Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Cancer Res ; 29(10): 1855-1868, 2023 05 15.
Article in English | MEDLINE | ID: mdl-36520504

ABSTRACT

Utilizing targeted therapies capable of reducing cancer metastasis, targeting chemoresistant and self-renewing cancer stem cells, and augmenting the efficacy of systemic chemo/radiotherapies is vital to minimize cancer-associated mortality. Targeting nitric oxide synthase (NOS), a protein within the tumor microenvironment, has gained interest as a promising therapeutic strategy to reduce metastatic capacity and augment the efficacy of chemo/radiotherapies in various solid malignancies. Our review highlights the influence of nitric oxide (NO) in tumor progression and cancer metastasis, as well as promising preclinical studies that evaluated NOS inhibitors as anticancer therapies. Lastly, we highlight the prospects and outstanding challenges of using NOS inhibitors in the clinical setting.


Subject(s)
Neoplasms , Nitric Oxide , Humans , Nitric Oxide/metabolism , Neoplasms/drug therapy , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II/metabolism , Tumor Microenvironment
2.
Breast Cancer Res ; 22(1): 121, 2020 11 04.
Article in English | MEDLINE | ID: mdl-33148288

ABSTRACT

Metaplastic breast cancer (MpBC) is an exceedingly rare breast cancer variant that is therapeutically challenging and aggressive. MpBC is defined by the histological presence of at least two cellular types, typically epithelial and mesenchymal components. This variant harbors a triple-negative breast cancer (TNBC) phenotype, yet has a worse prognosis and decreased survival compared to TNBC. There are currently no standardized treatment guidelines specifically for MpBC. However, prior studies have found that MpBC typically has molecular alterations in epithelial-to-mesenchymal transition, amplification of epidermal growth factor receptor, PI3K/Akt signaling, nitric oxide signaling, Wnt/ß-catenin signaling, altered immune response, and cell cycle dysregulation. Some of these molecular alterations have been studied as therapeutic targets, in both the preclinical and clinical setting. This current review discusses the histological organization and cellular origins of MpBC, molecular alterations, the role of radiation therapy, and current clinical trials for MpBC.


Subject(s)
Breast Neoplasms/pathology , Epithelial-Mesenchymal Transition , Genes, Neoplasm/genetics , Metaplasia/pathology , Triple Negative Breast Neoplasms/pathology , Wnt Signaling Pathway , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Cell Line, Tumor , Female , Humans , Metaplasia/genetics , Metaplasia/metabolism , Metaplasia/therapy , Molecular Targeted Therapy/methods , Nitric Oxide Synthase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/therapy
3.
Breast J ; 26(9): 1793-1796, 2020 09.
Article in English | MEDLINE | ID: mdl-32578306

ABSTRACT

Metastatic triple-negative breast cancer (mTNBC) patients tend to have a poor overall survival. The primary goals of treatment focus on palliation of symptoms and improvement in overall survival (OS). Single-agent sequential chemotherapy with anthracycline or taxane has remained the cornerstone of treatment for many years. The FDA has approved newer agents such as poly-adenosine diphosphate-ribose polymerase (PARP) inhibitors upfront in germline BRCA (gBRCA) 1/2 mutation carriers; atezolizumab and nab-paclitaxel combination frontline in patients with PD-L1 expression > 1%; and sacituzumab govitecan (IMMU-132), an antibody-drug conjugate in heavily pretreated mTNBC patients.


Subject(s)
Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/drug therapy
4.
Breast Cancer Res ; 22(1): 48, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32414394

ABSTRACT

BACKGROUND: The human epidermal growth factor receptor (HER) family, notably EGFR, is overexpressed in most triple-negative breast cancer (TNBC) cases and provides cancer cells with compensatory signals that greatly contribute to the survival and development of resistance in response to therapy. This study investigated the effects of Pan-HER (Symphogen, Ballerup, Denmark), a novel mixture of six monoclonal antibodies directed against members of the HER family EGFR, HER2, and HER3, in a preclinical trial of TNBC patient-derived xenografts (PDXs). METHODS: Fifteen low passage TNBC PDX tumor samples were transferred into the right mammary fat pad of mice for engraftment. When tumors reached an average size of 100-200 mm3, mice were randomized (n ≥ 6 per group) and treated following three 1-week cycles consisting of three times/week intraperitoneal (IP) injection of either formulation buffer (vehicle control) or Pan-HER (50 mg/kg). At the end of treatment, tumors were collected for Western blot, RNA, and immunohistochemistry analyses. RESULTS: All 15 TNBC PDXs were responsive to Pan-HER treatment, showing significant reductions in tumor growth consistent with Pan-HER-mediated tumor downmodulation of EGFR and HER3 protein levels and significantly decreased activation of associated HER family signaling pathways AKT and ERK. Tumor regression was observed in five of the models, which corresponded to those PDX tumor models with the highest level of HER family activation. CONCLUSIONS: The marked effect of Pan-HER in numerous HER family-dependent TNBC PDX models justifies further studies of Pan-HER in TNBC clinical trials as a potential therapeutic option.


Subject(s)
Antibodies, Monoclonal/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-3/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Animals , Cell Proliferation/drug effects , Disease Models, Animal , Drug Resistance, Neoplasm , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Humans , Mice , Molecular Targeted Therapy , Mutation , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/genetics , Receptor, ErbB-3/metabolism , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Tumor Cells, Cultured
5.
Hepatology ; 61(5): 1591-602, 2015 May.
Article in English | MEDLINE | ID: mdl-25529917

ABSTRACT

UNLABELLED: Sorafenib, a broad tyrosine kinase inhibitor, is the only approved systemic therapy for advanced hepatocellular carcinoma (HCC) but provides limited survival benefits. Recently, immunotherapy has emerged as a promising treatment strategy, but its role remains unclear in HCCs, which are associated with decreased cytotoxic CD8(+) T-lymphocyte infiltration in both murine and human tumors. Moreover, in mouse models after sorafenib treatment intratumoral hypoxia is increased and may fuel evasive resistance. Using orthotopic HCC models, we now show that increased hypoxia after sorafenib treatment promotes immunosuppression, characterized by increased intratumoral expression of the immune checkpoint inhibitor programmed death ligand-1 and accumulation of T-regulatory cells and M2-type macrophages. We also show that the recruitment of immunosuppressive cells is mediated in part by hypoxia-induced up-regulation of stromal cell-derived 1 alpha. Inhibition of the stromal cell-derived 1 alpha receptor (C-X-C receptor type 4 or CXCR4) using AMD3100 prevented the polarization toward an immunosuppressive microenvironment after sorafenib treatment, inhibited tumor growth, reduced lung metastasis, and improved survival. However, the combination of AMD3100 and sorafenib did not significantly change cytotoxic CD8(+) T-lymphocyte infiltration into HCC tumors and did not modify their activation status. In separate experiments, antibody blockade of the programmed death ligand-1 receptor programmed death receptor-1 (PD-1) showed antitumor effects in treatment-naive tumors in orthotopic (grafted and genetically engineered) models of HCC. However, anti-PD-1 antibody treatment had additional antitumor activity only when combined with sorafenib and AMD3100 and not when combined with sorafenib alone. CONCLUSION: Anti-PD-1 treatment can boost antitumor immune responses in HCC models; when used in combination with sorafenib, anti-PD-1 immunotherapy shows efficacy only with concomitant targeting of the hypoxic and immunosuppressive microenvironment with agents such as CXCR4 inhibitors.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/immunology , Immunotherapy/methods , Liver Neoplasms/drug therapy , Liver Neoplasms/immunology , Niacinamide/analogs & derivatives , Phenylurea Compounds/therapeutic use , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Receptors, CXCR4/antagonists & inhibitors , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Animals , Humans , Mice , Niacinamide/therapeutic use , Sorafenib
6.
Hum Mol Genet ; 21(13): 2973-90, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22492996

ABSTRACT

The purpose of this study was to investigate the protective effects of the mitochondria-targeted antioxidant catalase (MCAT) and lifespan extension in mice that express amyloid beta (Aß). Using immunoblotting and immunostaining analyses, we measured the production of full-length amyloid precursor protein (APP), soluble APPα, C-terminal fragments CTF99 and CTF83, monomeric and oligomeric Aß, Aß deposits and beta site amyloid precursor protein cleaving enzyme 1 (BACE1), in different stages of disease progression in MCAT/AßPP and AßPP mice. Using quantitative reverse transcriptase polymerase chain reaction and immunostaining analyses, we studied the expression of catalase, BACE1, the Alzheimer's disease (AD) markers, synaptophysin, APP, neprilysin, insulin-degrading enzyme and transthyretin in MCAT, AßPP, MCAT/AßPP and wild-type (WT) mice. Using the high pressure liquid chromatography analysis of 8-hydroxy-2-deoxyguanosine, we measured oxidative DNA damage in the cerebral cortical tissues from MCAT, AßPP, MCAT/AßPP and WT mice. We found that the AßPP transgenic mice that carried the human MCAT gene lived 5 months longer than did the AßPP mice. We also found that the overexpression of MCAT in the brain sections from the MCAT/AßPP transgenic mice significantly correlated with a reduction in the levels of full-length APP, CTF99, BACE1, Aß levels (40 and 42), Aß deposits and oxidative DNA damage relative to the brain sections from the AßPP mice. Interestingly, we found significantly increased levels of soluble APPα and CTF83 in the MCAT/AßPP mice, relative to the AßPP mice. These data provide direct evidence that oxidative stress plays a primary role in AD etiopathology and that in MCAT mice express Aß, MCAT prevents abnormal APP processing, reduces Aß levels and enhances Aß-degrading enzymes in mice at different ages, corresponding to different stages of disease progression. These findings indicate that mitochondria-targeted molecules may be an effective therapeutic approach to treat patients with AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/biosynthesis , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Aspartic Acid Endopeptidases/biosynthesis , Catalase/metabolism , Mitochondria/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Protein Precursor/biosynthesis , Animals , Brain/pathology , Catalase/genetics , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , DNA Damage/genetics , Disease Models, Animal , Female , Insulysin/biosynthesis , Insulysin/metabolism , Male , Mice , Mice, Transgenic , Neprilysin/biosynthesis , Neuroprotective Agents/metabolism , Oxidative Stress , Prealbumin/biosynthesis , RNA, Messenger/biosynthesis , Random Allocation , Synaptophysin/biosynthesis
7.
Biochim Biophys Acta ; 1822(5): 639-49, 2012 May.
Article in English | MEDLINE | ID: mdl-22037588

ABSTRACT

Synaptic pathology and mitochondrial oxidative damage are early events in Alzheimer's disease (AD) progression. Loss of synapses and synaptic damage are the best correlates of cognitive deficits found in AD patients. Recent research on amyloid beta (Aß) and mitochondria in AD revealed that Aß accumulates in synapses and synaptic mitochondria, leading to abnormal mitochondrial dynamics and synaptic degeneration in AD neurons. Further, recent studies using live-cell imaging and primary neurons from amyloid beta precursor protein (AßPP) transgenic mice revealed reduced mitochondrial mass, defective axonal transport of mitochondria and synaptic degeneration, indicating that Aß is responsible for mitochondrial and synaptic deficiencies. Tremendous progress has been made in studying antioxidant approaches in mouse models of AD and clinical trials of AD patients. This article highlights the recent developments made in Aß-induced abnormal mitochondrial dynamics, defective mitochondrial biogenesis, impaired axonal transport and synaptic deficiencies in AD. This article also focuses on mitochondrial approaches in treating AD, and also discusses latest research on mitochondria-targeted antioxidants in AD. This article is part of a Special Issue entitled: Antioxidants and Antioxidant Treatment in Disease.


Subject(s)
Alzheimer Disease/physiopathology , Antioxidants/therapeutic use , Mitochondria/drug effects , Synapses/pathology , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/metabolism , Animals , Antioxidants/pharmacology , Humans , Mice , Mice, Transgenic , Mitochondria/physiology
8.
Int J Environ Res Public Health ; 8(1): 203-21, 2011 01.
Article in English | MEDLINE | ID: mdl-21318024

ABSTRACT

The purpose of this study was to determine the neurotoxicity of two commonly used herbicides: picloram and triclopyr and the neuroprotective effects of the mitochondria-targeted antioxidant, SS31. Using mouse neuroblastoma (N2a) cells and primary neurons from C57BL/6 mice, we investigated the toxicity of these herbicides, and protective effects of SS1 peptide against picloram and triclopyr toxicity. We measured total RNA content, cell viability and mRNA expression of peroxiredoxins, neuroprotective genes, mitochondrial-encoded electron transport chain (ETC) genes in N2a cells treated with herbicides and SS31. Using primary neurons from C57BL/6 mice, neuronal survival was studied in neurons treated with herbicides, in neurons pretreated with SS31 plus treated with herbicides, neurons treated with SS31 alone, and untreated neurons. Significantly decreased total RNA content, and cell viability in N2a cells treated with picloram and triclopyr were found compared to untreated N2a cells. Decreased mRNA expression of neuroprotective genes, and ETC genes in cells treated with herbicides was found compared to untreated cells. Decreased mRNA expression of peroxiredoxins 1-6 in N2a cells treated with picloram was found, suggesting that picloram affects the antioxidant enzymes in N2a cells. Immunofluorescence analysis of primary neurons revealed that decreased neuronal branching and degenerating neurons in neurons treated with picloram and triclopyr. However, neurons pretreated with SS31 prevented degenerative process caused by herbicides. Based on these results, we propose that herbicides--picloram and triclopyr appear to damage neurons, and the SS31 peptide appears to protect neurons from herbicide toxicity.


Subject(s)
Antioxidants/physiology , Glycolates/toxicity , Herbicides/toxicity , Neurons/drug effects , Oligopeptides/physiology , Picloram/toxicity , Animals , Antioxidants/administration & dosage , Cell Line, Tumor , Cell Survival , Electron Transport Chain Complex Proteins/biosynthesis , Forkhead Box Protein O1 , Forkhead Transcription Factors/biosynthesis , Mice , Mice, Inbred C57BL , Neuroblastoma , Neurons/pathology , Neurons/physiology , Oligopeptides/administration & dosage , Oxidative Stress , Peroxiredoxins/biosynthesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , RNA/analysis , RNA, Messenger/biosynthesis , RNA, Messenger/drug effects , Receptors, N-Methyl-D-Aspartate/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/biosynthesis , Transcription Factors , Transcription, Genetic
9.
Brain Res Rev ; 67(1-2): 103-18, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21145355

ABSTRACT

The purpose of this article is to review the recent developments of abnormal mitochondrial dynamics, mitochondrial fragmentation, and neuronal damage in neurodegenerative diseases, including Alzheimer's, Parkinson's, Huntington's, and amyotrophic lateral sclerosis. The GTPase family of proteins, including fission proteins, dynamin related protein 1 (Drp1), mitochondrial fission 1 (Fis1), and fusion proteins (Mfn1, Mfn2 and Opa1) are essential to maintain mitochondrial fission and fusion balance, and to provide necessary adenosine triphosphate to neurons. Among these, Drp1 is involved in several important aspects of mitochondria, including shape, size, distribution, remodeling, and maintenance of mitochondria in mammalian cells. In addition, recent advancements in molecular, cellular, electron microscopy, and confocal imaging studies revealed that Drp1 is associated with several cellular functions, including mitochondrial and peroxisomal fragmentation, phosphorylation, SUMOylation, ubiquitination, and cell death. In the last two decades, tremendous progress has been made in researching mitochondrial dynamics, in yeast, worms, and mammalian cells; and this research has provided evidence linking Drp1 to neurodegenerative diseases. Researchers in the neurodegenerative disease field are beginning to recognize the possible involvement of Drp1 in causing mitochondrial fragmentation and abnormal mitochondrial dynamics in neurodegenerative diseases. This article summarizes research findings relating Drp1 to mitochondrial fission and fusion, in yeast, worms, and mammals. Based on findings from the Reddy laboratory and others', we propose that mutant proteins of neurodegenerative diseases, including AD, PD, HD, and ALS, interact with Drp1, activate mitochondrial fission machinery, fragment mitochondria excessively, and impair mitochondrial transport and mitochondrial dynamics, ultimately causing mitochondrial dysfunction and neuronal damage.


Subject(s)
GTP Phosphohydrolases/physiology , Microtubule-Associated Proteins/physiology , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Mitochondrial Proteins/physiology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Animals , Cell Respiration/genetics , Cell Respiration/physiology , Dynamins , GTP Phosphohydrolases/genetics , Humans , Microtubule-Associated Proteins/genetics , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Diseases/genetics , Mitochondrial Proteins/genetics , Models, Animal , Mutation , Neurodegenerative Diseases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...