Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
CPT Pharmacometrics Syst Pharmacol ; 13(6): 954-969, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38528813

ABSTRACT

Lenvatinib is a receptor tyrosine kinase (RTK) inhibitor targeting vascular endothelial growth factor (VEGF) receptors 1-3, fibroblast growth factor (FGF) receptors 1-4, platelet-derived growth factor receptor-α (PDGFRα), KIT, and RET that have been implicated in pathogenic angiogenesis, tumor growth, and cancer. The primary objective of this work was to evaluate, by establishing quantitative relationships, whether lenvatinib exposure and longitudinal serum biomarker data (VEGF, Ang-2, Tie-2, and FGF-23) are predictors for change in longitudinal tumor size which was assessed based on data from 558 patients with radioiodine-refractory differentiated thyroid cancer (RR-DTC) receiving either lenvatinib or placebo treatment. Lenvatinib PK was best described by a 3-compartment model with simultaneous first- and zero-order absorption and linear elimination from the central compartment with significant covariates (body weight, albumin <30 g/dL, ALP>ULN, RR-DTC, RCC, HCC subjects, and concomitant CYP3A inhibitors). Except for body weight, none of the covariates have any clinically meaningful effect on exposure to lenvatinib. Longitudinal biomarker measurements over time were reasonably well defined by a PK/PD model with common EC50, Emax, and a slope for disease progression for all biomarkers. Longitudinal tumor measurements over time were reasonably well defined by a tumor growth inhibition Emax model, which in addition to lenvatinib exposure, included model-predicted relative changes from baseline over time for Tie-2 and Ang-2 as having significant association with tumor response. The developed PK/PD models pave the way for dose optimization and potential prediction of clinical response.


Subject(s)
Iodine Radioisotopes , Phenylurea Compounds , Quinolines , Thyroid Neoplasms , Humans , Quinolines/pharmacokinetics , Quinolines/administration & dosage , Quinolines/therapeutic use , Quinolines/blood , Quinolines/pharmacology , Phenylurea Compounds/pharmacokinetics , Phenylurea Compounds/administration & dosage , Phenylurea Compounds/therapeutic use , Phenylurea Compounds/blood , Phenylurea Compounds/pharmacology , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/blood , Thyroid Neoplasms/pathology , Male , Female , Middle Aged , Adult , Iodine Radioisotopes/pharmacokinetics , Iodine Radioisotopes/therapeutic use , Aged , Biomarkers, Tumor/blood , Models, Biological , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/blood , Vascular Endothelial Growth Factor A/blood , Receptor, TIE-2/blood , Young Adult , Angiopoietin-2/blood
2.
Biochem Biophys Res Commun ; 513(1): 1-7, 2019 05 21.
Article in English | MEDLINE | ID: mdl-30944079

ABSTRACT

Lenvatinib inhibits VEGF- and FGF-driven angiogenesis, and proliferation of tumor cells with activated FGF signaling pathways in preclinical models, and we previously demonstrated antitumor activity in human HCC xenograft tumor models. Here, we examined the inhibitory activity of lenvatinib against FGF-driven survival of human HCC cell lines. First, we conducted a histological analysis of FGF19-overexpressing Hep3B2.1-7 xenograft tumors collected from mice treated with lenvatinib. Second, we examined the effects of pharmacological inhibition on survival of cultured HCC cells with an activated FGF signaling pathway under nutrient-starved culture condition to mimic tumor microenvironments induced by angiogenesis inhibition. In the first analysis, area of histological focal necrosis was greater in Hep3B2.1-7 xenograft tumors with the lenvatinib treatment than that after the treatment with sorafenib, which does not inhibit FGFRs. Lenvatinib and E7090 (a selective FGFR1-3 inhibitor), but not sorafenib, induced death of Hep3B2.1-7, and another FGF19 overexpressing HuH-7 cells. Lenvatinib and E7090 decreased phosphorylation of downstream molecules of the FGF signaling pathway (such as FRS2, Erk, and p38 MAPK), and induced PARP cleavage, even under limited nutrients. PD0325901, MEK inhibitor, caused the same changes in HCC cells as those described above for lenvatinib and E7090. These results reveal that the FGF signaling pathway through MAPK cascades plays an important role in survival of HCC cell lines with an activated FGF signaling pathway under limited nutrients, and FGFR-MAPK cascades likely contribute to survival of HCC cells with an activated FGF signaling pathway under tumor microenvironments with limited nutrients, where tumor angiogenesis is inhibited.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Fibroblast Growth Factors/metabolism , Liver Neoplasms/drug therapy , Phenylurea Compounds/therapeutic use , Quinolines/therapeutic use , Signal Transduction/drug effects , Animals , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Female , Humans , Liver Neoplasms/metabolism , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred BALB C , Mice, Nude , Receptors, Fibroblast Growth Factor/metabolism
3.
Cancer Med ; 7(6): 2641-2653, 2018 06.
Article in English | MEDLINE | ID: mdl-29733511

ABSTRACT

Unresectable hepatocellular carcinoma (uHCC) is one of the most lethal and prevalent cancers worldwide, and current systemic therapeutic options for uHCC are limited. Lenvatinib, a multiple receptor tyrosine kinase inhibitor targeting vascular endothelial growth factor receptors (VEGFRs) and fibroblast growth factor receptors (FGFRs), recently demonstrated a treatment effect on overall survival by statistical confirmation of noninferiority to sorafenib in a phase 3 study of uHCC. Here, we investigated mechanisms underlying the antitumor activity of lenvatinib in preclinical HCC models. In vitro proliferation assay of nine human HCC cell lines showed that lenvatinib selectively inhibited proliferation of FGF signal-activated HCC cells including FGF19-expressing Hep3B2.1-7. Lenvatinib suppressed phosphorylation of FRS2, a substrate of FGFR1-4, in these cells in a concentration-dependent manner. Lenvatinib inhibited in vivo tumor growth in Hep3B2.1-7 and SNU-398 xenografts and decreased phosphorylation of FRS2 and Erk1/2 within the tumor tissues. Lenvatinib also exerted antitumor activity and potently reduced tumor microvessel density in PLC/PRF/5 xenograft model and two HCC patient-derived xenograft models. These results suggest that lenvatinib has antitumor activity consistently across diverse HCC models, and that targeting of tumor FGF signaling pathways and anti-angiogenic activity underlies its antitumor activity against HCC tumors.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Fibroblast Growth Factors/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Neovascularization, Pathologic/metabolism , Phenylurea Compounds/pharmacology , Quinolines/pharmacology , Signal Transduction/drug effects , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Female , Humans , Immunohistochemistry , Liver Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
4.
Cancer Sci ; 108(4): 763-771, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28107584

ABSTRACT

The combination of lenvatinib, a multiple receptor tyrosine kinase inhibitor, plus everolimus, a mammalian target of rapamycin (mTOR) inhibitor, significantly improved clinical outcomes versus everolimus monotherapy in a phase II clinical study of metastatic renal cell carcinoma (RCC). We investigated potential mechanisms underlying the antitumor activity of the combination treatment in preclinical RCC models. Lenvatinib plus everolimus showed greater antitumor activity than either monotherapy in three human RCC xenograft mouse models (A-498, Caki-1, and Caki-2). In particular, the combination led to tumor regression in the A-498 and Caki-1 models. In the A-498 model, everolimus showed antiproliferative activity, whereas lenvatinib showed anti-angiogenic effects. The anti-angiogenic activity was potentiated by the lenvatinib plus everolimus combination in Caki-1 xenografts, in which fibroblast growth factor (FGF)-driven angiogenesis may contribute to tumor growth. The combination showed mostly additive activity in vascular endothelial growth factor (VEGF)-activated, and synergistic activity against FGF-activated endothelial cells, in cell proliferation and tube formation assays, as well as strongly suppressed mTOR-S6K-S6 signaling. Enhanced antitumor activities of the combination versus each monotherapy were also observed in mice bearing human pancreatic KP-1 xenografts overexpressing VEGF or FGF. Our results indicated that simultaneous targeting of tumor cell growth and angiogenesis by lenvatinib plus everolimus resulted in enhanced antitumor activity. The enhanced inhibition of both VEGF and FGF signaling pathways by the combination underlies its superior anti-angiogenic activity in human RCC xenograft models.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Neovascularization, Pathologic/prevention & control , Xenograft Model Antitumor Assays/methods , Animals , Blotting, Western , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cells, Cultured , Drug Synergism , Everolimus/administration & dosage , Everolimus/pharmacology , Fibroblast Growth Factor 1/genetics , Fibroblast Growth Factor 1/metabolism , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Phenylurea Compounds/administration & dosage , Phenylurea Compounds/pharmacology , Quinolines/administration & dosage , Quinolines/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Tumor Burden/drug effects , Tumor Burden/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
5.
Proc Natl Acad Sci U S A ; 111(46): 16556-61, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25368159

ABSTRACT

The motoneural control of skeletal muscle contraction requires the neuromuscular junction (NMJ), a midmuscle synapse between the motor nerve and myotube. The formation and maintenance of NMJs are orchestrated by the muscle-specific receptor tyrosine kinase (MuSK). Motor neuron-derived agrin activates MuSK via binding to MuSK's coreceptor Lrp4, and genetic defects in agrin underlie a congenital myasthenic syndrome (an NMJ disorder). However, MuSK-dependent postsynaptic differentiation of NMJs occurs in the absence of a motor neuron, indicating a need for nerve/agrin-independent MuSK activation. We previously identified the muscle protein Dok-7 as an essential activator of MuSK. Although NMJ formation requires agrin under physiological conditions, it is dispensable for NMJ formation experimentally in the absence of the neurotransmitter acetylcholine, which inhibits postsynaptic specialization. Thus, it was hypothesized that MuSK needs agrin together with Lrp4 and Dok-7 to achieve sufficient activation to surmount inhibition by acetylcholine. Here, we show that forced expression of Dok-7 in muscle enhanced MuSK activation in mice lacking agrin or Lrp4 and restored midmuscle NMJ formation in agrin-deficient mice, but not in Lrp4-deficient mice, probably due to the loss of Lrp4-dependent presynaptic differentiation. However, these NMJs in agrin-deficient mice rapidly disappeared after birth, and postsynaptic specializations emerged ectopically throughout myotubes whereas exogenous Dok-7-mediated MuSK activation was maintained. These findings demonstrate that the MuSK activator agrin plays another role essential for the postnatal maintenance, but not for embryonic formation, of NMJs and also for the postnatal, but not prenatal, midmuscle localization of postsynaptic specializations, providing physiological and pathophysiological insight into NMJ homeostasis.


Subject(s)
Agrin/physiology , Neuromuscular Junction/enzymology , Receptor Protein-Tyrosine Kinases/metabolism , Agrin/deficiency , Agrin/genetics , Alternative Splicing , Animals , Diaphragm/embryology , Diaphragm/growth & development , Enzyme Activation , Female , LDL-Receptor Related Proteins , Longevity/genetics , Male , Mice , Mice, Transgenic , Muscle Fibers, Skeletal/enzymology , Muscle Fibers, Skeletal/physiology , Muscle Proteins/deficiency , Muscle Proteins/physiology , Neuromuscular Junction/embryology , Neuromuscular Junction/growth & development , Neuromuscular Junction Diseases/enzymology , Neuromuscular Junction Diseases/genetics , Neuromuscular Junction Diseases/physiopathology , Phosphorylation , Post-Synaptic Density/physiology , Protein Processing, Post-Translational , Receptors, Cholinergic/physiology , Receptors, LDL/deficiency , Receptors, LDL/physiology , Recombinant Fusion Proteins/metabolism , Rotarod Performance Test
6.
FEBS Lett ; 587(23): 3749-54, 2013 Nov 29.
Article in English | MEDLINE | ID: mdl-24140340

ABSTRACT

Low-density lipoprotein receptor-related protein 4 (Lrp4) is essential for pre- and post-synaptic specialization at the neuromuscular junction (NMJ), an indispensable synapse between a motor nerve and skeletal muscle. Muscle-specific receptor tyrosine kinase MuSK must form a complex with Lrp4 to organize postsynaptic specialization at NMJs. Here, we show that the chaperon Mesdc2 binds to the intracellular form of Lrp4 and promotes its glycosylation and cell-surface expression. Furthermore, knockdown of Mesdc2 suppresses cell-surface expression of Lrp4, activation of MuSK, and postsynaptic specialization in muscle cells. These results suggest that Mesdc2 plays an essential role in NMJ formation by promoting Lrp4 maturation.


Subject(s)
Molecular Chaperones/metabolism , Muscle Fibers, Skeletal/metabolism , Neuromuscular Junction/metabolism , Receptors, LDL/metabolism , Animals , Cell Differentiation , Cell Membrane/metabolism , Glycosylation , HEK293 Cells , Humans , LDL-Receptor Related Proteins , Mice , Molecular Chaperones/genetics , Muscle Fibers, Skeletal/cytology , Myoblasts/cytology , Myoblasts/metabolism , Protein Processing, Post-Translational , Protein Transport , RNA, Small Interfering , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, LDL/genetics
7.
Curr Aging Sci ; 4(1): 1-7, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21204775

ABSTRACT

Various protein factors, including telomerase and WRN helicase, are involved in telomere maintenance. Resveratrol (Rsv), a polyphenol that extends the lifespan of diverse species is an activator of SIRT1, a NAD(+) dependent deacetylating enzyme in mammalian cells. Here, we examined the changes in gene expressions and promoter activities of WRN helicase and telomerase after Rsv treatment. This treatment increased the amount of WRN transcript and protein product by activating its promoter and telomerase promoter activity and gene expression. However cell proliferation was not changed. This suggests that Rsv induces telomere maintenance factors like WRN helicase without affecting cell proliferation.


Subject(s)
Antioxidants/pharmacology , Exodeoxyribonucleases/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Promoter Regions, Genetic/drug effects , RecQ Helicases/metabolism , Stilbenes/pharmacology , Telomerase/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Exodeoxyribonucleases/genetics , Female , HeLa Cells , Humans , RecQ Helicases/genetics , Resveratrol , Telomerase/genetics , Up-Regulation/drug effects , Uterine Cervical Neoplasms/enzymology , Uterine Cervical Neoplasms/pathology , Werner Syndrome Helicase
SELECTION OF CITATIONS
SEARCH DETAIL
...