Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters

Publication year range
1.
FASEB J ; 38(10): e23659, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38733301

ABSTRACT

HDAC3 inhibition has been shown to improve memory and reduce amyloid-ß (Aß) in Alzheimer's disease (AD) models, but the underlying mechanisms are unclear. We investigated the molecular effects of HDAC3 inhibition on AD pathology, using in vitro and ex vivo models of AD, based on our finding that HDAC3 expression is increased in AD brains. For this purpose, N2a mouse neuroblastoma cells as well as organotypic brain cultures (OBCSs) of 5XFAD and wild-type mice were incubated with various concentrations of the HDAC3 selective inhibitor RGFP966 (0.1-10 µM) for 24 h. Treatment with RGFP966 or HDAC3 knockdown in N2a cells was associated with an increase on amyloid precursor protein (APP) and mRNA expressions, without alterations in Aß42 secretion. In vitro chromatin immunoprecipitation analysis revealed enriched HDAC3 binding at APP promoter regions. The increase in APP expression was also detected in OBCSs from 5XFAD mice incubated with 1 µM RGFP966, without changes in Aß. In addition, HDAC3 inhibition resulted in a reduction of activated Iba-1-positive microglia and astrocytes in 5XFAD slices, which was not observed in OBCSs from wild-type mice. mRNA sequencing analysis revealed that HDAC3 inhibition modulated neuronal regenerative pathways related to neurogenesis, differentiation, axonogenesis, and dendritic spine density in OBCSs. Our findings highlight the complexity and diversity of the effects of HDAC3 inhibition on AD models and suggest that HDAC3 may have multiple roles in the regulation of APP expression and processing, as well as in the modulation of neuroinflammatory and neuroprotective genes.


Subject(s)
Alzheimer Disease , Amyloid beta-Protein Precursor , Histone Deacetylases , Animals , Mice , Acrylamides , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/genetics , Brain/metabolism , Brain/pathology , Cell Line, Tumor , Disease Models, Animal , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Mice, Inbred C57BL , Mice, Transgenic , Microglia/metabolism , Phenylenediamines/pharmacology
2.
Proc Natl Acad Sci U S A ; 119(32): e2116289119, 2022 08 09.
Article in English | MEDLINE | ID: mdl-35917342

ABSTRACT

Glioblastoma (GBM) is an aggressive malignant primary brain tumor with limited therapeutic options. We show that the angiotensin II (AngII) type 2 receptor (AT2R) is a therapeutic target for GBM and that AngII, endogenously produced in GBM cells, promotes proliferation through AT2R. We repurposed EMA401, an AT2R antagonist originally developed as a peripherally restricted analgesic, for GBM and showed that it inhibits the proliferation of AT2R-expressing GBM spheroids and blocks their invasiveness and angiogenic capacity. The crystal structure of AT2R bound to EMA401 was determined and revealed the receptor to be in an active-like conformation with helix-VIII blocking G-protein or ß-arrestin recruitment. The architecture and interactions of EMA401 in AT2R differ drastically from complexes of AT2R with other relevant compounds. To enhance central nervous system (CNS) penetration of EMA401, we exploited the crystal structure to design an angiopep-2-tethered EMA401 derivative, A3E. A3E exhibited enhanced CNS penetration, leading to reduced tumor volume, inhibition of proliferation, and increased levels of apoptosis in an orthotopic xenograft model of GBM.


Subject(s)
Angiotensin II Type 2 Receptor Blockers , Benzhydryl Compounds , Brain Neoplasms , Drug Repositioning , Glioblastoma , Isoquinolines , Receptor, Angiotensin, Type 2 , Analgesics/pharmacology , Angiotensin II/chemistry , Angiotensin II/pharmacology , Angiotensin II Type 2 Receptor Blockers/therapeutic use , Apoptosis , Benzhydryl Compounds/chemistry , Benzhydryl Compounds/pharmacology , Benzhydryl Compounds/therapeutic use , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Humans , Isoquinolines/chemistry , Isoquinolines/pharmacology , Isoquinolines/therapeutic use , Protein Conformation, alpha-Helical , Receptor, Angiotensin, Type 2/chemistry , Receptor, Angiotensin, Type 2/metabolism , Tumor Burden/drug effects
3.
Int J Cancer ; 154(7): 1298-1308, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38146864

ABSTRACT

Here, we describe a blood test for the detection of glial malignancies (GLI-M) based on the identification of circulating glial cells (CGCs). The test is highly specific for GLI-M and can detect multiple grades (II-IV) and subtypes including gliomas, astrocytomas, oligodendrogliomas, oligoastrocytomas and glioblastomas, irrespective of gender and age. Analytical validation of the test was performed as per Clinical and Laboratory Standards Institute (CLSI) guidelines. Real-world performance characteristics of the test were evaluated in four clinical (observational) studies. The test has high analytical sensitivity (95%), specificity (100%) and precision (coefficient of variation [CV] = 13.7% for repeatability and CV = 23.5% for within laboratory precision, both at the detection threshold) and is not prone to interference from common drugs and serum factors. The ability of the test to detect and differentiate GLI-M from non-malignant brain tumours (NBT), brain metastases from primary epithelial malignancies (EPI-M) and healthy individual donors (HD) was evaluated in four clinical cohorts. Across these clinical studies, the test showed 99.35% sensitivity (95% confidence interval [CI]: 96.44%-99.98%) and 100% specificity (95% CI: 99.37%-100%). The performance characteristics of this test support its clinical utility for diagnostic triaging of individuals presenting with intracranial space-occupying lesions (ICSOL).


Subject(s)
Astrocytoma , Brain Neoplasms , Glioma , Oligodendroglioma , Humans , Astrocytoma/diagnosis , Astrocytoma/pathology , Brain Neoplasms/diagnosis , Brain Neoplasms/pathology , Glioma/pathology , Neuroglia/pathology , Oligodendroglioma/diagnosis , Oligodendroglioma/pathology , Observational Studies as Topic
4.
Chemistry ; 26(47): 10690-10694, 2020 Aug 21.
Article in English | MEDLINE | ID: mdl-32691857

ABSTRACT

Mutating the side-chains of amino acids in a peptide ligand, with unnatural amino acids, aiming to mitigate its short half-life is an established approach. However, it is hypothesized that mutating specific backbone peptide bonds with bioisosters can be exploited not only to enhance the proteolytic stability of parent peptides, but also to tune its receptor subtype selectivity. Towards this end, four [Y]6 -Angiotensin II analogues are synthesized where amide bonds have been replaced by 1,4-disubstituted 1,2,3-triazole isosteres in four different backbone locations. All the analogues possessed enhanced stability in human plasma in comparison with the parent peptide, whereas only two of them achieved enhanced AT2 R/AT1 R subtype selectivity. This diversification has been studied through 2D NMR spectroscopy and unveiled a putative more structured microenvironment for the two selective ligands accompanied with increased number of NOE cross-peaks. The most potent analogue, compound 2, has been explored regarding its neurotrophic potential and resulted in an enhanced neurite growth with respect to the established agent C21.


Subject(s)
Angiotensin II/chemistry , Angiotensin II/metabolism , Mutation , Peptides/genetics , Receptors, Angiotensin/chemistry , Receptors, Angiotensin/metabolism , Amino Acids/genetics , Angiotensin II/genetics , Animals , HEK293 Cells , Humans , Ligands , Peptides/chemistry , Peptides/metabolism , Substrate Specificity
5.
BMC Cancer ; 18(1): 167, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29422017

ABSTRACT

BACKGROUND: Tumour cells have a high demand for arginine. However, a subset of glioblastomas has a defect in the arginine biosynthetic pathway due to epigenetic silencing of the rate limiting enzyme argininosuccinate synthetase (ASS1). These tumours are auxotrophic for arginine and susceptible to the arginine degrading enzyme, pegylated arginine deiminase (ADI-PEG20). Moreover, ASS1 deficient GBM have a worse prognosis compared to ASS1 positive tumours. Since altered tumour metabolism is one of the hallmarks of cancer we were interested to determine if these two subtypes exhibited different metabolic profiles that could allow for their non-invasive detection as well as unveil additional novel therapeutic opportunities. METHODS: We looked for basal metabolic differences using one and two-dimensional gas chromatography-time-of-flight mass spectrometry (1D/2D GC-TOFMS) followed by targeted analysis of 29 amino acids using liquid chromatography-time-of-flight mass spectrometry (LC-TOFMS). We also looked for differences upon arginine deprivation in a single ASS1 negative and positive cell line (SNB19 and U87 respectively). The acquired data was evaluated by chemometric based bioinformatic methods. RESULTS: Orthogonal partial least squares-discriminant analysis (OPLS-DA) of both the 1D and 2D GC-TOFMS data revealed significant systematic difference in metabolites between the two subgroups with ASS1 positive cells generally exhibiting an overall elevation of identified metabolites, including those involved in the arginine biosynthetic pathway. Pathway and network analysis of the metabolite profile show that ASS1 negative cells have altered arginine and citrulline metabolism as well as altered amino acid metabolism. As expected, we observed significant metabolite perturbations in ASS negative cells in response to ADI-PEG20 treatment. CONCLUSIONS: This study has highlighted significant differences in the metabolome of ASS1 negative and positive GBM which warrants further study to determine their diagnostic and therapeutic potential for the treatment of this devastating disease.


Subject(s)
Argininosuccinate Synthase/metabolism , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Metabolomics/methods , Cell Line, Tumor , Humans , Phenotype
6.
BMC Cancer ; 18(1): 268, 2018 03 08.
Article in English | MEDLINE | ID: mdl-29519248

ABSTRACT

In the original publication of this article [1], published on 8 February 2018, it was noticed that the acknowledgement of the source of the drug ADI-PEG20 was missing.

7.
J Proteome Res ; 15(1): 280-90, 2016 Jan 04.
Article in English | MEDLINE | ID: mdl-26629888

ABSTRACT

Altered metabolism in tumor cells is required for rapid proliferation but also can influence other phenotypes that affect clinical outcomes such as metastasis and sensitivity to chemotherapy. Here, a genome-wide association study (GWAS)-guided integration of NCI-60 transcriptome and metabolome data identified ecto-5'-nucleotidase (NT5E or CD73) as a major determinant of metabolic phenotypes in cancer cells. NT5E expression and associated metabolome variations were also correlated with sensitivity to several chemotherapeutics including platinum-based treatment. NT5E mRNA levels were observed to be elevated in cells upon in vitro and in vivo acquisition of platinum resistance in ovarian cancer cells, and specific targeting of NT5E increased tumor cell sensitivity to platinum. We observed that tumor NT5E levels were prognostic for outcomes in ovarian cancer and were elevated after treatment with platinum, supporting the translational relevance of our findings. In this work, we integrated and analyzed a plethora of public data, demonstating the merit of such a systems oncology approach for the discovery of novel players in cancer biology and therapy. We experimentally validated the main findings of the NT5E gene being involved in both intrinsic and acquired resistance to platinum-based drugs. We propose that the efficacy of conventional chemotherapy could be improved by NT5E inhibition and that NT5E expression may be a useful prognostic and predictive clinical biomarker.


Subject(s)
5'-Nucleotidase/metabolism , Antineoplastic Agents/pharmacology , Ovarian Neoplasms/metabolism , 5'-Nucleotidase/genetics , Carboplatin/pharmacology , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Expression , Humans , Organoplatinum Compounds/pharmacology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/mortality , Proportional Hazards Models , Systems Biology , Treatment Outcome
8.
Cancer Cell ; 13(2): 91-104, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18242510

ABSTRACT

Therapies that target estrogen signaling have transformed the treatment of breast cancer. However, the effectiveness of these agents is limited by the development of resistance. Here, an RNAi screen was used to identify modifiers of tamoxifen sensitivity. We demonstrate that CDK10 is an important determinant of resistance to endocrine therapies and show that CDK10 silencing increases ETS2-driven transcription of c-RAF, resulting in MAPK pathway activation and loss of tumor cell reliance upon estrogen signaling. Patients with ER alpha-positive tumors that express low levels of CDK10 relapse early on tamoxifen, demonstrating the clinical significance of these observations. The association of low levels of CDK10 with methylation of the CDK10 promoter suggests a mechanism by which CDK10 expression is reduced in tumors.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Cyclin-Dependent Kinases/metabolism , Drug Resistance, Neoplasm , Antineoplastic Agents, Hormonal/pharmacology , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Drug Resistance, Neoplasm/drug effects , Enzyme Activation/drug effects , Estrogen Receptor alpha/metabolism , Estrogens/deficiency , G1 Phase/drug effects , Gene Silencing/drug effects , Humans , Ligands , Mitogen-Activated Protein Kinase 1/metabolism , Proto-Oncogene Proteins c-raf/genetics , RNA, Small Interfering/metabolism , Repressor Proteins/metabolism , Reproducibility of Results , Signal Transduction/drug effects , Survival Analysis , Tamoxifen/pharmacology , Tamoxifen/therapeutic use , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Transfection
9.
Mol Cancer ; 14: 110, 2015 Jun 03.
Article in English | MEDLINE | ID: mdl-26037383

ABSTRACT

BACKGROUND: Gene therapy has been an attractive paradigm for cancer treatment. However, cancer gene therapy has been challenged by the inherent limitation of vectors that are able to deliver therapeutic genes to tumors specifically and efficiently following systemic administration. Bacteriophage (phage) are viruses that have shown promise for targeted systemic gene delivery. Yet, they are considered poor vectors for gene transfer. Recently, we generated a tumor-targeted phage named adeno-associated virus/phage (AAVP), which is a filamentous phage particle whose genome contains the adeno-associated virus genome. Its effectiveness in delivering therapeutic genes to tumors specifically both in vitro and in vivo has been shown in numerous studies. Despite being a clinically useful vector, a multitude of barriers impede gene transduction to tumor cells. We hypothesized that one such factor is the tumor extracellular matrix (ECM). METHODS: We used a number of tumor cell lines from different species and histological types in 2D monolayers or 3D multicellular tumor spheroid (MCTS) models. To assess whether the ECM is a barrier to tumor cell targeting by AAVP, we depleted the ECM using collagenase, hyaluronidase, or combination of both. We employed multiple techniques to investigate and quantify the effect of ECM depletion on ECM composition (including collagen type I, hyaluronic acid, fibronectin and laminin), and how AAVP adsorption, internalisation, gene expression and therapeutic efficacy are subsequently affected. Data were analyzed using a student's t test when comparing two groups or one-way ANOVA and post hoc Tukey tests when using more than two groups. RESULTS: We demonstrate that collagenase and hyaluronidase-mediated degradation of tumor ECM affects the composition of collagen, hyaluronic acid and fibronectin. Consequently, AAVP diffusion, internalisation, gene expression and tumor cell killing were enhanced after enzymatic treatment. Our data suggest that enhancement of gene transfer by the AAVP is solely attributed to ECM depletion. We provide substantial evidence that ECM modulation is relevant in clinically applicable settings by using 3D MCTS, which simulates in vivo environments more accurately. CONCLUSION: Our findings suggest that ECM depletion is an effective strategy to enhance the efficiency of viral vector-guided gene therapy.


Subject(s)
Bacteriophages/metabolism , Extracellular Matrix/metabolism , Genetic Vectors/metabolism , Neoplasms/metabolism , Animals , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Collagenases/pharmacology , Dependovirus , Diffusion , Endocytosis , Enzyme-Linked Immunosorbent Assay , Extracellular Matrix/drug effects , Fibronectins/metabolism , Ganciclovir/pharmacology , Gene Expression/drug effects , Humans , Hyaluronoglucosaminidase/pharmacology , Integrin alphaV/metabolism , Laminin/metabolism , Losartan/pharmacology , Neoplasms/pathology , Rats , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Transduction, Genetic
10.
Nat Genet ; 38(10): 1133-41, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16964264

ABSTRACT

iASPP is one of the most evolutionarily conserved inhibitors of p53, whereas ASPP1 and ASPP2 are activators of p53. We show here that, in addition to the DNA-binding domain, the ASPP family members also bind to the proline-rich region of p53, which contains the most common p53 polymorphism at codon 72. Furthermore, the ASPP family members, particularly iASPP, bind to and regulate the activity of p53Pro72 more efficiently than that of p53Arg72. Hence, escape from negative regulation by iASPP is a newly identified mechanism by which p53Arg72 activates apoptosis more efficiently than p53Pro72.


Subject(s)
Apoptosis/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Polymorphism, Genetic , Proline/metabolism , Tumor Suppressor Protein p53/genetics , Amino Acid Sequence , Arginine , Binding Sites , Breast Neoplasms/genetics , Carcinoma/genetics , Cells, Cultured , Codon , Conserved Sequence , Female , Gene Expression Regulation , Homozygote , Humans , Intracellular Signaling Peptides and Proteins/genetics , Molecular Sequence Data , Repressor Proteins , Tumor Suppressor Protein p53/metabolism , Tyrosine/metabolism
11.
Cancers (Basel) ; 16(11)2024 May 29.
Article in English | MEDLINE | ID: mdl-38893173

ABSTRACT

Glioblastoma (GBM) cells require high levels of nicotinamide adenine dinucleotide (NAD) to fuel metabolic reactions, regulate their cell cycle and support DNA repair in response to chemotherapy and radiation. Inhibition of a key enzyme in NAD biosynthesis, NAMPT, has demonstrated significant anti-neoplastic activity. Here, we sought to characterise NAD biosynthetic pathways in GBM to determine resistance mechanisms to NAD inhibitors. GBM cells were treated with the NAMPT inhibitor FK866 with and without NAD precursors, and were analysed by qPCR, Western blot and proliferation assays (monolayer and spheroid). We also measured changes in the cell cycle, apoptosis, NAD/NADH levels and energy production. We performed orthoptic xenograft experiments in athymic nude mice to test the efficacy of FK866 in combination with temozolomide (TMZ). We show that the expression of key genes involved in NAD biosynthesis is highly variable across GBM tumours. FK866 inhibits proliferation, reduces NAD levels and limits oxidative metabolism, leading to G2/M cell cycle arrest; however, this can be reversed by supplementation with specific NAD precursors. Furthermore, FK866 potentiates the effects of radiation and TMZ in vitro and in vivo. NAMPT inhibitors should be considered for the treatment of GBM, with patients stratified based on their expression of key enzymes in other NAD biosynthetic pathways.

12.
Nature ; 448(7157): 1063-7, 2007 Aug 30.
Article in English | MEDLINE | ID: mdl-17728759

ABSTRACT

The acetyl-transferase Tip60 might influence tumorigenesis in multiple ways. First, Tip60 is a co-regulator of transcription factors that either promote or suppress tumorigenesis, such as Myc and p53. Second, Tip60 modulates DNA-damage response (DDR) signalling, and a DDR triggered by oncogenes can counteract tumour progression. Using E(mu)-myc transgenic mice that are heterozygous for a Tip60 gene (Htatip) knockout allele (hereafter denoted as Tip60+/- mice), we show that Tip60 counteracts Myc-induced lymphomagenesis in a haplo-insufficient manner and in a time window that is restricted to a pre- or early-tumoral stage. Tip60 heterozygosity severely impaired the Myc-induced DDR but caused no general DDR defect in B cells. Myc- and p53-dependent transcription were not affected, and neither were Myc-induced proliferation, activation of the ARF-p53 tumour suppressor pathway or the resulting apoptotic response. We found that the human TIP60 gene (HTATIP) is a frequent target for mono-allelic loss in human lymphomas and head-and-neck and mammary carcinomas, with concomitant reduction in mRNA levels. Immunohistochemical analysis also demonstrated loss of nuclear TIP60 staining in mammary carcinomas. These events correlated with disease grade and frequently concurred with mutation of p53. Thus, in both mouse and human, Tip60 has a haplo-insufficient tumour suppressor activity that is independent from-but not contradictory with-its role within the ARF-p53 pathway. We suggest that this is because critical levels of Tip60 are required for mounting an oncogene-induced DDR in incipient tumour cells, the failure of which might synergize with p53 mutation towards tumour progression.


Subject(s)
DNA Damage , Histone Acetyltransferases/metabolism , Oncogene Protein p55(v-myc)/metabolism , Oncogenes/genetics , Tumor Suppressor Proteins/metabolism , Alleles , Animals , B-Lymphocytes/metabolism , Carcinoma/genetics , Carcinoma/pathology , Cells, Cultured , Genes, Tumor Suppressor , Genes, myc/genetics , Heterozygote , Histone Acetyltransferases/genetics , Homeostasis , Humans , Lymphoma/genetics , Lymphoma/pathology , Lysine Acetyltransferase 5 , Mice , Mice, Transgenic , Oncogene Protein p55(v-myc)/genetics , Trans-Activators , Transcription, Genetic/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics
13.
Cell Death Discov ; 9(1): 172, 2023 May 19.
Article in English | MEDLINE | ID: mdl-37202382

ABSTRACT

Glioblastomas are a highly aggressive cancer type which respond poorly to current pharmaceutical treatments, thus novel therapeutic approaches need to be investigated. One such approach involves the use of the bioactive natural product Tanshinone IIA (T2A) derived from the Chinese herb Danshen, where mechanistic insight for this anti-cancer agent is needed to validate its use. Here, we employ a tractable model system, Dictyostelium discoideum, to provide this insight. T2A potently inhibits cellular proliferation of Dictyostelium, suggesting molecular targets in this model. We show that T2A rapidly reduces phosphoinositide 3 kinase (PI3K) and protein kinase B (PKB) activity, but surprisingly, the downstream complex mechanistic target of rapamycin complex 1 (mTORC1) is only inhibited following chronic treatment. Investigating regulators of mTORC1, including PKB, tuberous sclerosis complex (TSC), and AMP-activated protein kinase (AMPK), suggests these enzymes were not responsible for this effect, implicating an additional molecular mechanism of T2A. We identify this mechanism as the increased expression of sestrin, a negative regulator of mTORC1. We further show that combinatory treatment using a PI3K inhibitor and T2A gives rise to a synergistic inhibition of cell proliferation. We then translate our findings to human and mouse-derived glioblastoma cell lines, where both a PI3K inhibitor (Paxalisib) and T2A reduces glioblastoma proliferation in monolayer cultures and in spheroid expansion, with combinatory treatment significantly enhancing this effect. Thus, we propose a new approach for cancer treatment, including glioblastomas, through combinatory treatment with PI3K inhibitors and T2A.

14.
Cancer Med ; 12(8): 9116-9127, 2023 04.
Article in English | MEDLINE | ID: mdl-36718027

ABSTRACT

BACKGROUND: The low specificity of serum PSA resulting in the inability to effectively differentiate prostate cancer from benign prostate conditions is a persistent clinical challenge. The low sensitivity of serum PSA results in false negatives and can miss high-grade prostate cancers. We describe a non-invasive test for detection of prostate cancer based on functional enrichment of prostate adenocarcinoma associated circulating tumor cells (PrAD-CTCs) from blood samples followed by their identification by immunostaining for pan-cytokeratins (PanCK), prostate specific membrane antigen (PSMA), alpha methyl-acyl coenzyme-A racemase (AMACR), epithelial cell adhesion molecule (EpCAM), and common leucocyte antigen (CD45). METHODS: Analytical validation studies were performed to establish the performance characteristics of the test using VCaP prostate cancer cells spiked into healthy donor blood (HDB). The clinical performance characteristics of the test were evaluated in a case-control study with 160 known prostate cancer cases and 800 healthy males, followed by a prospective clinical study of 210 suspected cases of prostate cancer. RESULTS: Analytical validation established analyte stability as well as acceptable performance characteristics. The test showed 100% specificity and 100% sensitivity to differentiate prostate cancer cases from healthy individuals in the case control study and 91.2% sensitivity and 100% specificity to differentiate prostate cancers from benign prostate conditions in the prospective clinical study. CONCLUSIONS: The test accurately detects PrAD-CTCs with high sensitivity and specificity irrespective of stage, serum PSA or Gleason score, which translates into low risks of false negatives or overdiagnosis. The high accuracy of the test could offer advantages over PSA based prostate cancer detection.


Subject(s)
Neoplastic Cells, Circulating , Prostatic Neoplasms , Male , Humans , Prostate-Specific Antigen , Prostate/pathology , Case-Control Studies , Prospective Studies , Prostatic Neoplasms/pathology , Biomarkers, Tumor
15.
Cancer Cell ; 3(4): 387-402, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12726864

ABSTRACT

Intact p73 function is shown to be an important determinant of cellular sensitivity to anticancer agents. Inhibition of p73 function by dominant-negative proteins or by mutant p53 abrogates apoptosis and cytotoxicity induced by these agents. A polymorphism encoding either arginine (72R) or proline (72P) at codon 72 of p53 influences inhibition of p73 by a range of p53 mutants identified in squamous cancers. Clinical response following cisplatin-based chemo-radiotherapy for advanced head and neck cancer is influenced by this polymorphism, cancers expressing 72R mutants having lower response rates than those expressing 72P mutants. Polymorphism in p53 may influence individual responsiveness to cancer therapy.


Subject(s)
Apoptosis/genetics , Carcinoma, Squamous Cell/genetics , DNA-Binding Proteins/physiology , Drug Resistance, Neoplasm/genetics , Genes, p53/physiology , Nuclear Proteins/physiology , Adult , Aged , Drug Therapy , Female , Genes, Tumor Suppressor , Head and Neck Neoplasms/genetics , Humans , Immunoblotting , Immunohistochemistry , Male , Middle Aged , Mutation , Plasmids , Polymorphism, Single Nucleotide , Prognosis , RNA, Small Interfering/metabolism , Tumor Cells, Cultured , Tumor Protein p73 , Tumor Suppressor Proteins
16.
Pharmaceutics ; 14(2)2022 Feb 04.
Article in English | MEDLINE | ID: mdl-35214094

ABSTRACT

The unmet need to develop novel approaches for cancer diagnosis and treatment has led to the evolution of theranostic agents, which usually include, in addition to the anticancer drug, an imaging agent based mostly on fluorescent agents. Over the past few years, a non-invasive photoacoustic imaging modality has been effectively integrated into theranostic agents. Herein, we shed light on the design principles governing the development of theranostic agents with photoacoustic properties, which can be formulated into nanocarriers to enhance their potency. Specifically, we provide an extensive analysis of their individual constituents including the imaging dyes, drugs, linkers, targeting moieties, and their formulation into nanocarriers. Along these lines, we present numerous relevant paradigms. Finally, we discuss the clinical relevance of the specific strategy, as also the limitations and future perspectives, and through this review, we envisage paving the way for the development of theranostic agents endowed with photoacoustic properties as effective anticancer medicines.

17.
Biomolecules ; 12(10)2022 10 11.
Article in English | MEDLINE | ID: mdl-36291666

ABSTRACT

There is compelling evidence that head injury is a significant environmental risk factor for Alzheimer's disease (AD) and that a history of traumatic brain injury (TBI) accelerates the onset of AD. Amyloid-ß plaques and tau aggregates have been observed in the post-mortem brains of TBI patients; however, the mechanisms leading to AD neuropathology in TBI are still unknown. In this study, we hypothesized that focal TBI induces changes in miRNA expression in and around affected areas, resulting in the altered expression of genes involved in neurodegeneration and AD pathology. For this purpose, we performed a miRNA array in extracts from rats subjected to experimental TBI, using the controlled cortical impact (CCI) model. In and around the contusion, we observed alterations of miRNAs associated with dementia/AD, compared to the contralateral side. Specifically, the expression of miR-9 was significantly upregulated, while miR-29b, miR-34a, miR-106b, miR-181a and miR-107 were downregulated. Via qPCR, we confirmed these results in an additional group of injured rats when compared to naïve animals. Interestingly, the changes in those miRNAs were concomitant with alterations in the gene expression of mRNAs involved in amyloid generation and tau pathology, such as ß-APP cleaving enzyme (BACE1) and Glycogen synthase-3-ß (GSK3ß). In addition increased levels of neuroinflammatory markers (TNF-α), glial activation, neuronal loss, and tau phosphorylation were observed in pericontusional areas. Therefore, our results suggest that the secondary injury cascade in TBI affects miRNAs regulating the expression of genes involved in AD dementia.


Subject(s)
Alzheimer Disease , Brain Injuries, Traumatic , Contusions , MicroRNAs , Animals , Rats , Amyloid Precursor Protein Secretases/metabolism , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Tumor Necrosis Factor-alpha/metabolism , Glycogen Synthase/metabolism , Aspartic Acid Endopeptidases/genetics , Brain Injuries, Traumatic/metabolism , Amyloid beta-Peptides/metabolism , Alzheimer Disease/metabolism , MicroRNAs/metabolism , Plaque, Amyloid/complications , Plaque, Amyloid/metabolism , Brain/metabolism , Contusions/complications , Contusions/metabolism
18.
J Clin Invest ; 132(6)2022 03 15.
Article in English | MEDLINE | ID: mdl-35113813

ABSTRACT

New approaches for the management of glioblastoma (GBM) are an urgent and unmet clinical need. Here, we illustrate that the efficacy of radiotherapy for GBM is strikingly potentiated by concomitant therapy with the arginine-depleting agent ADI-PEG20 in a non-arginine-auxotrophic cellular background (argininosuccinate synthetase 1 positive). Moreover, this combination led to durable and complete radiological and pathological response, with extended disease-free survival in an orthotopic immune-competent model of GBM, with no significant toxicity. ADI-PEG20 not only enhanced the cellular sensitivity of argininosuccinate synthetase 1-positive GBM to ionizing radiation by elevated production of nitric oxide (˙NO) and hence generation of cytotoxic peroxynitrites, but also promoted glioma-associated macrophage/microglial infiltration into tumors and turned their classical antiinflammatory (protumor) phenotype into a proinflammatory (antitumor) phenotype. Our results provide an effective, well-tolerated, and simple strategy to improve GBM treatment that merits consideration for early evaluation in clinical trials.


Subject(s)
Antineoplastic Agents , Glioblastoma , Antineoplastic Agents/therapeutic use , Arginine , Argininosuccinate Synthase/genetics , Cell Line, Tumor , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Humans , Hydrolases , Microglia , Polyethylene Glycols
19.
Ann Hematol ; 90(9): 1037-45, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21340720

ABSTRACT

Polo-like kinase 2 (SNK/PLK2), a transcriptional target for wild-type p53 and is hypermethylated in a high percentage of multiple myeloma and B cell lymphomas patients. Given these data, we sought to study the methylation status of the specific gene in acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS), and to correlate it with clinical and genetic features. Using methylation-specific PCR MSP, we analyzed the methylation profile of 45 cases of AML and 43 cases of MDS. We also studied the distribution of MTHFR A1298C and MTHFR C677T polymorphisms and FLT3 mutations in AML patients and correlated the results with hypermethylation in the SNK/PLK2 CpG island. The SNK/PLK2 CpG island was hypermethylated in 68.9% and 88.4% of AML and MDS cases, respectively. Cases with hypermethylation had a trend towards more favorable overall survival (OS). There was no association between different MTHFR genotypes and susceptibility to develop AML. SNK/PLK2 hypermethylation combined with the MTHFR AA1298 genotype was associated with a tendency for a better OS. Similarly, patients with SNK/PLK2 hypermethylation combined with the MTHFR CT677 polymorphism had a better OS (HR = 0.34; p = 0.017). SNK/PLK2 methylation associated with unmutated FLT3 cases had a trend for better OS compared to patients with mutated FLT3 gene. SNK/PLK2 is a novel epigenetically regulated gene in AML and MDS, and methylation occurs at high frequency in both diseases. As such, SNK/PLK2 could represent a potential pathogenetic factor, although additional studies are necessary to verify its exact role in disease pathogenesis.


Subject(s)
Epigenesis, Genetic/physiology , Epistasis, Genetic/physiology , Leukemia, Myeloid, Acute/genetics , Myelodysplastic Syndromes/genetics , Protein Serine-Threonine Kinases/physiology , Adolescent , Adult , Aged , Aged, 80 and over , CpG Islands/genetics , DNA Methylation , Female , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Young Adult
20.
J Neurol Surg Rep ; 82(2): e11-e16, 2021 Jan.
Article in English | MEDLINE | ID: mdl-34141517

ABSTRACT

We present a case report of a 51-year-old left-handed male with a background of human papillomovairus 16-positive tonsil squamous cell carcinoma presenting with tonic-clonic seizure and a radiological diagnosis of secondary metastatic deposits. These were initially treated with stereotactic radiosurgery and subsequently with surgery. Surgical resection was performed under general anesthesia with right-sided temporal and parietal approaches. Both the parietal and temporal deposits were removed, while the intraventricular mass was intentionally left to avoid postoperative deficits. Adjuvant radiotherapy and chemotherapy were administered postoperatively. The patient experienced a satisfactory recovery postoperatively and was reoperated for recurrence 4 months later. He maintained a good quality of life and an excellent performance status throughout, but unfortunately he passed away in November 2018 due to septic complications. This case history stresses the difficulty in managing squamous cell carcinomas (SCC) with brain metastatic deposits. There are no current guidelines about the management of patients presenting with such a rare condition. More data are thus desirable to better define treatment guidelines and protocols when SCC brain metastases are present.

SELECTION OF CITATIONS
SEARCH DETAIL