Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Transl Res ; 260: 46-60, 2023 10.
Article in English | MEDLINE | ID: mdl-37353110

ABSTRACT

Head and neck cancers, which include oral squamous cell carcinoma (OSCC) as a major subsite, exhibit cellular plasticity that includes features of an epithelial-mesenchymal transition (EMT), referred to as partial-EMT (p-EMT). To identify molecular mechanisms contributing to OSCC plasticity, we performed a multiphase analysis of single cell RNA sequencing (scRNAseq) data from human OSCC. This included a multiresolution characterization of cancer cell subgroups to identify pathways and cell states that are heterogeneously represented, followed by casual inference analysis to elucidate activating and inhibitory relationships between these pathways and cell states. This approach revealed signaling networks associated with hierarchical cell state transitions, which notably included an association between ß-catenin-driven CREB-binding protein (CBP) activity and mTORC1 signaling. This network was associated with subpopulations of cancer cells that were enriched for markers of the p-EMT state and poor patient survival. Functional analyses revealed that ß-catenin/CBP induced mTORC1 activity in part through the transcriptional regulation of a raptor-interacting protein, chaperonin containing TCP1 subunit 5 (CCT5). Inhibition of ß-catenin-CBP activity through the use of the orally active small molecule, E7386, reduced the expression of CCT5 and mTORC1 activity in vitro, and inhibited p-EMT-associated markers and tumor development in a murine model of OSCC. Our study highlights the use of multiresolution network analyses of scRNAseq data to identify targetable signals for therapeutic benefit, thus defining an underappreciated association between ß-catenin/CBP and mTORC1 signaling in head and neck cancer plasticity.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Animals , Humans , Mice , beta Catenin/genetics , beta Catenin/metabolism , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement , CREB-Binding Protein/metabolism , Epithelial-Mesenchymal Transition , Mouth Neoplasms/drug therapy , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Squamous Cell Carcinoma of Head and Neck , Wnt Signaling Pathway
2.
Mol Cancer Res ; 19(9): 1510-1521, 2021 09.
Article in English | MEDLINE | ID: mdl-34039647

ABSTRACT

Immune checkpoint blockade (ICB) therapy has substantially improved the outcomes of patients with many types of cancers, including renal cell carcinoma (RCC). Initially studied as monotherapy, immunotherapy-based combination regimens have improved the clinical benefit achieved by ICB monotherapy and have revolutionized RCC treatment. While biomarkers like PD-L1 and tumor mutational burden (TMB) are FDA approved as biomarkers for ICB monotherapy, there are no known biomarkers for combination immunotherapies. Here, we describe the clinical outcomes and genomic determinants of response from a phase Ib/II clinical trial on patients with advanced RCC evaluating the efficacy of lenvatinib, a multi-kinase inhibitor mainly targeting VEGFR and FGFR plus pembrolizumab, an anti-PD1 immunotherapy. Concurrent treatment with lenvatinib and pembrolizumab resulted in an objective response rate of 79% (19/24) and tumor shrinkage in 96% (23/24) of patients. While tumor mutational burden (TMB) did not predict for clinical benefit, germline HLA-I diversity strongly impacted treatment efficacy. Specifically, HLA-I evolutionary divergence (HED), which measures the breadth of a patient's immunopeptidome, was associated with both improved clinical benefit and durability of response. Our results identify lenvatinib plus pembrolizumab as a highly active treatment strategy in RCC and reveal HLA-I diversity as a critical determinant of efficacy for this combination. HED also predicted better survival in a separate cohort of patients with RCC following therapy with anti-PD-1-based combination therapy. IMPLICATIONS: These findings have substantial implications for RCC therapy and for understanding immunogenetic mechanisms of efficacy and warrants further investigation.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/genetics , Carcinoma, Renal Cell/pathology , Genetic Variation , HLA Antigens/genetics , Kidney Neoplasms/pathology , Aged , Antibodies, Monoclonal, Humanized/administration & dosage , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , Female , Follow-Up Studies , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , Male , Middle Aged , Phenylurea Compounds/administration & dosage , Prognosis , Quinolines/administration & dosage , Survival Rate
3.
Cancer Res ; 81(4): 1052-1062, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33408116

ABSTRACT

The Wnt/ß-catenin signaling pathway plays crucial roles in embryonic development and the development of multiple types of cancer, and its aberrant activation provides cancer cells with escape mechanisms from immune checkpoint inhibitors. E7386, an orally active selective inhibitor of the interaction between ß-catenin and CREB binding protein, which is part of the Wnt/ß-catenin signaling pathway, disrupts the Wnt/ß-catenin signaling pathway in HEK293 and adenomatous polyposis coli (APC)-mutated human gastric cancer ECC10 cells. It also inhibited tumor growth in an ECC10 xenograft model and suppressed polyp formation in the intestinal tract of ApcMin /+ mice, in which mutation of Apc activates the Wnt/ß-catenin signaling pathway. E7386 demonstrated antitumor activity against mouse mammary tumors developed in mouse mammary tumor virus (MMTV)-Wnt1 transgenic mice. Gene expression profiling using RNA sequencing data of MMTV-Wnt1 tumor tissue from mice treated with E7386 showed that E7386 downregulated genes in the hypoxia signaling pathway and immune responses related to the CCL2, and IHC analysis showed that E7386 induced infiltration of CD8+ cells into tumor tissues. Furthermore, E7386 showed synergistic antitumor activity against MMTV-Wnt1 tumor in combination with anti-PD-1 antibody. In conclusion, E7386 demonstrates clear antitumor activity via modulation of the Wnt/ß-catenin signaling pathway and alteration of the tumor and immune microenvironments, and its antitumor activity can be enhanced in combination with anti-PD-1 antibody. SIGNIFICANCE: These findings demonstrate that the novel anticancer agent, E7386, modulates Wnt/ß-catenin signaling, altering the tumor immune microenvironment and exhibiting synergistic antitumor activity in combination with anti-PD-1 antibody.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/pathology , Peptide Fragments/metabolism , Pyrazines/pharmacology , Sialoglycoproteins/metabolism , Triazines/pharmacology , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism , Animals , Antineoplastic Agents/therapeutic use , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Female , Genes, APC , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Peptide Fragments/antagonists & inhibitors , Protein Binding/drug effects , Pyrazines/therapeutic use , Sialoglycoproteins/antagonists & inhibitors , Triazines/therapeutic use , Wnt Signaling Pathway/genetics , Wnt1 Protein/genetics , Wnt1 Protein/metabolism , beta Catenin/antagonists & inhibitors
4.
Breast Cancer ; 27(4): 706-715, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32133606

ABSTRACT

BACKGROUND: Eribulin, a nontaxane synthetic inhibitor of microtubule dynamics, is widely used to manage locally advanced or metastatic breast cancer (MBC). Eribulin has demonstrated immunomodulatory activity on the tumour microenvironment. Baseline neutrophil-to-lymphocyte ratio (NLR), a marker of immune status, may predict progression-free survival in eribulin treatment. This post hoc analysis assessed predictors for overall survival (OS). METHODS: The phase 3 open-label study (EMBRACE) of eribulin versus treatment of physician's choice (TPC) in patients with MBC provided source data. Baseline absolute lymphocyte counts (ALCs) and NLR were evaluable in 751 and 713 patients, respectively. RESULTS: Eribulin prolonged OS versus TPC in patients with baseline ALC ≥ 1500/µl (hazard ratio [HR] 0.586; 95% confidence interval [CI] 0.437-0.784; P < 0.001). There was no significant difference by treatment for ALC < 1500/µl (HR 1.002; 95% CI 0.800-1.253; P = 0.989). Univariate and multivariate analyses were performed and identified baseline ALC as a potential predictor of OS in eribulin-treated patients. Interaction analysis of OS supported 1500/µl as a potentially differential cutoff value. NLR at a cutoff value of 3 was associated with prolonged OS (eribulin group). However, similar results were also observed in the TPC group, without apparent interaction effect, suggesting that NLR may be a general prognostic marker rather than a specific predictor of OS for eribulin. DISCUSSION: This hypothesis-generating study speculates that baseline ALC may be an independent predictor for longer OS in eribulin-treated MBC patients and could be clinically impactful because it can be evaluated without the need for additional invasive procedures. TRIAL REGISTRATION: www.ClinicalTrials.gov code: NCT00388726.


Subject(s)
Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Furans/administration & dosage , Ketones/administration & dosage , Lymphocytes/immunology , Aged , Breast Neoplasms/blood , Breast Neoplasms/immunology , Breast Neoplasms/mortality , Drug Administration Schedule , Female , Humans , Infusions, Intravenous , Kaplan-Meier Estimate , Lymphocyte Count , Middle Aged , Neutrophils/immunology , Prognosis , Progression-Free Survival , Tumor Microenvironment
5.
Mol Omics ; 16(3): 195-209, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32203567

ABSTRACT

Epidermal growth factor receptor (EGFR) is a major driver of head and neck cancer, a devastating malignancy with a major sub-site in the oral cavity manifesting as oral squamous cell carcinoma (OSCC). EGFR is a glycoprotein receptor tyrosine kinase (RTK) whose activity is upregulated in >80% OSCC. Current anti-EGFR therapy relies on the use of cetuximab, a monoclonal antibody against EGFR, although it has had only a limited response in patients. Here, we uncover a novel mechanism regulating EGFR activity, identifying a role of the nuclear branch of the Wnt/ß-catenin signaling pathway, the ß-catenin/CBP axis, in control of post-translational modification of N-glycans on the EGFR. Genomic and structural analyses reveal that ß-catenin/CBP signaling represses fucosylation on the antennae of N-linked glycans on EGFR. By employing nUPLC-MS/MS, we determined that malignant human OSCC cells harbor EGFR with a paucity of N-glycan antennary fucosylation, while indolent cells display higher levels of fucosylation at sites N420 and N579. Additionally, treatment with either ICG-001 or E7386, which are both small molecule inhibitors of ß-catenin/CBP signaling, leads to increased transcriptional expression of fucosyltransferases FUT2 and FUT3, with a concomitant increase in EGFR N-glycan antennary fucosylation. In order to discover which fucosylated glycan epitopes are involved in the observed effect, we performed in-depth characterization of multiply-fucosylated N-glycans via tandem mass spectrometry analysis of the EGFR tryptic glycopeptides. Data are available via ProteomeXchange with identifier PXD017060. We propose that ß-catenin/CBP signaling promotes EGFR oncogenic activity in OSCC by inhibiting its N-glycan antennary fucosylation through transcriptional repression of FUT2 and FUT3.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Fucose/metabolism , Fucosyltransferases/genetics , Mouth Neoplasms/drug therapy , Small Molecule Libraries/administration & dosage , Animals , Binding Sites , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , CREB-Binding Protein/metabolism , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , ErbB Receptors/chemistry , ErbB Receptors/metabolism , Fucosyltransferases/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Models, Molecular , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Neoplasm Metastasis , Polysaccharides/metabolism , Protein Structure, Tertiary , Pyrimidinones/administration & dosage , Pyrimidinones/pharmacology , Small Molecule Libraries/pharmacology , Wnt Signaling Pathway/drug effects , Xenograft Model Antitumor Assays , beta Catenin/metabolism , Galactoside 2-alpha-L-fucosyltransferase
6.
Cell Commun Signal ; 17(1): 67, 2019 06 17.
Article in English | MEDLINE | ID: mdl-31208428

ABSTRACT

BACKGROUND: Chlorotoxin (Cltx) isolated from scorpion venom is an established tumor targeting and antiangiogenic peptide. Radiolabeled Cltx therapeutic (131I-TM601) yielded promising results in human glioma clinical studies, and the imaging agent tozuleristide, is under investigation in CNS cancer studies. Several binding targets have previously been proposed for Cltx but none effectively explain its pleiotropic effects; its true target remains ambiguous and is the focus of this study. METHODS: A peptide-drug conjugate (ER-472) composed of Cltx linked to cryptophycin as warhead was developed as a tool to probe the molecular target and mechanism of action of Cltx, using multiple xenograft models. RESULTS: Neuropilin-1 (NRP1), an endocytic receptor on tumor and endothelial cells, was identified as a novel Cltx target, and NRP1 binding by Cltx increased drug uptake into tumor. Metabolism of Cltx to peptide bearing free C-terminal arginine, a prerequisite for NRP1 binding, took place in the tumor microenvironment, while native scorpion Cltx with amidated C-terminal arginine did not bind NRP1, and instead acts as a cryptic peptide. Antitumor activity of ER-472 in xenografts correlated to tumor NRP1 expression. Potency was significantly reduced by treatment with NRP1 blocking antibodies or knockout in tumor cells, confirming a role for NRP1-binding in ER-472 activity. Higher cryptophycin metabolite levels were measured in NRP1-expressing tumors, evidence of NRP1-mediated enhanced drug uptake and presumably responsible for the superior antitumor efficacy. CONCLUSIONS: NRP1 was identified as a novel Cltx target which enhances tumor drug uptake. This finding should facilitate tumor selection for chlorotoxin-based therapeutics and diagnostics.


Subject(s)
Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Neuropilin-1/metabolism , Scorpion Venoms/metabolism , Scorpion Venoms/pharmacology , Amino Acid Sequence , Animals , Antineoplastic Agents/chemistry , Biological Transport , Cell Line, Tumor , Depsipeptides/chemistry , Humans , Mice , Neuropilin-1/chemistry , Scorpion Venoms/chemistry
7.
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
8.
Cancer Invest ; 37(4-5): 185-198, 2019.
Article in English | MEDLINE | ID: mdl-31006280

ABSTRACT

High expression of vascular endothelial growth factor (VEGF) in patients with hepatocellular carcinoma (HCC) is associated with poor prognosis. Here, we investigated the antitumor activity of lenvatinib, a multiple receptor tyrosine kinase inhibitor, in VEGF-overexpressing HCC models. In human umbilical vein endothelial cells, lenvatinib showed potent inhibitory activities against VEGF-induced proliferation and VEGF/basic fibroblast growth factor-induced tube formation. In VEGF-overexpressing HCC xenograft models, characterized by aggressive tumor growth and hypervascularity, lenvatinib had significant antitumor and antiangiogenic activities. These results suggest that potent activity of lenvatinib against VEGF signaling underlies its antitumor and antiangiogenic activities in the hypervascular HCC models.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Phenylurea Compounds/pharmacology , Quinolines/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Female , Humans , Liver Neoplasms/blood supply , Liver Neoplasms/pathology , Mice , Neovascularization, Pathologic/pathology , Phenylurea Compounds/therapeutic use , Quinolines/therapeutic use , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
9.
PLoS One ; 14(2): e0212513, 2019.
Article in English | MEDLINE | ID: mdl-30811474

ABSTRACT

Lenvatinib is a multiple receptor tyrosine kinase inhibitor targeting mainly vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF) receptors. We investigated the immunomodulatory activities of lenvatinib in the tumor microenvironment and its mechanisms of enhanced antitumor activity when combined with a programmed cell death-1 (PD-1) blockade. Antitumor activity was examined in immunodeficient and immunocompetent mouse tumor models. Single-cell analysis, flow cytometric analysis, and immunohistochemistry were used to analyze immune cell populations and their activation. Gene co-expression network analysis and pathway analysis using RNA sequencing data were used to identify lenvatinib-driven combined activity with anti-PD-1 antibody (anti-PD-1). Lenvatinib showed potent antitumor activity in the immunocompetent tumor microenvironment compared with the immunodeficient tumor microenvironment. Antitumor activity of lenvatinib plus anti-PD-1 was greater than that of either single treatment. Flow cytometric analysis revealed that lenvatinib reduced tumor-associated macrophages (TAMs) and increased the percentage of activated CD8+ T cells secreting interferon (IFN)-γ+ and granzyme B (GzmB). Combination treatment further increased the percentage of T cells, especially CD8+ T cells, among CD45+ cells and increased IFN-γ+ and GzmB+ CD8+ T cells. Transcriptome analyses of tumors resected from treated mice showed that genes specifically regulated by the combination were significantly enriched for type-I IFN signaling. Pretreatment with lenvatinib followed by anti-PD-1 treatment induced significant antitumor activity compared with anti-PD-1 treatment alone. Our findings show that lenvatinib modulates cancer immunity in the tumor microenvironment by reducing TAMs and, when combined with PD-1 blockade, shows enhanced antitumor activity via the IFN signaling pathway. These findings provide a scientific rationale for combination therapy of lenvatinib with PD-1 blockade to improve cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Neoplasms, Experimental/immunology , Neoplasms, Experimental/therapy , Phenylurea Compounds/administration & dosage , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Quinolines/administration & dosage , Animals , Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Gene Expression/drug effects , Gene Expression/immunology , Immunologic Factors/administration & dosage , Interferons/metabolism , Lymphocyte Activation/drug effects , Macrophages/drug effects , Macrophages/immunology , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Neoplasms, Experimental/genetics , Protein Kinase Inhibitors/administration & dosage , Signal Transduction/drug effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
10.
Cancer Sci ; 109(12): 3993-4002, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30447042

ABSTRACT

Angiogenesis inhibitors such as lenvatinib and sorafenib, and an immune checkpoint inhibitor (ICI), nivolumab, are used for anticancer therapies against advanced hepatocellular carcinoma (HCC). Combination treatments comprising angiogenesis inhibitors plus ICIs are promising options for improving clinical benefits in HCC patients, and clinical trials are ongoing. Here, we investigated the antitumor and immunomodulatory activities of lenvatinib (a multiple receptor tyrosine kinase inhibitor targeting vascular endothelial growth factor receptor 1-3, fibroblast growth factor receptor 1-4, platelet-derived growth factor receptor α, KIT and RET) and the combined antitumor activity of lenvatinib plus anti-programmed cell death 1 (PD-1) antibody in the Hepa1-6 mouse HCC syngeneic model. We found that the antitumor activities of lenvatinib and sorafenib were not different in immunodeficient mice, but lenvatinib showed more potent antitumor activity than sorafenib in immunocompetent mice. The antitumor activity of lenvatinib was greater in immunocompetent mice than in immunodeficient mice and was attenuated by CD8+ T cell depletion. Treatment with lenvatinib plus anti-PD-1 antibody resulted in more tumor regression and a higher response rate compared with either treatment alone in immunocompetent mice. Single-cell RNA sequencing analysis demonstrated that treatment with lenvatinib with or without anti-PD-1 antibody decreased the proportion of monocytes and macrophages population and increased that of CD8+ T cell populations. These data suggest that lenvatinib has immunomodulatory activity that contributes to the antitumor activity of lenvatinib and enhances the antitumor activity in combination treatment with anti-PD-1 antibody. Combination treatment of lenvatinib plus anti-PD-1 antibody therefore warrants further investigation against advanced HCC.


Subject(s)
Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents/administration & dosage , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Phenylurea Compounds/administration & dosage , Quinolines/administration & dosage , Sorafenib/administration & dosage , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Immunological/pharmacology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Immunocompetence , Immunomodulation , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Mice , Phenylurea Compounds/pharmacology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Quinolines/pharmacology , Sequence Analysis, RNA , Single-Cell Analysis , Sorafenib/pharmacology , Xenograft Model Antitumor Assays
11.
Phys Rev Lett ; 121(10): 102701, 2018 Sep 07.
Article in English | MEDLINE | ID: mdl-30240253

ABSTRACT

The isotope ^{98}Tc decays to ^{98}Ru with a half-life of 4.2×10^{6} yr and could have been present in the early Solar System. In this Letter, we report on the first calculations of the production of ^{98}Tc by neutrino-induced reactions in core-collapse supernovae (the ν process). Our predicted ^{98}Tc abundance at the time of solar system formation is not much lower than the current measured upper limit raising the possibility for its detection in the not too distant future. We show that, if the initial abundance were to be precisely measured, the ^{98}Tc nuclear cosmochronometer could be used to evaluate a much more precise value of the duration time from the last core-collapse supernova to the formation of the solar system. Moreover, a unique and novel feature of the ^{98}Tc ν-process nucleosynthesis is the large contribution (∼20%) from charged current reactions with electron antineutrinos. This means that ^{98}Tc becomes a unique new ν-process probe of the temperature of the electron antineutrinos.

12.
Anticancer Res ; 38(6): 3375-3385, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29848686

ABSTRACT

BACKGROUND: Eribulin is used in many countries to treat patients with advanced breast cancer or liposarcoma and exerts in vivo anticancer activity under monotherapy conditions against various human tumor xenograft models. Here, eribulin in combination with mechanistically different anticancer agents was evaluated. MATERIALS AND METHODS: Eribulin was combined with cytotoxic agents (capecitabine, carboplatin, cisplatin, doxorubicin, gemcitabine) or targeted agents (bevacizumab, BKM-120, E7449, erlotinib, everolimus, lenvatinib, palbociclib) in tumor xenograft models of breast cancer, melanoma, non-small cell lung cancer (NSCLC), and ovarian cancer. RESULTS: Across nearly all models, eribulin with either cytotoxic or targeted agents demonstrated combination activity, defined as the activity demonstrably greater than that of either agent alone. Combination activity was absent only with doxorubicin (MDA-MB-435 model) and with lenvatinib (NCI-H1975 model), both of which responded to the agents as monotherapy. CONCLUSION: Eribulin has combination activity with multiple agents from different mechanistic classes in several human cancer models, including breast, NSCLC, ovarian, and melanoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Neoplasms/drug therapy , Xenograft Model Antitumor Assays/methods , A549 Cells , Animals , Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Drug Synergism , Female , Furans/administration & dosage , Humans , Ketones/administration & dosage , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , MCF-7 Cells , Melanoma/drug therapy , Melanoma/pathology , Mice , Neoplasms/pathology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology
13.
Cancer Res ; 78(3): 817-829, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29191802

ABSTRACT

Chemotherapy-induced peripheral neuropathy (CIPN) is a major cause of disability in cancer survivors. CIPN investigations in preclinical model systems have focused on either behaviors or acute changes in nerve conduction velocity (NCV) and amplitude, but greater understanding of the underlying nature of axonal injury and its long-term processes is needed as cancer patients live longer. In this study, we used multiple independent endpoints to systematically characterize CIPN recovery in mice exposed to the antitubulin cancer drugs eribulin, ixabepilone, paclitaxel, or vinorelbine at MTDs. All of the drugs ablated intraepidermal nerve fibers and produced axonopathy, with a secondary disruption in myelin structure within 2 weeks of drug administration. In addition, all of the drugs reduced sensory NCV and amplitude, with greater deficits after paclitaxel and lesser deficits after ixabepilone. These effects correlated with degeneration in dorsal root ganglia (DRG) and sciatic nerve and abundance of Schwann cells. Although most injuries were fully reversible after 3-6 months after administration of eribulin, vinorelbine, and ixabepilone, we observed delayed recovery after paclitaxel that produced a more severe, pervasive, and prolonged neurotoxicity. Compared with other agents, paclitaxel also displayed a unique prolonged exposure in sciatic nerve and DRG. The most sensitive indicator of toxicity was axonopathy and secondary myelin changes accompanied by a reduction in intraepidermal nerve fiber density. Taken together, our findings suggest that intraepidermal nerve fiber density and changes in NCV and amplitude might provide measures of axonal injury to guide clinical practice.Significance: This detailed preclinical study of the long-term effects of widely used antitubulin cancer drugs on the peripheral nervous system may help guide clinical evaluations to improve personalized care in limiting neurotoxicity in cancer survivors. Cancer Res; 78(3); 817-29. ©2017 AACR.


Subject(s)
Ganglia, Spinal/drug effects , Microtubules/drug effects , Peripheral Nervous System Diseases/chemically induced , Recovery of Function/drug effects , Schwann Cells/drug effects , Sciatic Nerve/drug effects , Tubulin Modulators/toxicity , Acute Disease , Animals , Cells, Cultured , Female , Ganglia, Spinal/injuries , Ganglia, Spinal/pathology , Mice , Mice, Inbred BALB C , Microtubules/pathology , Peripheral Nervous System Diseases/pathology , Schwann Cells/pathology , Sciatic Nerve/injuries , Sciatic Nerve/pathology
14.
Nature ; 550(7674): 80-83, 2017 10 04.
Article in English | MEDLINE | ID: mdl-28980637

ABSTRACT

Type Ia supernovae arise from the thermonuclear explosion of white-dwarf stars that have cores of carbon and oxygen. The uniformity of their light curves makes these supernovae powerful cosmological distance indicators, but there have long been debates about exactly how their explosion is triggered and what kind of companion stars are involved. For example, the recent detection of the early ultraviolet pulse of a peculiar, subluminous type Ia supernova has been claimed as evidence for an interaction between a red-giant or a main-sequence companion and ejecta from a white-dwarf explosion. Here we report observations of a prominent but red optical flash that appears about half a day after the explosion of a type Ia supernova. This supernova shows hybrid features of different supernova subclasses, namely a light curve that is typical of normal-brightness supernovae, but with strong titanium absorption, which is commonly seen in the spectra of subluminous ones. We argue that this early flash does not occur through previously suggested mechanisms such as the companion-ejecta interaction. Instead, our simulations show that it could occur through detonation of a thin helium shell either on a near-Chandrasekhar-mass white dwarf, or on a sub-Chandrasekhar-mass white dwarf merging with a less-massive white dwarf. Our finding provides evidence that one branch of previously proposed explosion models-the helium-ignition branch-does exist in nature, and that such a model may account for the explosions of white dwarfs in a mass range wider than previously supposed.

15.
Oncoimmunology ; 6(8): e1338239, 2017.
Article in English | MEDLINE | ID: mdl-28920002

ABSTRACT

Reprogramming of immunosuppressive tumor microenvironment (TME) by targeting alternatively activated tumor associated macrophages (M2TAM), myeloid-derived suppressor cells (MDSC), and regulatory T cells (Tregs), represents a promising strategy for developing novel cancer immunotherapy. Prostaglandin E2 (PGE2), an arachidonic acid pathway metabolite and mediator of chronic inflammation, has emerged as a powerful immunosuppressor in the TME through engagement with one or more of its 4 receptors (EP1-EP4). We have developed E7046, an orally bioavailable EP4-specific antagonist and show here that E7046 has specific and potent inhibitory activity on PGE2-mediated pro-tumor myeloid cell differentiation and activation. E7046 treatment reduced the growth or even rejected established tumors in vivo in a manner dependent on both myeloid and CD8+ T cells. Furthermore, co-administration of E7046 and E7777, an IL-2-diphtheria toxin fusion protein that preferentially kills Tregs, synergistically disrupted the myeloid and Treg immunosuppressive networks, resulting in effective and durable anti-tumor immune responses in mouse tumor models. In the TME, E7046 and E7777 markedly increased ratios of CD8+granzymeB+ cytotoxic T cells (CTLs)/live Tregs and of M1-like/M2TAM, and converted a chronic inflammation phenotype into acute inflammation, shown by substantial induction of STAT1/IRF-1 and IFNγ-controlled genes. Notably, E7046 also showed synergistic anti-tumor activity when combined with anti-CTLA-4 antibodies, which have been reported to diminish intratumoral Tregs. Our studies thus reveal a specific myeloid cell differentiation-modifying activity by EP4 blockade and a novel combination of E7046 and E7777 as a means to synergistically mitigate both myeloid and Treg-derived immunosuppression for cancer treatment in preclinical models.

16.
Cancer Res ; 76(11): 3332-9, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27197173

ABSTRACT

Chemotherapy-induced peripheral neuropathy is a dose-limiting side effect of many antineoplastic agents, but the mechanisms underlying the toxicities are unclear. At their MTDs, the microtubule-binding drugs paclitaxel and ixabepilone induce more severe neuropathy in mice relative to eribulin mesylate, paralleling their toxicity profiles in clinic. We hypothesized that the severity of their neurotoxic effects might be explained by the levels at which they accumulate in the peripheral nervous system. To test this hypothesis, we compared their pharmacokinetics and distribution in peripheral nerve tissue. After administration of a single intravenous dose, each drug was rapidly cleared from plasma but all persisted in the dorsal root ganglia (DRG) and sciatic nerve (SN) for up to 72 hours. Focusing on paclitaxel and eribulin, we performed a 2-week MTD-dosing regimen, followed by a determination of drug pharmacokinetics, tissue distribution, and multiple functional measures of peripheral nerve toxicity for 4 weeks. Consistent with the acute dosing study, both drugs persisted in peripheral nervous tissues for weeks, in contrast to their rapid clearance from plasma. Notably, although eribulin exhibited greater DRG and SN penetration than paclitaxel, the neurotoxicity observed functionally was consistently more severe with paclitaxel. Overall, our results argue that sustained exposure of microtubule-binding chemotherapeutic agents in peripheral nerve tissues cannot by itself account for their associated neurotoxicity. Cancer Res; 76(11); 3332-9. ©2016 AACR.


Subject(s)
Apoptosis/drug effects , Ganglia, Spinal/pathology , Microtubules/pathology , Paclitaxel/pharmacology , Peripheral Nervous System Diseases/pathology , Sciatic Nerve/pathology , Animals , Blotting, Western , Cell Proliferation , Dose-Response Relationship, Drug , Electrophysiology , Epothilones/pharmacokinetics , Epothilones/pharmacology , Female , Furans/pharmacokinetics , Furans/pharmacology , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Ketones/pharmacokinetics , Ketones/pharmacology , Mice , Mice, Inbred BALB C , Microtubules/drug effects , Microtubules/metabolism , Paclitaxel/pharmacokinetics , Peripheral Nervous System Diseases/drug therapy , Peripheral Nervous System Diseases/metabolism , Sciatic Nerve/drug effects , Sciatic Nerve/metabolism , Time Factors , Tissue Distribution , Tubulin Modulators/pharmacokinetics , Tubulin Modulators/pharmacology
17.
Mol Cancer Ther ; 15(6): 1208-16, 2016 06.
Article in English | MEDLINE | ID: mdl-27196783

ABSTRACT

Apratoxin A is a natural product with potent antiproliferative activity against many human cancer cell lines. However, we and other investigators observed that it has a narrow therapeutic window in vivo Previous mechanistic studies have suggested its involvement in the secretory pathway as well as the process of chaperone-mediated autophagy. Still the link between the biologic activities of apratoxin A and its in vivo toxicity has remained largely unknown. A better understanding of this relationship is critically important for any further development of apratoxin A as an anticancer drug. Here, we describe a detailed pathologic analysis that revealed a specific pancreas-targeting activity of apratoxin A, such that severe pancreatic atrophy was observed in apratoxin A-treated animals. Follow-up tissue distribution studies further uncovered a unique drug distribution profile for apratoxin A, showing high drug exposure in pancreas and salivary gland. It has been shown previously that apratoxin A inhibits the protein secretory pathway by preventing cotranslational translocation. However, the molecule targeted by apratoxin A in this pathway has not been well defined. By using a (3)H-labeled apratoxin A probe and specific Sec 61α/ß antibodies, we identified that the Sec 61 complex is the molecular target of apratoxin A. We conclude that apratoxin A in vivo toxicity is likely caused by pancreas atrophy due to high apratoxin A exposure. Mol Cancer Ther; 15(6); 1208-16. ©2016 AACR.


Subject(s)
Antineoplastic Agents/toxicity , Depsipeptides/toxicity , Neoplasms/drug therapy , Pancreas/drug effects , SEC Translocation Channels/metabolism , A549 Cells , Animals , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Depsipeptides/pharmacokinetics , Humans , MCF-7 Cells , Maximum Tolerated Dose , Mice , Neoplasm Transplantation , Neoplasms/metabolism , Organ Specificity , Protein Binding , Rats
18.
Cancer Res ; 76(6): 1528-37, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26822154

ABSTRACT

Fms-like tyrosine kinase 3 (FLT3) inhibition has elicited encouraging responses in acute myeloid leukemia (AML) therapy. Unfortunately, unless combined with a bone marrow transplant, disease relapse is frequent. In addition to the acquired point mutations in the FLT3 kinase domain that contribute to FLT3 inhibitor resistance, MEK/ERK signaling is persistently activated in AML cells even when FLT3 phosphorylation is continually suppressed. Thus, concomitant targeting of FLT3 and MAPK may potentially exert synergistic activity to counteract the resistance of AML cells to FLT3-targeted therapy. In this study, we investigated the antileukemia activity of a MEK1 and FLT3 dual inhibitor, E6201, in AML cells resistant to FLT3 inhibition. We found that E6201 exerted profound apoptogenic effects on AML cells harboring resistance-conferring FLT3 mutations. This activity appeared to be p53 dependent, and E6201-induced cytotoxicity was retained under hypoxic culture conditions and during coculture with mesenchymal stem cells that mimic the AML microenvironment. Furthermore, E6201 markedly reduced leukemia burden and improved the survival of mice in a human FLT3-mutated AML model. Collectively, our data provide a preclinical basis for the clinical evaluation of E6201 in AML patients harboring FLT3 mutations, including those who relapse following FLT3-targeted monotherapy.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Lactones/pharmacology , Leukemia, Myeloid, Acute/drug therapy , MAP Kinase Signaling System/drug effects , Point Mutation/drug effects , Signal Transduction/drug effects , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Female , Humans , Leukemia, Myeloid, Acute/genetics , MAP Kinase Signaling System/genetics , Mice , Phosphorylation/drug effects , Phosphorylation/genetics , Point Mutation/genetics , Protein Kinase Inhibitors/pharmacology , Signal Transduction/genetics
19.
Gan To Kagaku Ryoho ; 43(12): 1538-1540, 2016 Nov.
Article in Japanese | MEDLINE | ID: mdl-28133049

ABSTRACT

Case 1 involved an 87-year-old woman. She had increased serum amylase levels, and intraductal papillary mucinous adenocarcinoma( IPMC)in the accessory pancreatic duct was diagnosed following various medical examinations. Radical surgery is possible for such a lesion when there are no issues detected following evaluation. The patient and her family decided that she should undergo surgery; therefore, pancreatoduodenectomy was performed for Stage III IPMC. Various morbidities required medical treatment, but she was finally discharged from hospital 89 days after surgery. She has had no recurrence 2 years and 11 months after surgery. Case 2 involved a 90-year-old man. He had liver dysfunction, and progressive hepatocellular carcinoma( HCC)was diagnosed based on computed tomography(CT)findings. The tumor size reduced to around 10 cm with enforced 2 degrees TACE. The patient and his family chose for him to undergo radical surgery; therefore, right trisegmentectomy of the liver was performed for Stage III HCC. He was discharged from the hospital 22 days after surgery, and has had no recurrence for the past 1.6 years. More careful examination and a general assessment are necessary to make decisions on surgical indications in super elderly patients.


Subject(s)
Adenocarcinoma, Mucinous/surgery , Carcinoma, Hepatocellular/surgery , Liver Neoplasms/surgery , Pancreatic Neoplasms/surgery , Adenocarcinoma, Mucinous/diagnostic imaging , Aged, 80 and over , Carcinoma, Hepatocellular/diagnostic imaging , Female , Humans , Liver Neoplasms/diagnostic imaging , Male , Pancreatic Neoplasms/pathology , Pancreaticoduodenectomy , Tomography, X-Ray Computed , Treatment Outcome
20.
Oncotarget ; 6(38): 41307-23, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26513298

ABSTRACT

Inhibition of Poly(ADP-ribose) Polymerase1 (PARP1) impairs DNA damage repair, and early generation PARP1/2 inhibitors (olaparib, niraparib, etc.) have demonstrated clinical proof of concept for cancer treatment. Here, we describe the development of the novel PARP inhibitor E7449, a potent PARP1/2 inhibitor that also inhibits PARP5a/5b, otherwise known as tankyrase1 and 2 (TNKS1 and 2), important regulators of canonical Wnt/ß-catenin signaling. E7449 inhibits PARP enzymatic activity and additionally traps PARP1 onto damaged DNA; a mechanism previously shown to augment cytotoxicity. Cells deficient in DNA repair pathways beyond homologous recombination were sensitive to E7449 treatment. Chemotherapy was potentiated by E7449 and single agent had significant antitumor activity in BRCA-deficient xenografts. Additionally, E7449 inhibited Wnt/ß-catenin signaling in colon cancer cell lines, likely through TNKS inhibition. Consistent with this possibility, E7449 stabilized axin and TNKS proteins resulting in ß-catenin de-stabilization and significantly altered expression of Wnt target genes. Notably, hair growth mediated by Wnt signaling was inhibited by E7449. A pharmacodynamic effect of E7449 on Wnt target genes was observed in tumors, although E7449 lacked single agent antitumor activity in vivo, a finding typical for selective TNKS inhibitors. E7449 antitumor activity was increased through combination with MEK inhibition. Particularly noteworthy was the lack of toxicity, most significantly the lack of intestinal toxicity reported for other TNKS inhibitors. E7449 represents a novel dual PARP1/2 and TNKS1/2 inhibitor which has the advantage of targeting Wnt/ß-catenin signaling addicted tumors. E7449 is currently in early clinical development.


Subject(s)
Cell Proliferation/drug effects , DNA Repair/drug effects , Isoquinolines/pharmacology , Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Quinazolinones/pharmacology , Wnt Signaling Pathway/drug effects , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Azo Compounds , Blotting, Western , Carboplatin/administration & dosage , Cell Line, Tumor , Cell Proliferation/genetics , DNA Repair/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/drug effects , Isoquinolines/chemistry , Mice, Inbred C57BL , Mice, Nude , Mice, SCID , Neoplasms/genetics , Neoplasms/pathology , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Poly(ADP-ribose) Polymerase Inhibitors/administration & dosage , Poly(ADP-ribose) Polymerase Inhibitors/chemistry , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Quinazolinones/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Tankyrases/antagonists & inhibitors , Tankyrases/genetics , Tankyrases/metabolism , Temozolomide , Tumor Burden/drug effects , Wnt Signaling Pathway/genetics , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...