of ketoconazole and ritonavir (IC(50) values less than 0.02 microM), which are established cytochrome P450 (CYP) 3A4 inhibitors. Ritonavir 104-113 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 175-200 10714567-5 2000 RESULTS: Those taking ritonavir (n = 11) had significantly higher levels of plasma triglyceride, VLDL cholesterol, IDL cholesterol, apolipoprotein B, and lipoprotein (a) compared with placebo (n = 8). Ritonavir 22-31 apolipoprotein B Homo sapiens 132-148 10714567-7 2000 Post-heparin lipoprotein lipase (LpL) activity did not change but hepatic lipase activity decreased 20% (P < 0.01) in those taking ritonavir-compared with placebo. Ritonavir 134-143 lipase C, hepatic type Homo sapiens 66-80 10714567-9 2000 CONCLUSION: Treatment with ritonavir in the absence of HIV infection or changes in body composition results in hypertriglyceridemia that is apparently not mediated by impaired LpL activity or the defective removal of remnant lipoproteins, but could be caused by enhanced formation of VLDL. Ritonavir 27-36 lipoprotein lipase Homo sapiens 176-179 10668858-6 2000 Clinically important CYP3A4 inhibitors include itraconazole, ketoconazole, clarithromycin, erythromycin, nefazodone, ritonavir and grapefruit juice. Ritonavir 117-126 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 21-27 10641980-3 2000 Because ritonavir can induce CYP3A, it can decrease methadone plasma levels. Ritonavir 8-17 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 29-34 10545207-9 1999 Although Rit, Rin, and Ras have highly related effector domain sequences, Rit and Rin were found to interact with the known Ras binding proteins RalGDS, Rlf, and AF-6/Canoe but not with the Raf kinases, RIN1, or the p110 subunit of phosphatidylinositol 3-kinase. Ritonavir 9-12 ral guanine nucleotide dissociation stimulator Homo sapiens 145-151 10545207-9 1999 Although Rit, Rin, and Ras have highly related effector domain sequences, Rit and Rin were found to interact with the known Ras binding proteins RalGDS, Rlf, and AF-6/Canoe but not with the Raf kinases, RIN1, or the p110 subunit of phosphatidylinositol 3-kinase. Ritonavir 9-12 afadin, adherens junction formation factor Homo sapiens 162-166 10545207-10 1999 These interactions were GTP and effector domain dependent and suggest that RalGDS, Rlf, and AF-6 are Rit and Rin effectors. Ritonavir 101-104 ral guanine nucleotide dissociation stimulator Homo sapiens 75-81 10545207-10 1999 These interactions were GTP and effector domain dependent and suggest that RalGDS, Rlf, and AF-6 are Rit and Rin effectors. Ritonavir 101-104 RLF zinc finger Homo sapiens 83-86 10545207-10 1999 These interactions were GTP and effector domain dependent and suggest that RalGDS, Rlf, and AF-6 are Rit and Rin effectors. Ritonavir 101-104 afadin, adherens junction formation factor Homo sapiens 92-96 10509562-1 1999 OBJECTIVES: To determine the effect of the protease inhibitors ritonavir, nelfinavir and indinavir on the P-glycoprotein (P-gp)-mediated transport of saquinavir in Caco-2 cell monolayers. Ritonavir 63-72 ATP binding cassette subfamily B member 1 Homo sapiens 106-120 10589373-14 1999 Potent inhibitors of CYP3A (e.g., omeprazole and ritonavir) may also alter the metabolism of clarithromycin and its metabolites. Ritonavir 49-58 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 21-26 10509562-1 1999 OBJECTIVES: To determine the effect of the protease inhibitors ritonavir, nelfinavir and indinavir on the P-glycoprotein (P-gp)-mediated transport of saquinavir in Caco-2 cell monolayers. Ritonavir 63-72 ATP binding cassette subfamily B member 1 Homo sapiens 122-126 10509562-14 1999 CONCLUSIONS: We have demonstrated that saquinavir is a substrate for P-gp and that ritonavir, nelfinavir and indinavir modulate P-gp function in both human lymphocytes and Caco-2 cells. Ritonavir 83-92 ATP binding cassette subfamily B member 1 Homo sapiens 128-132 10509562-2 1999 To study the modulation of P-gp function in human lymphocytes by saquinavir, ritonavir, nelfinavir and indinavir. Ritonavir 77-86 ATP binding cassette subfamily B member 1 Homo sapiens 27-31 10509562-13 1999 The protease inhibitors displayed some P-gp modulation with ritonavir having the most potent effect. Ritonavir 60-69 ATP binding cassette subfamily B member 1 Homo sapiens 39-43 10421617-2 1999 In the present study, the effect of ritonavir, a potent inhibitor of cytochrome P-450 (CYP) 3A, on the in vitro metabolism of ABT-378 was examined. Ritonavir 36-45 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 69-94 10461827-3 1999 We investigated the effect of potent antiretroviral therapy (ritonavir and saquinavir) on the production of MIP-1alpha, MIP-1beta, and RANTES in 19 HIV-infected patients who had sustained decreases in plasma HIV RNA levels (<200 copies/ml). Ritonavir 61-70 C-C motif chemokine ligand 5 Homo sapiens 135-141 10419894-11 1999 In the presence of ritonavir, CD4(+) T cells from HIV-infected patients showed similar changes in ICE intracellular levels without alteration of Fas expression. Ritonavir 19-28 CD4 molecule Homo sapiens 30-33 10419894-11 1999 In the presence of ritonavir, CD4(+) T cells from HIV-infected patients showed similar changes in ICE intracellular levels without alteration of Fas expression. Ritonavir 19-28 caspase 1 Homo sapiens 98-101 10551732-4 1999 The susceptibility of PBMCs to apoptosis was markedly decreased after ritonavir treatment and correlated with lower levels of caspase-1 expression, decreases in annexin V staining, and reduced caspase-3 activity. Ritonavir 70-79 caspase 1 Homo sapiens 126-135 10551732-4 1999 The susceptibility of PBMCs to apoptosis was markedly decreased after ritonavir treatment and correlated with lower levels of caspase-1 expression, decreases in annexin V staining, and reduced caspase-3 activity. Ritonavir 70-79 caspase 3 Homo sapiens 193-202 10551732-5 1999 Induction in vitro of tumor necrosis factor (TNF) production by PBMCs and monocytes was inhibited by ritonavir in a time- and dose-dependent manner at nontoxic concentrations. Ritonavir 101-110 tumor necrosis factor Homo sapiens 22-43 10551732-5 1999 Induction in vitro of tumor necrosis factor (TNF) production by PBMCs and monocytes was inhibited by ritonavir in a time- and dose-dependent manner at nontoxic concentrations. Ritonavir 101-110 tumor necrosis factor Homo sapiens 45-48 10461827-3 1999 We investigated the effect of potent antiretroviral therapy (ritonavir and saquinavir) on the production of MIP-1alpha, MIP-1beta, and RANTES in 19 HIV-infected patients who had sustained decreases in plasma HIV RNA levels (<200 copies/ml). Ritonavir 61-70 C-C motif chemokine ligand 3 Homo sapiens 108-118 10461827-3 1999 We investigated the effect of potent antiretroviral therapy (ritonavir and saquinavir) on the production of MIP-1alpha, MIP-1beta, and RANTES in 19 HIV-infected patients who had sustained decreases in plasma HIV RNA levels (<200 copies/ml). Ritonavir 61-70 C-C motif chemokine ligand 4 Homo sapiens 120-129 10428109-13 1999 CONCLUSIONS: Ritonavir combination therapy results in prompt and sustained restoration of intestinal function, which is associated with reduction in viral load, increase in CD4 counts, and gain in body weight. Ritonavir 13-22 CD4 molecule Homo sapiens 173-176 10421617-8 1999 The ABT-378-ritonavir combinations (at 3:1 and 29:1 ratios) inhibited CYP3A (IC(50) = 1.1 and 4.6 microM), albeit less potently than ritonavir (IC(50) = 0.14 microM). Ritonavir 12-21 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 70-75 11230802-0 1999 HIV protease inhibitor ritonavir: a more potent inhibitor of P-glycoprotein than the cyclosporine analog SDZ PSC 833. Ritonavir 23-32 ATP binding cassette subfamily B member 1 Homo sapiens 61-75 10445808-1 1999 The phenotype of circulating CD8+ T lymphocytes and its association with plasma HIV-1 RNA were analyzed in 34 HIV-1-infected subjects, who were treated with saquinavir, ritonavir, and two nucleoside analogs (HAART) for 1 year. Ritonavir 169-178 CD8a molecule Homo sapiens 29-32 11230802-6 1999 In addition, the HIV protease inhibitor ritonavir inhibited p-glycoprotein-mediated extrusion of saquinavir with an IC(50) of 0.2 microM, indicating a high affinity of ritonavir for p-glycoprotein. Ritonavir 40-49 ATP binding cassette subfamily B member 1 Homo sapiens 60-74 11230802-6 1999 In addition, the HIV protease inhibitor ritonavir inhibited p-glycoprotein-mediated extrusion of saquinavir with an IC(50) of 0.2 microM, indicating a high affinity of ritonavir for p-glycoprotein. Ritonavir 40-49 ATP binding cassette subfamily B member 1 Homo sapiens 182-196 11230802-6 1999 In addition, the HIV protease inhibitor ritonavir inhibited p-glycoprotein-mediated extrusion of saquinavir with an IC(50) of 0.2 microM, indicating a high affinity of ritonavir for p-glycoprotein. Ritonavir 168-177 ATP binding cassette subfamily B member 1 Homo sapiens 60-74 11230802-6 1999 In addition, the HIV protease inhibitor ritonavir inhibited p-glycoprotein-mediated extrusion of saquinavir with an IC(50) of 0.2 microM, indicating a high affinity of ritonavir for p-glycoprotein. Ritonavir 168-177 ATP binding cassette subfamily B member 1 Homo sapiens 182-196 11230802-7 1999 In conclusion, we showed that the HIV protease inhibitor ritonavir is a more potent inhibitor of P-glycoprotein than the multidrug resistance (MDR)-reversing agent SDZ PSC 833. Ritonavir 57-66 ATP binding cassette subfamily B member 1 Homo sapiens 97-111 10082072-2 1999 All are inhibitors of CYP3A4, ranging from weak inhibition for saquinavir to very potent inhibition for ritonavir. Ritonavir 104-113 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 22-28 10228296-10 1999 CONCLUSIONS: Early increases in lymphocytes after ritonavir therapy are a result of recirculation, as shown by increases in B cells and CD4(+) CD45RO and CD8(+) T cells. Ritonavir 50-59 CD4 molecule Homo sapiens 136-139 10228296-10 1999 CONCLUSIONS: Early increases in lymphocytes after ritonavir therapy are a result of recirculation, as shown by increases in B cells and CD4(+) CD45RO and CD8(+) T cells. Ritonavir 50-59 CD8a molecule Homo sapiens 154-157 10203056-5 1999 Previous studies revealed that monotherapy with ritonavir, a protease inhibitor, resulted in a slight improvement in memory CD4+ T cell responses to recall Ags only when detectable prior to onset of therapy, suggesting that the loss of CD4+ T cell reactivity might be irreversible at advanced stages of the disease [2]. Ritonavir 48-57 CD4 molecule Homo sapiens 124-127 10203056-5 1999 Previous studies revealed that monotherapy with ritonavir, a protease inhibitor, resulted in a slight improvement in memory CD4+ T cell responses to recall Ags only when detectable prior to onset of therapy, suggesting that the loss of CD4+ T cell reactivity might be irreversible at advanced stages of the disease [2]. Ritonavir 48-57 CD4 molecule Homo sapiens 236-239 10219639-14 1999 Antiretroviral combinations including ritonavir were tolerated by six of eight children and produced substantial benefits with respect to increased numbers of CD4 cells and a decline in plasma viral RNA concentration. Ritonavir 38-47 CD4 molecule Homo sapiens 159-162 10219639-15 1999 It can be concluded that the administration of ritonavir is possible in a significant proportion of HIV-infected children, and leads to improvement of the CD4 cell count and viral load. Ritonavir 47-56 CD4 molecule Homo sapiens 155-158 10221532-1 1999 This study investigated the effects of a combination antiretroviral drug regimen (indinavir and two nucleoside analogs or ritonavir and saquinavir) on the levels of CD34+ colony-forming units (CFU-Cs) in the peripheral blood of HIV-1+ patients. Ritonavir 122-131 CD34 molecule Homo sapiens 165-169 9835508-1 1998 The human immunodeficiency virus type 1 (HIV-1) protease inhibitors (PIs)-saquinavir, ritonavir, nelfinavir, and indinavir-interact with the ABC-type multidrug transporter proteins MDR1 and MRP1 in CEM T-lymphocytic cell lines. Ritonavir 86-95 ATP binding cassette subfamily B member 1 Homo sapiens 181-185 10207548-15 1999 CONCLUSION: Despite variable viral response, antiretroviral-experienced HIV-infected children demonstrated a substantial CD4 cell increase during a median period of 15 months of ritonavir containing combination therapy. Ritonavir 178-187 CD4 molecule Homo sapiens 121-124 9876810-7 1998 Ritonavir has been shown to be a potent inhibitor of CYP3A4, an enzyme responsible for warfarin metabolism. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 53-59 9835508-1 1998 The human immunodeficiency virus type 1 (HIV-1) protease inhibitors (PIs)-saquinavir, ritonavir, nelfinavir, and indinavir-interact with the ABC-type multidrug transporter proteins MDR1 and MRP1 in CEM T-lymphocytic cell lines. Ritonavir 86-95 ATP binding cassette subfamily C member 1 Homo sapiens 190-194 9875393-3 1998 K-12 was active against different strains of HIV-1 (including AZT- and ritonavir-resistant HIV-1 strains), HIV-2 and simian immunodeficiency virus, in MT-4, CEM, C8166 and peripheral blood mononuclear cells. Ritonavir 71-80 keratin 12 Homo sapiens 0-4 9826719-3 1998 To gain experimental evidence supporting one of the evolutionary models, we investigated whether the development of resistance to the protease inhibitor ritonavir affected the evolution of the env gene. Ritonavir 153-162 endogenous retrovirus group K member 20 Homo sapiens 193-196 9826719-4 1998 Sequential serum samples from five patients treated with ritonavir were used for analysis of the protease gene and the V3 domain of the env gene. Ritonavir 57-66 endogenous retrovirus group K member 20 Homo sapiens 136-139 9812178-7 1998 Ritonavir is primarily metabolised by cytochrome P450 (CYP) 3A isozymes and, to a lesser extent, by CYP2D6. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 38-62 9812178-7 1998 Ritonavir is primarily metabolised by cytochrome P450 (CYP) 3A isozymes and, to a lesser extent, by CYP2D6. Ritonavir 0-9 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 100-106 9812178-13 1998 In vitro, ritonavir is a potent inhibitor of CYP3A. Ritonavir 10-19 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 45-50 9812178-14 1998 In vivo, ritonavir significantly increases the AUC of drugs primarily eliminated by CYP3A metabolism (e.g. clarithromycin, ketoconazole, rifabutin, and other HIV protease inhibitors, including indinavir, saquinavir and nelfinavir) with effects ranging from an increase of 77% to 20-fold in humans. Ritonavir 9-18 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 84-89 9812178-16 1998 Since ritonavir is also an inducer of several metabolising enzymes [CYP1A4, glucuronosyl transferase (GT), and possibly CYP2C9 and CYP2C19], the magnitude of drug interactions is difficult to predict, particularly for drugs that are metabolised by multiple enzymes or have low intrinsic clearance by CYP3A. Ritonavir 6-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 120-126 9812178-16 1998 Since ritonavir is also an inducer of several metabolising enzymes [CYP1A4, glucuronosyl transferase (GT), and possibly CYP2C9 and CYP2C19], the magnitude of drug interactions is difficult to predict, particularly for drugs that are metabolised by multiple enzymes or have low intrinsic clearance by CYP3A. Ritonavir 6-15 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 131-138 9812178-16 1998 Since ritonavir is also an inducer of several metabolising enzymes [CYP1A4, glucuronosyl transferase (GT), and possibly CYP2C9 and CYP2C19], the magnitude of drug interactions is difficult to predict, particularly for drugs that are metabolised by multiple enzymes or have low intrinsic clearance by CYP3A. Ritonavir 6-15 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 300-305 9812178-18 1998 Ritonavir is minimally affected by other CYP3A inhibitors, including ketoconazole. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 41-46 9812178-19 1998 Rifampicin (rifampin), a potent CYP3A inducer, decreased the AUC of ritonavir by only 35%. Ritonavir 68-77 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 32-37 9803961-6 1998 Saquinavir, ritonavir and nelfinavir significantly inhibited the efflux of [3H]paclitaxel and [3H]vinblastine in P-gp-positive cells, resulting in an increase in intracellular accumulation of these drugs. Ritonavir 12-21 ATP binding cassette subfamily B member 1 Homo sapiens 113-117 9803961-8 1998 In photoaffinity labeling studies, saquinavir and ritonavir displaced [3H]azidopine, a substrate for P-gp, in a dose-dependent manner. Ritonavir 50-59 ATP binding cassette subfamily B member 1 Homo sapiens 101-105 9803961-9 1998 These data suggest that saquinavir, ritonavir, and nelfinavir are inhibitors and possibly substrates of P-gp. Ritonavir 36-45 ATP binding cassette subfamily B member 1 Homo sapiens 104-108 9616191-3 1998 Ritonavir was similarly biotransformed by microsomes containing expressed CYP3A4 or CYP3A5 isozymes (KM = 0.05-0.07 microM, Vmax = 1-1.4 nmol/min/nmol CYP). Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 74-80 9616191-3 1998 Ritonavir was similarly biotransformed by microsomes containing expressed CYP3A4 or CYP3A5 isozymes (KM = 0.05-0.07 microM, Vmax = 1-1.4 nmol/min/nmol CYP). Ritonavir 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-77 9616191-3 1998 Ritonavir was similarly biotransformed by microsomes containing expressed CYP3A4 or CYP3A5 isozymes (KM = 0.05-0.07 microM, Vmax = 1-1.4 nmol/min/nmol CYP). Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 84-90 9616191-7 1998 The metabolism of ritonavir in liver and enterocyte microsomes was associated with inactivation of CYP3A. Ritonavir 18-27 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 99-104 9616191-10 1998 Metabolic inactivation of CYP3A by ritonavir explains the improved bioavailability and pharmacokinetics of ritonavir and the sustained elevation of blood levels of other, concomitantly administered, substrates of CYP3A. Ritonavir 35-44 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 26-31 9616191-10 1998 Metabolic inactivation of CYP3A by ritonavir explains the improved bioavailability and pharmacokinetics of ritonavir and the sustained elevation of blood levels of other, concomitantly administered, substrates of CYP3A. Ritonavir 35-44 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 213-218 9616191-10 1998 Metabolic inactivation of CYP3A by ritonavir explains the improved bioavailability and pharmacokinetics of ritonavir and the sustained elevation of blood levels of other, concomitantly administered, substrates of CYP3A. Ritonavir 107-116 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 26-31 9492772-3 1998 We undertook an international, multicentre, randomised, double-blind, placebo-controlled trial of ritonavir in patients with HIV-1 infection and CD4-lymphocyte counts of 100 cells/microL or less, who had previously been treated with antiretroviral drugs. Ritonavir 98-107 CD4 molecule Homo sapiens 145-148 9585795-8 1998 CONCLUSIONS: Ritonavir inhibited the metabolism of rifabutin and 25-O-desacetylrifabutin, suggesting that both are metabolized at least in part by CYP3A. Ritonavir 13-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 147-152 9533981-9 1998 Higher plasma drug concentrations may also be obtained by combining the drug with CYP blockers, such as ritonavir or ketoconazole. Ritonavir 104-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-85 9542203-1 1998 The virologic and immunologic efficacy of ritonavir, an HIV protease inhibitor, was examined in 14 HIV-infected patients with a mean CD4+ cell count of 183 x 10(6)/l at baseline. Ritonavir 42-51 CD4 molecule Homo sapiens 133-136 9486958-0 1997 HIV protease inhibitors, saquinavir, indinavir and ritonavir: inhibition of CYP3A4-mediated metabolism of testosterone and benzoxazinorifamycin, KRM-1648, in human liver microsomes. Ritonavir 51-60 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 76-82 9549640-4 1998 Ritonavir had high inhibition potency against cytochrome P450-2C9 (tolbutamide hydroxylation), -2C19 (S-mephenytoin hydroxylation), and -2D6 (dextromethorphan O-demethylation and desipramine hydroxylation), while the other protease inhibitors had one or more orders of magnitude lower inhibitory activity against these reactions. Ritonavir 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-65 9420308-3 1997 Compared with both RT inhibitors, treatment with the protease inhibitor ritonavir resulted in the most significant and persistent elevation of CD4+ and CD8+ T-cell counts. Ritonavir 72-81 CD4 molecule Homo sapiens 143-146 9420308-3 1997 Compared with both RT inhibitors, treatment with the protease inhibitor ritonavir resulted in the most significant and persistent elevation of CD4+ and CD8+ T-cell counts. Ritonavir 72-81 CD8a molecule Homo sapiens 152-155 9486958-1 1997 The protease inhibitors, ritonavir, indinavir and saquinavir, the most potent anti-HIV drugs developed to date, interact with many drugs by competing for CYP3A4, an enzyme central to the metabolism of a wide variety of compounds. Ritonavir 25-34 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 154-160 9056009-2 1997 In rat and human liver microsomes, ritonavir potently inhibited the cytochrome P450 (CYP)-mediated metabolism of saquinavir, indinavir, nelfinavir, and VX-478. Ritonavir 35-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 9278209-6 1997 RESULTS: Ritonavir was a very potent inhibitor of CYP3A4 mediated testosterone 6beta-hydroxylation (mean K(i) = 0.019 +/- 0.004 microM, mean +/- s.d. Ritonavir 9-18 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 50-56 9278209-1 1997 AIMS: To compare the inhibitory potential of the HIV protease inhibitors saquinavir, ritonavir and indinavir against CYP1A2, CYP2C9, CYP2E1 and CYP3A4 catalysed metabolic reactions in human liver microsomes in vitro. Ritonavir 85-94 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 117-123 9278209-1 1997 AIMS: To compare the inhibitory potential of the HIV protease inhibitors saquinavir, ritonavir and indinavir against CYP1A2, CYP2C9, CYP2E1 and CYP3A4 catalysed metabolic reactions in human liver microsomes in vitro. Ritonavir 85-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 125-131 9278209-1 1997 AIMS: To compare the inhibitory potential of the HIV protease inhibitors saquinavir, ritonavir and indinavir against CYP1A2, CYP2C9, CYP2E1 and CYP3A4 catalysed metabolic reactions in human liver microsomes in vitro. Ritonavir 85-94 cytochrome P450 family 2 subfamily E member 1 Homo sapiens 133-139 9278209-1 1997 AIMS: To compare the inhibitory potential of the HIV protease inhibitors saquinavir, ritonavir and indinavir against CYP1A2, CYP2C9, CYP2E1 and CYP3A4 catalysed metabolic reactions in human liver microsomes in vitro. Ritonavir 85-94 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 144-150 9250556-3 1997 The probabilities of one interaction or more were 31%, 42%, and 77% of patients if treated with indinavir, saquinavir, and ritonavir, respectively, across all CD4 groups; when the CD4 count was below 100 cells/microl, the probabilities were 55%, 63%, and 93%. Ritonavir 123-132 CD4 molecule Homo sapiens 159-162 9250556-3 1997 The probabilities of one interaction or more were 31%, 42%, and 77% of patients if treated with indinavir, saquinavir, and ritonavir, respectively, across all CD4 groups; when the CD4 count was below 100 cells/microl, the probabilities were 55%, 63%, and 93%. Ritonavir 123-132 CD4 molecule Homo sapiens 180-183 9250556-5 1997 The potential for drug interactions is high when starting protease inhibitor therapy, especially in patients with low CD4 cell counts who receive ritonavir. Ritonavir 146-155 CD4 molecule Homo sapiens 118-121 9084785-2 1997 Ritonavir (RIT) is a potent inhibitor of CYP3A4 and inhibits saquinavir (SQV) metabolism in healthy volunteers. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 41-47 9084785-2 1997 Ritonavir (RIT) is a potent inhibitor of CYP3A4 and inhibits saquinavir (SQV) metabolism in healthy volunteers. Ritonavir 11-14 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 41-47 9056009-2 1997 In rat and human liver microsomes, ritonavir potently inhibited the cytochrome P450 (CYP)-mediated metabolism of saquinavir, indinavir, nelfinavir, and VX-478. Ritonavir 35-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 9056009-3 1997 The structural features of ritonavir responsible for CYP binding and inhibition were examined. Ritonavir 27-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-56 8881129-5 1996 Clinical trials in HIV infection with saquinavir, indinavir, and ritonavir have demonstrated a decrease in viral load measured by plasma HIV-RNA PCR and an increase in CD4 lymphocyte counts. Ritonavir 65-74 CD4 molecule Homo sapiens 168-171 8948369-7 1996 In contrast, ritonavir monotherapy caused a peak increase of 892 CD8+ cells/mm3, which remained significantly above baseline for 32 weeks. Ritonavir 13-22 CD8a molecule Homo sapiens 65-68 8948369-9 1996 These findings suggest that the greater CD8+ response seen with ritonavir may be due to its specific inhibition of HIV protease and also that the CD8+ response is dependent on new CD4+ cell production. Ritonavir 64-73 CD8a molecule Homo sapiens 40-43 8948369-9 1996 These findings suggest that the greater CD8+ response seen with ritonavir may be due to its specific inhibition of HIV protease and also that the CD8+ response is dependent on new CD4+ cell production. Ritonavir 64-73 CD4 molecule Homo sapiens 180-183 8859229-2 1996 To investigate the mechanisms of its action, we studied changes in the serum levels of total IgE, allergen-specific IgE and IgG4, and expression of CD23 on peripheral blood B cells in patients receiving RIT. Ritonavir 203-206 Fc epsilon receptor II Homo sapiens 148-152 8859229-5 1996 Furthermore, after 6 months of RIT, the percentage of CD23+B cells and its CD23 receptor density significantly decreased. Ritonavir 31-34 Fc epsilon receptor II Homo sapiens 54-58 8859229-5 1996 Furthermore, after 6 months of RIT, the percentage of CD23+B cells and its CD23 receptor density significantly decreased. Ritonavir 31-34 Fc epsilon receptor II Homo sapiens 75-79 8891466-4 1996 In patients with HIV-1 infection, ritonavir markedly reduced viral load within 2 weeks of treatment onset and also increased CD4+ cell counts. Ritonavir 34-43 CD4 molecule Homo sapiens 125-128 8891466-7 1996 Early results suggest combination therapy with ritonavir and saquinavir increases CD4+ cell counts and decreases HIV RNA levels in patients with previously untreated HIV infection. Ritonavir 47-56 CD4 molecule Homo sapiens 82-85 8613951-5 1996 Ritonavir was found to be a potent inhibitor of CYP3A-mediated biotransformations (nifedipine oxidation, IC50) = 0.07 microM; 17alpha-ethynylestradiol 2-hydroxylation, IC50 = 2 microM; terfenadine hydroxylation, IC50 = 0.14 microM). Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 48-53 8613951-6 1996 Ritonavir was also found to be an inhibitor of the reactions mediated by CYP2D6 (IC50 = 2.5 microM) and CYP2C9/10 (IC50 = 8.0 microM). Ritonavir 0-9 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 73-79 8613951-6 1996 Ritonavir was also found to be an inhibitor of the reactions mediated by CYP2D6 (IC50 = 2.5 microM) and CYP2C9/10 (IC50 = 8.0 microM). Ritonavir 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 8613951-7 1996 The results of this study indicate the potential for in vivo inhibition of the metabolism by ritonavir of drugs that are CYP3A, CYP2D6 and, to a lesser extent, CYP2C9/10 substrates. Ritonavir 93-102 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 121-126 8613951-7 1996 The results of this study indicate the potential for in vivo inhibition of the metabolism by ritonavir of drugs that are CYP3A, CYP2D6 and, to a lesser extent, CYP2C9/10 substrates. Ritonavir 93-102 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 128-134 8613951-7 1996 The results of this study indicate the potential for in vivo inhibition of the metabolism by ritonavir of drugs that are CYP3A, CYP2D6 and, to a lesser extent, CYP2C9/10 substrates. Ritonavir 93-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 160-166 8143603-4 1993 RIT was found to increase background rates of chromosome-type aberrations and frequencies of hprt mutations and there was a strong correlation between levels of therapy-induced chromosome damage sustained in vivo and in vitro sensitivity to radiation-induced chromosome damage. Ritonavir 0-3 hypoxanthine phosphoribosyltransferase 1 Homo sapiens 93-97 7659390-8 1995 Therefore, determination of changes in the T-lymphocyte subsets and the CD4+:CD8+ ratio could be used as clinical and prognostic indicators in patients who have received RIT. Ritonavir 170-173 CD4 molecule Homo sapiens 72-75 7659390-8 1995 Therefore, determination of changes in the T-lymphocyte subsets and the CD4+:CD8+ ratio could be used as clinical and prognostic indicators in patients who have received RIT. Ritonavir 170-173 CD8a molecule Homo sapiens 77-80 11362207-1 1995 ABT-538, a new protease inhibitor, appears to increase CD4 counts and viral load in HIV-infected persons. Ritonavir 0-7 CD4 molecule Homo sapiens 55-58 7477167-7 1995 After 32 weeks, in the seven patients in the highest-dosage group (600 mg of ritonavir every 12 hours), the median increase from base line in the CD4+ lymphocyte count was 230 cells per cubic millimeter, and the mean decrease in the plasma concentration of HIV-1 RNA (as measured by a branched-DNA assay) was 0.81 log (95 percent confidence interval, 0.40 to 1.22). Ritonavir 77-86 CD4 molecule Homo sapiens 146-149 1727128-8 1992 In dogs, bone marrow transplantation with autologous cryopreserved bone marrow cells or G-CSF treatment can accelerate hemopoietic recovery and granulopoiesis, respectively, after RIT. Ritonavir 180-183 colony stimulating factor 3 Canis lupus familiaris 88-93 1944353-7 1991 Molecular analysis of mutants demonstrated a greater proportion of mutations with hprt gene changes on Southern blots after RIT treatment than before (40% versus 20%). Ritonavir 124-127 hypoxanthine phosphoribosyltransferase 1 Homo sapiens 82-86 33821626-4 2021 In order to determine if CYP3A7 inhibition could lead to the adverse outcomes associated with Kaletra therapy, we conducted in vitro metabolic studies to determine the extent and mechanism of CYP3A7 inhibition by both ritonavir and lopinavir and the relative intrinsic clearance of lopinavir with and without ritonavir in both neonatal and adult human liver microsomes (HLMs). Ritonavir 218-227 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 192-198 33821626-5 2021 We identified ritonavir as a potent inhibitor of CYP3A7 oxidation of DHEA-S (IC50 = 0.0514 muM), while lopinavir is a much weaker inhibitor (IC50 = 5.88 muM). Ritonavir 14-23 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 49-55 33821626-6 2021 Furthermore, ritonavir is a time-dependent inhibitor of CYP3A7 with a KI of 0.392 muM and a kinact of 0.119 min-1, illustrating the potential for CYP3A mediated drug-drug interactions with Kaletra. Ritonavir 13-22 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 56-62 33821626-6 2021 Furthermore, ritonavir is a time-dependent inhibitor of CYP3A7 with a KI of 0.392 muM and a kinact of 0.119 min-1, illustrating the potential for CYP3A mediated drug-drug interactions with Kaletra. Ritonavir 13-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 56-61 33821626-8 2021 Our results suggest that several of the observed adverse outcomes of Kaletra therapy may be due to the direct inhibition of CYP3A7 by ritonavir and that the necessity for the inclusion of this drug in the therapy may be obviated by the lower rate of lopinavir clearance in the neonatal liver. Ritonavir 134-143 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 124-130 34949673-2 2022 RTV is primarily metabolized by cytochrome P450 3A4 (CYP3A4) in the liver. Ritonavir 0-3 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 32-51 34952136-13 2022 Similar to rifampin, dual-inhibition functions of ritonavir affecting both CYP3A enzymes and enterohepatic Abcb1 transporters enhanced larotrectinib oral availability. Ritonavir 50-59 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 75-80 34952136-13 2022 Similar to rifampin, dual-inhibition functions of ritonavir affecting both CYP3A enzymes and enterohepatic Abcb1 transporters enhanced larotrectinib oral availability. Ritonavir 50-59 ATP-binding cassette, sub-family B (MDR/TAP), member 1B Mus musculus 107-112 34949673-2 2022 RTV is primarily metabolized by cytochrome P450 3A4 (CYP3A4) in the liver. Ritonavir 0-3 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 53-59 34949673-8 2022 HNF4A-AS1 knockdown increased PXR and CYP3A4 expression and exacerbated RTV-induced cytotoxicity, whereas HNF1A-AS1 knockdown generated the opposite phenotype. Ritonavir 72-75 HNF4A antisense RNA 1 Homo sapiens 0-9 34949673-11 2022 Alterations in RTV-induced hepatotoxicity caused by decreasing HNF4A-AS1 or HNF1A-AS1 were reversed by knockdown or overexpression of PXR. Ritonavir 15-18 HNF4A antisense RNA 1 Homo sapiens 63-72 34949673-11 2022 Alterations in RTV-induced hepatotoxicity caused by decreasing HNF4A-AS1 or HNF1A-AS1 were reversed by knockdown or overexpression of PXR. Ritonavir 15-18 HNF1A antisense RNA 1 Homo sapiens 76-85 34949673-11 2022 Alterations in RTV-induced hepatotoxicity caused by decreasing HNF4A-AS1 or HNF1A-AS1 were reversed by knockdown or overexpression of PXR. Ritonavir 15-18 nuclear receptor subfamily 1 group I member 2 Homo sapiens 134-137 34949673-12 2022 Increased susceptibility to RTV-induced liver injury caused by the PXR activator rifampicin was attenuated by HNF4A-AS1 overexpression or HNF1A-AS1 knockdown. Ritonavir 28-31 nuclear receptor subfamily 1 group I member 2 Homo sapiens 67-70 34949673-12 2022 Increased susceptibility to RTV-induced liver injury caused by the PXR activator rifampicin was attenuated by HNF4A-AS1 overexpression or HNF1A-AS1 knockdown. Ritonavir 28-31 HNF4A antisense RNA 1 Homo sapiens 110-119 34949673-12 2022 Increased susceptibility to RTV-induced liver injury caused by the PXR activator rifampicin was attenuated by HNF4A-AS1 overexpression or HNF1A-AS1 knockdown. Ritonavir 28-31 HNF1A antisense RNA 1 Homo sapiens 138-147 34949673-13 2022 Taken together, these results revealed that HNF4A-AS1 and HNF1A-AS1 modulated RTV-induced hepatotoxicity by regulating CYP3A4 expression, primarily by affecting the binding of PXR and histone modification status in the CYP3A4 promoter. Ritonavir 78-81 HNF4A antisense RNA 1 Homo sapiens 44-53 34949673-13 2022 Taken together, these results revealed that HNF4A-AS1 and HNF1A-AS1 modulated RTV-induced hepatotoxicity by regulating CYP3A4 expression, primarily by affecting the binding of PXR and histone modification status in the CYP3A4 promoter. Ritonavir 78-81 HNF1A antisense RNA 1 Homo sapiens 58-67 34949673-13 2022 Taken together, these results revealed that HNF4A-AS1 and HNF1A-AS1 modulated RTV-induced hepatotoxicity by regulating CYP3A4 expression, primarily by affecting the binding of PXR and histone modification status in the CYP3A4 promoter. Ritonavir 78-81 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 119-125 34949673-13 2022 Taken together, these results revealed that HNF4A-AS1 and HNF1A-AS1 modulated RTV-induced hepatotoxicity by regulating CYP3A4 expression, primarily by affecting the binding of PXR and histone modification status in the CYP3A4 promoter. Ritonavir 78-81 nuclear receptor subfamily 1 group I member 2 Homo sapiens 176-179 34949673-13 2022 Taken together, these results revealed that HNF4A-AS1 and HNF1A-AS1 modulated RTV-induced hepatotoxicity by regulating CYP3A4 expression, primarily by affecting the binding of PXR and histone modification status in the CYP3A4 promoter. Ritonavir 78-81 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 219-225 34949673-14 2022 Significance Statement HNF4A-AS1 and HNF1A-AS1, transcribed separately from neighboring antisense genes of the human transcription factor genes HNF4A and HNF1A, were identified as lncRNAs that can affect RTV-induced hepatotoxicity and susceptibility to RTV-induced hepatotoxicity caused by rifampicin exposure, mainly by affecting the expression of CY3A4 via alterations in PXR enrichment and histone modification status in the CYP3A4 promoter. Ritonavir 204-207 HNF4A antisense RNA 1 Homo sapiens 23-32 34949673-14 2022 Significance Statement HNF4A-AS1 and HNF1A-AS1, transcribed separately from neighboring antisense genes of the human transcription factor genes HNF4A and HNF1A, were identified as lncRNAs that can affect RTV-induced hepatotoxicity and susceptibility to RTV-induced hepatotoxicity caused by rifampicin exposure, mainly by affecting the expression of CY3A4 via alterations in PXR enrichment and histone modification status in the CYP3A4 promoter. Ritonavir 204-207 HNF1A antisense RNA 1 Homo sapiens 37-46 34949673-14 2022 Significance Statement HNF4A-AS1 and HNF1A-AS1, transcribed separately from neighboring antisense genes of the human transcription factor genes HNF4A and HNF1A, were identified as lncRNAs that can affect RTV-induced hepatotoxicity and susceptibility to RTV-induced hepatotoxicity caused by rifampicin exposure, mainly by affecting the expression of CY3A4 via alterations in PXR enrichment and histone modification status in the CYP3A4 promoter. Ritonavir 204-207 hepatocyte nuclear factor 4 alpha Homo sapiens 144-149 34949673-14 2022 Significance Statement HNF4A-AS1 and HNF1A-AS1, transcribed separately from neighboring antisense genes of the human transcription factor genes HNF4A and HNF1A, were identified as lncRNAs that can affect RTV-induced hepatotoxicity and susceptibility to RTV-induced hepatotoxicity caused by rifampicin exposure, mainly by affecting the expression of CY3A4 via alterations in PXR enrichment and histone modification status in the CYP3A4 promoter. Ritonavir 204-207 HNF1 homeobox A Homo sapiens 154-159 34949673-14 2022 Significance Statement HNF4A-AS1 and HNF1A-AS1, transcribed separately from neighboring antisense genes of the human transcription factor genes HNF4A and HNF1A, were identified as lncRNAs that can affect RTV-induced hepatotoxicity and susceptibility to RTV-induced hepatotoxicity caused by rifampicin exposure, mainly by affecting the expression of CY3A4 via alterations in PXR enrichment and histone modification status in the CYP3A4 promoter. Ritonavir 253-256 HNF4A antisense RNA 1 Homo sapiens 23-32 34949673-14 2022 Significance Statement HNF4A-AS1 and HNF1A-AS1, transcribed separately from neighboring antisense genes of the human transcription factor genes HNF4A and HNF1A, were identified as lncRNAs that can affect RTV-induced hepatotoxicity and susceptibility to RTV-induced hepatotoxicity caused by rifampicin exposure, mainly by affecting the expression of CY3A4 via alterations in PXR enrichment and histone modification status in the CYP3A4 promoter. Ritonavir 253-256 HNF1A antisense RNA 1 Homo sapiens 37-46 34949673-14 2022 Significance Statement HNF4A-AS1 and HNF1A-AS1, transcribed separately from neighboring antisense genes of the human transcription factor genes HNF4A and HNF1A, were identified as lncRNAs that can affect RTV-induced hepatotoxicity and susceptibility to RTV-induced hepatotoxicity caused by rifampicin exposure, mainly by affecting the expression of CY3A4 via alterations in PXR enrichment and histone modification status in the CYP3A4 promoter. Ritonavir 253-256 HNF1 homeobox A Homo sapiens 154-159 34175754-3 2021 Worldwide Conference on Harmonization approval rules were taken after to completely approve the strategy, and linearity was gotten for the two drugs over the extend of 0.4-2.4 microg mL-1 for Lopinavir and 0.1-0.6 microg mL-1 for ritonavir. Ritonavir 230-239 L1 cell adhesion molecule Mus musculus 221-225 34848452-2 2021 RTS-V5 is the first dual histone deacetylase-proteasome inhibitor, and we anticipated that combining it with the cytochrome P450 family 3 subfamily A member 4 inhibitor ritonavir would enhance its activity in bladder cancer cells. Ritonavir 169-178 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 113-158 34885967-5 2021 In this work, the interaction mechanism of four HIV protease inhibitors Darunavir (DRV), Lopinavir (LPV), Nelfinavir (NFV), and Ritonavire (RTV) targeting SARS-CoV-2 Mpro was explored by applying docking, molecular dynamics (MD) simulations, and MM-GBSA methods using the broad-spectrum antiviral drug Ribavirin (RBV) as the negative and nonspecific control. Ritonavir 140-143 NEWENTRY Severe acute respiratory syndrome-related coronavirus 166-170 34397829-13 2021 CONCLUSION: Limited evidence suggests that lopinavir, ritonavir, atazanavir, and saquinavir could cause PR prolongation, QRS widening, and QT prolongation. Ritonavir 54-63 transmembrane protein 37 Homo sapiens 104-106 34741481-7 2022 Our in silico analysis revealed that the three drugs Lopinavir, Ritonavir, and Remdesivir showed interaction with the active site residues of Mpro. Ritonavir 64-73 NEWENTRY Severe acute respiratory syndrome-related coronavirus 142-146 34408078-0 2021 The HIV protease inhibitor, ritonavir, corrects diverse brain phenotypes across development in mouse model of DYT-TOR1A dystonia. Ritonavir 28-37 torsin family 1, member A (torsin A) Mus musculus 110-119 34408078-5 2021 Using a DYT1 knock-in mouse model to test efficacy on brain pathologies, we found that ritonavir restored multiple brain abnormalities across development. Ritonavir 87-96 torsin family 1, member A (torsin A) Mus musculus 8-12 34426442-1 2021 PURPOSE: To evaluate drug-drug interactions between the HER2-targeted antibody-drug conjugate trastuzumab deruxtecan (T-DXd; DS-8201a) and the OATP1B/CYP3A inhibitor ritonavir or the strong CYP3A inhibitor itraconazole. Ritonavir 166-175 erb-b2 receptor tyrosine kinase 2 Homo sapiens 56-60 34426442-1 2021 PURPOSE: To evaluate drug-drug interactions between the HER2-targeted antibody-drug conjugate trastuzumab deruxtecan (T-DXd; DS-8201a) and the OATP1B/CYP3A inhibitor ritonavir or the strong CYP3A inhibitor itraconazole. Ritonavir 166-175 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 150-155 34497279-5 2021 Lopinavir and ritonavir in low micromolar concentrations inhibited BSEP and MATE1 exporters, as well as OATP1B1/1B3 uptake transporters. Ritonavir 14-23 ATP binding cassette subfamily B member 11 Homo sapiens 67-71 34497279-5 2021 Lopinavir and ritonavir in low micromolar concentrations inhibited BSEP and MATE1 exporters, as well as OATP1B1/1B3 uptake transporters. Ritonavir 14-23 solute carrier family 47 member 1 Homo sapiens 76-81 34497279-5 2021 Lopinavir and ritonavir in low micromolar concentrations inhibited BSEP and MATE1 exporters, as well as OATP1B1/1B3 uptake transporters. Ritonavir 14-23 solute carrier organic anion transporter family member 1B1 Homo sapiens 104-115 34497279-6 2021 Ritonavir had a similar inhibitory pattern, also inhibiting OCT1. Ritonavir 0-9 solute carrier family 22 member 1 Homo sapiens 60-64 34392751-4 2021 From our results, three complexes containing the ligands with Pubchem IDs: 153012995, 12149203, and 123608715 showed lower binding energies than the control (Ritonavir), indicating that they may become promising inhibitors for PLpro. Ritonavir 158-167 ORF1a polyprotein;ORF1ab polyprotein Severe acute respiratory syndrome coronavirus 2 227-232 34139548-7 2021 Heterologous expression of these candidates and activity testing led to the identification of a novel P450 that efficiently converts ritonavir resembling the activity of the human CYP3A4. Ritonavir 133-142 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 180-186 34184163-7 2021 Lopinavir (LPV) and ritonavir (RTV), available as FDA-approved fixed-dose combination products, were chosen as model ART drugs, and the formulation and processing parameters of spray-dried cyclodextrin (CD)-based LPV and RTV complexes were studied. Ritonavir 221-224 cyclodextrin None 189-201 34197501-1 2021 BACKGROUND: Some mutations in the HIV-1 Gag gene are known to confer resistance to ritonavir-boosted protease inhibitors (PI/r), but their clinical implications remain controversial. Ritonavir 83-92 Pr55(Gag) Human immunodeficiency virus 1 40-43 34130375-9 2021 The best hit compounds on the human ACE2 were the lopinavir (-10.1 kcal/mol), ritonavir (-8.9 kcal/mol), and nafamostat (-8.7 kcal/mol). Ritonavir 78-87 angiotensin converting enzyme 2 Homo sapiens 36-40 34130375-3 2021 Thus, in this current computational study we carried out molecular docking experiments to assess the bridging potentials of some commercial drugs such as chloroquine, hydroxychloroquine, lopinavir, ritonavir, nafamostat, camostat, famotidine, umifenovir, nitazoxanide, ivermectin, and fluvoxamine at the interface between human ACE2 and the coronavirus spike glycoprotein complex. Ritonavir 198-207 angiotensin converting enzyme 2 Homo sapiens 328-332 35567754-3 2022 The ritonavir component boosts plasma concentrations of nirmatrelvir through the potent and rapid inhibition of the key drug metabolizing enzyme cytochrome P450 (CYP) 3A4. Ritonavir 4-13 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 145-170 35584053-1 2022 The derivative of protease inhibitor ritonavir (5-methyl-4-oxohexanoic acid ritonavir ester; RD) was recently recognized as a potent P-gp inhibitor and cancerostatic drug inhibiting the proteasome and STAT3 signaling. Ritonavir 37-46 phosphoglycolate phosphatase Mus musculus 133-137 35584053-1 2022 The derivative of protease inhibitor ritonavir (5-methyl-4-oxohexanoic acid ritonavir ester; RD) was recently recognized as a potent P-gp inhibitor and cancerostatic drug inhibiting the proteasome and STAT3 signaling. Ritonavir 37-46 signal transducer and activator of transcription 3 Mus musculus 201-206 35628472-7 2022 Favipiravir, hydroxychloroquine, lopinavir, and the combination of lopinavir with ritonavir were found to inhibit pannexin1 channel activity without affecting pannexin1 protein or mRNA levels. Ritonavir 82-91 pannexin 1 Homo sapiens 114-123 35543797-0 2022 Interactions between HIV protease inhibitor ritonavir and human DNA repair enzyme ALKBH2: a molecular dynamics simulation study. Ritonavir 44-53 alkB homolog 2, alpha-ketoglutarate dependent dioxygenase Homo sapiens 82-88 35543797-4 2022 Intending to repurpose a drug as an inhibitor of ALKBH2, we performed in silico evaluation of HIV protease inhibitors and identified Ritonavir as an ALKBH2-interacting molecule. Ritonavir 133-142 alkB homolog 2, alpha-ketoglutarate dependent dioxygenase Homo sapiens 49-55 35543797-4 2022 Intending to repurpose a drug as an inhibitor of ALKBH2, we performed in silico evaluation of HIV protease inhibitors and identified Ritonavir as an ALKBH2-interacting molecule. Ritonavir 133-142 alkB homolog 2, alpha-ketoglutarate dependent dioxygenase Homo sapiens 149-155 35543797-5 2022 Using molecular dynamics simulation, we elucidated the molecular details of Ritonavir-ALKBH2 interaction. Ritonavir 76-85 alkB homolog 2, alpha-ketoglutarate dependent dioxygenase Homo sapiens 86-92 35524830-3 2022 The expression profiles of miR-223-3p, miR-17-5p, miR-24-3p, and TLR2 - 196 to - 174 del/ins polymorphisms from the blood/serum of 34 hepatitis C virus (HCV)-infected patients pre- and post-ombitavir/paritaprevir/ritonavir + dasabuvir treatment were determined by RT-qPCR. Ritonavir 213-222 toll like receptor 2 Homo sapiens 65-69 35543797-6 2022 The present work highlights that Ritonavir might be used to target the ALKBH2-mediated DNA alkylation repair. Ritonavir 33-42 alkB homolog 2, alpha-ketoglutarate dependent dioxygenase Homo sapiens 71-77 35153195-10 2022 First-in-human pharmacokinetics studies have demonstrated a considerable boost in the oral systemic exposure of nirmatrelvir upon co-administration with the CYP3A4 inhibitor ritonavir, consistent with the predominant role of CYP3A4 in nirmatrelvir metabolism. Ritonavir 174-183 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 157-163 35153195-10 2022 First-in-human pharmacokinetics studies have demonstrated a considerable boost in the oral systemic exposure of nirmatrelvir upon co-administration with the CYP3A4 inhibitor ritonavir, consistent with the predominant role of CYP3A4 in nirmatrelvir metabolism. Ritonavir 174-183 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 225-231 35486324-15 2022 CONCLUSIONS: The current model, which incorporates formulation characteristics and mechanistic disposition parameters, can be used to assess the DDI potential of CYP3A4/5 and CYP2D6 substrates administered with a twice-daily dose of 100 mg of ritonavir for 14 days. Ritonavir 243-252 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 162-170 35486324-0 2022 A Mechanistic Absorption and Disposition Model of Ritonavir to Predict Exposure and Drug-Drug Interaction Potential of CYP3A4/5 and CYP2D6 Substrates. Ritonavir 50-59 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 119-127 35486324-15 2022 CONCLUSIONS: The current model, which incorporates formulation characteristics and mechanistic disposition parameters, can be used to assess the DDI potential of CYP3A4/5 and CYP2D6 substrates administered with a twice-daily dose of 100 mg of ritonavir for 14 days. Ritonavir 243-252 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 175-181 35486324-0 2022 A Mechanistic Absorption and Disposition Model of Ritonavir to Predict Exposure and Drug-Drug Interaction Potential of CYP3A4/5 and CYP2D6 Substrates. Ritonavir 50-59 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 132-138 35486324-2 2022 Ritonavir is a strong inhibitor for CYP3A4/5-mediated DDIs and has been proposed as a suitable alternative to ketoconazole. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 36-44 35486324-5 2022 Thus, the main purpose of the current study was to verify the predictive performance of a mechanistic absorption and disposition model of ritonavir when it was applied to the inhibition of CYP2D6 and CYP3A4/5 by ritonavir. Ritonavir 138-147 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 189-195 35486324-5 2022 Thus, the main purpose of the current study was to verify the predictive performance of a mechanistic absorption and disposition model of ritonavir when it was applied to the inhibition of CYP2D6 and CYP3A4/5 by ritonavir. Ritonavir 138-147 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 200-208 35486324-5 2022 Thus, the main purpose of the current study was to verify the predictive performance of a mechanistic absorption and disposition model of ritonavir when it was applied to the inhibition of CYP2D6 and CYP3A4/5 by ritonavir. Ritonavir 212-221 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 189-195 35486324-5 2022 Thus, the main purpose of the current study was to verify the predictive performance of a mechanistic absorption and disposition model of ritonavir when it was applied to the inhibition of CYP2D6 and CYP3A4/5 by ritonavir. Ritonavir 212-221 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 200-208 35486324-9 2022 RESULTS: Maximal inhibition of hepatic (3.53% of the activity remaining) and gut (5.16% of the activity remaining) CYP3A4 activity was observed when ritonavir was orally administered in doses of 100 mg or higher. Ritonavir 149-158 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 115-121 35429391-5 2022 RESULTS: Sixteen evaluable patients received 64 cycles of PCb, including 6 patients treated with CYP3A4 inhibiting ART (ritonavir). Ritonavir 120-129 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 97-103 35490356-15 2022 There was significantly lower mRNA gene expression of ASK-1 in the heart tissues of the treated rats with zidovudine (2.67 +- 0.09, p < 0.0001), ritonavir (2.57 +-0.11, p < 0.0001) and a combination of both (2.75 +- 0.06, p < 0.0001) when compared to rats in the untreated group. Ritonavir 145-154 mitogen-activated protein kinase kinase kinase 5 Rattus norvegicus 54-59 35461811-2 2022 Nirmatrelvir (PF-07321332) is a reversible, covalent inhibitor targeting the main protease (Mpro) of SARS-CoV-2 and the active protease inhibitor in PAXLOVID (nirmatrelvir tablets and ritonavir tablets). Ritonavir 185-194 NEWENTRY Severe acute respiratory syndrome-related coronavirus 92-96 35618549-1 2022 OBJECTIVES: Nirmatrelvir in association with ritonavir (PAXLOVID , Pfizer) is an antiviral agent targeting the 3-chymotrypsin-like cysteine protease enzyme (3C-like protease or Mpro) which is a key enzyme of the viral cycle of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Ritonavir 45-54 NEWENTRY Severe acute respiratory syndrome-related coronavirus 177-181 35302484-2 2022 In the drug combination nirmatrelvir + ritonavir (nirmatrelvir/r), the active agent, nirmatrelvir, is made bioavailable in clinically adequate amounts by the additional administration of a potent inhibitor of its first-pass metabolism by way of cytochrome P450 (CYP) 3A in the gut and liver. Ritonavir 39-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 245-269 35138967-1 2022 PURPOSE: Direct oral anticoagulants (DOACs) pose a challenge when given with potent CYP3A4 and P-gp inhibitors, such as the commonly prescribed pharmacokinetic booster ritonavir. Ritonavir 168-177 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 84-90 35305258-2 2022 Nirmatrelvir is a peptidomimetic inhibitor of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) main protease, while ritonavir is a human immunodeficiency virus type 1 (HIV-1) protease inhibitor and CYP3A inhibitor. Ritonavir 132-141 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 214-219 35107380-14 2022 Two hit drugs (azithromycin and ritonavir) strongly inhibited HEV production in culture supernatants, as well as intracellular expression of ORF2 protein, and may therefore be candidate novel anti-HEV drugs. Ritonavir 32-41 capsid protein Orthohepevirus A 141-145 35347032-1 2022 BACKGROUND/AIM: The antiviral agent ritonavir is a substrate for cytochrome P450 3A4 (CYP3A4); therefore, concomitant use of CYP3A4-metabolising drugs might cause adverse reactions to this drug. Ritonavir 36-45 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 65-84 35347032-1 2022 BACKGROUND/AIM: The antiviral agent ritonavir is a substrate for cytochrome P450 3A4 (CYP3A4); therefore, concomitant use of CYP3A4-metabolising drugs might cause adverse reactions to this drug. Ritonavir 36-45 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 86-92 35347032-1 2022 BACKGROUND/AIM: The antiviral agent ritonavir is a substrate for cytochrome P450 3A4 (CYP3A4); therefore, concomitant use of CYP3A4-metabolising drugs might cause adverse reactions to this drug. Ritonavir 36-45 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 125-131 35352280-3 2022 Boosting pharmacokinetic exposure of KIs metabolized by cytochrome P450 (CYP)3A4 with ritonavir might result in lower doses needed and subsequently reduces treatment costs. Ritonavir 86-95 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 56-80 35352280-12 2022 Ritonavir-boosting is a promising strategy to reduce erlotinib treatment costs and provides a rationale for other expensive therapies metabolized by CYP3A4. Ritonavir 0-9 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 149-155 35138967-1 2022 PURPOSE: Direct oral anticoagulants (DOACs) pose a challenge when given with potent CYP3A4 and P-gp inhibitors, such as the commonly prescribed pharmacokinetic booster ritonavir. Ritonavir 168-177 phosphoglycolate phosphatase Homo sapiens 95-99 35082353-1 2022 Response to ritonavir-boosted-protease inhibitors (PI/r)-based regimen is associated with some Gag mutations among HIV-1 B-clade. Ritonavir 12-21 Pr55(Gag) Human immunodeficiency virus 1 95-98 35186496-7 2022 Results indicated that birinapant, atazanavir, and ritonavir potently bound and stabilized SARS-CoV-2 Mpro structure. Ritonavir 51-60 NEWENTRY Severe acute respiratory syndrome-related coronavirus 102-106 33900318-6 2021 Ritonavir, Arbidol, and Chloroquine consistently showed an outstanding binding ability to monomeric Mpro under various methods. Ritonavir 0-9 NEWENTRY Severe acute respiratory syndrome-related coronavirus 100-104 33649517-2 2021 In this study, we aimed to assess the functional significance of 14 polymorphisms in the CYP3A, CYP1B1, ABCB1, ABCC2, and SLCO1B3 genes for the pharmacokinetics and pharmacodynamics of oral docetaxel, co-administered with ritonavir. Ritonavir 222-231 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 89-94 33649517-2 2021 In this study, we aimed to assess the functional significance of 14 polymorphisms in the CYP3A, CYP1B1, ABCB1, ABCC2, and SLCO1B3 genes for the pharmacokinetics and pharmacodynamics of oral docetaxel, co-administered with ritonavir. Ritonavir 222-231 cytochrome P450 family 1 subfamily B member 1 Homo sapiens 96-102 33649517-2 2021 In this study, we aimed to assess the functional significance of 14 polymorphisms in the CYP3A, CYP1B1, ABCB1, ABCC2, and SLCO1B3 genes for the pharmacokinetics and pharmacodynamics of oral docetaxel, co-administered with ritonavir. Ritonavir 222-231 ATP binding cassette subfamily B member 1 Homo sapiens 104-109 33649517-2 2021 In this study, we aimed to assess the functional significance of 14 polymorphisms in the CYP3A, CYP1B1, ABCB1, ABCC2, and SLCO1B3 genes for the pharmacokinetics and pharmacodynamics of oral docetaxel, co-administered with ritonavir. Ritonavir 222-231 ATP binding cassette subfamily C member 2 Homo sapiens 111-116 33649517-2 2021 In this study, we aimed to assess the functional significance of 14 polymorphisms in the CYP3A, CYP1B1, ABCB1, ABCC2, and SLCO1B3 genes for the pharmacokinetics and pharmacodynamics of oral docetaxel, co-administered with ritonavir. Ritonavir 222-231 solute carrier organic anion transporter family member 1B3 Homo sapiens 122-129 33629924-7 2021 Eplerenone is metabolized by the hepatic P450 cytochrome isoenzyme CYP3A4; therefore, concomitant administration with CYP3A4 inhibitors, like ritonavir, may increase plasma levels of eplerenone and, therefore, the risk of side effects, mainly hyperkalaemia. Ritonavir 142-151 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 67-73 33629924-7 2021 Eplerenone is metabolized by the hepatic P450 cytochrome isoenzyme CYP3A4; therefore, concomitant administration with CYP3A4 inhibitors, like ritonavir, may increase plasma levels of eplerenone and, therefore, the risk of side effects, mainly hyperkalaemia. Ritonavir 142-151 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 118-124 34006500-9 2021 The strong CYP3A4 inhibitor ritonavir, increased exposure to ivacaftor 7 times. Ritonavir 28-37 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 11-17 33900318-7 2021 Ritonavir, Arbidol, and Saquinavir presented the best performance when binding to a dimer, which was independent of the protonated state of Hie41 (protonated at Nepsilon) and Hid41 (protonated at Ndelta), and these findings suggest that Chloroquine may not effectively inhibit the activity of dimeric Mpro in vivo. Ritonavir 0-9 NEWENTRY Severe acute respiratory syndrome-related coronavirus 301-305 33900318-8 2021 Furthermore, three common hot-spot residues of Met165, Hie41, and Gln189 of monomeric Mpro systems dominated the binding of Ritonavir, Arbidol, and Chloroquine. Ritonavir 124-133 NEWENTRY Severe acute respiratory syndrome-related coronavirus 86-90 33900318-9 2021 In dimeric Mpro, Gln189, Met165, and Met49 contributed significantly to binding with Ritonavir, Arbidol, and Saquinavir; therefore, Gln189 and Met165 might serve as the focus in the discovery and development of anti-COVID-19 drugs. Ritonavir 85-94 NEWENTRY Severe acute respiratory syndrome-related coronavirus 11-15 33797130-10 2021 Overall, the present analysis yielded two potential inhibitors (DB02986 and DB08573) that are predicted to bind with the main protease of Covid-19 better than currently used drug molecules such as N3 (co-crystallized native ligand), Lopinavir and Ritonavir. Ritonavir 247-256 n3 None 197-199 34017865-6 2021 Methods: Atazanavir, remdesivir, ritonavir, lopinavir and favipiravir were docked to in silico models of the pore domain of hERG, derived from cryo-EM structures of hERG and the closely related EAG channel. Ritonavir 33-42 ETS transcription factor ERG Homo sapiens 124-128 33709626-1 2021 BACKGROUND: ModraDoc006 is an oral formulation of docetaxel, which is co-administered with the cytochrome P450 3A4 and P-glycoprotein inhibitor ritonavir (r): ModraDoc006/r. Ritonavir 144-153 ATP binding cassette subfamily B member 1 Homo sapiens 119-133 33759544-1 2021 Lopinavir and ritonavir are substrates of permeability glycoprotein encoded by ABCB1. Ritonavir 14-23 ATP binding cassette subfamily B member 1 Homo sapiens 79-84 33450312-7 2021 RTV (2.5 - 31.5 muM) dose-dependently inhibited SERCA1-mediated, ATP-dependent Ca2+ transport. Ritonavir 0-3 ATPase sarcoplasmic/endoplasmic reticulum Ca2+ transporting 1 Homo sapiens 48-54 33421743-5 2021 The results suggested that by using the combination of Lopinavir, Ritonavir along with Hydroxychloroquine and Vitamin C may turned out to be the effective line of treatment for SARS-CoV-2 as it shows the involvement of PARP-1, MAPK-8, EGFR, PRKCB, PTGS-2, and BCL-2. Ritonavir 66-75 prostaglandin-endoperoxide synthase 2 Homo sapiens 248-254 33421743-5 2021 The results suggested that by using the combination of Lopinavir, Ritonavir along with Hydroxychloroquine and Vitamin C may turned out to be the effective line of treatment for SARS-CoV-2 as it shows the involvement of PARP-1, MAPK-8, EGFR, PRKCB, PTGS-2, and BCL-2. Ritonavir 66-75 poly(ADP-ribose) polymerase 1 Homo sapiens 219-225 33421743-5 2021 The results suggested that by using the combination of Lopinavir, Ritonavir along with Hydroxychloroquine and Vitamin C may turned out to be the effective line of treatment for SARS-CoV-2 as it shows the involvement of PARP-1, MAPK-8, EGFR, PRKCB, PTGS-2, and BCL-2. Ritonavir 66-75 mitogen-activated protein kinase 8 Homo sapiens 227-233 33421743-5 2021 The results suggested that by using the combination of Lopinavir, Ritonavir along with Hydroxychloroquine and Vitamin C may turned out to be the effective line of treatment for SARS-CoV-2 as it shows the involvement of PARP-1, MAPK-8, EGFR, PRKCB, PTGS-2, and BCL-2. Ritonavir 66-75 BCL2 apoptosis regulator Homo sapiens 260-265 33421743-5 2021 The results suggested that by using the combination of Lopinavir, Ritonavir along with Hydroxychloroquine and Vitamin C may turned out to be the effective line of treatment for SARS-CoV-2 as it shows the involvement of PARP-1, MAPK-8, EGFR, PRKCB, PTGS-2, and BCL-2. Ritonavir 66-75 epidermal growth factor receptor Homo sapiens 235-239 33421743-5 2021 The results suggested that by using the combination of Lopinavir, Ritonavir along with Hydroxychloroquine and Vitamin C may turned out to be the effective line of treatment for SARS-CoV-2 as it shows the involvement of PARP-1, MAPK-8, EGFR, PRKCB, PTGS-2, and BCL-2. Ritonavir 66-75 protein kinase C beta Homo sapiens 241-246 33645457-5 2021 Current study, based on docking and extensive set of MD simulations, finds the combination of Elbasvir, Glecaprevir and Ritonavir to be a viable candidate for further experimental drug testing/pharmacophore design for Mpro.Communicated by Ramaswamy H. Sarma. Ritonavir 120-129 NEWENTRY Severe acute respiratory syndrome-related coronavirus 218-222 33464545-1 2021 INTRODUCTION: ModraDoc006 is a novel docetaxel tablet formulation that is co-administrated with the cytochrome P450 3A4 and P-glycoprotein inhibitor ritonavir (r): ModraDoc006/r. Ritonavir 149-158 ATP binding cassette subfamily B member 1 Homo sapiens 124-138 33679150-5 2021 In this study, we docked five clinically used drug molecules, favipiravir, hydroxychloroquine, remdesivir, lopinavir, and ritonavir onto three target proteins, the receptor-binding domain of SARS-CoV-2 spike protein, SARS-CoV-2 main protease, and human furin protease. Ritonavir 122-131 surface glycoprotein Severe acute respiratory syndrome coronavirus 2 202-207