Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Int J Mol Sci ; 25(10)2024 May 10.
Article in English | MEDLINE | ID: mdl-38791253

ABSTRACT

The application of metal-based nanoparticles (mNPs) in cancer therapy and diagnostics (theranostics) has been a hot research topic since the early days of nanotechnology, becoming even more relevant in recent years. However, the clinical translation of this technology has been notably poor, with one of the main reasons being a lack of understanding of the disease and conceptual errors in the design of mNPs. Strikingly, throughout the reported studies to date on in vivo experiments, the concepts of "tumor targeting" and "tumor cell targeting" are often intertwined, particularly in the context of active targeting. These misconceptions may lead to design flaws, resulting in failed theranostic strategies. In the context of mNPs, tumor targeting can be described as the process by which mNPs reach the tumor mass (as a tissue), while tumor cell targeting refers to the specific interaction of mNPs with tumor cells once they have reached the tumor tissue. In this review, we conduct a critical analysis of key challenges that must be addressed for the successful targeting of either tumor tissue or cancer cells within the tumor tissue. Additionally, we explore essential features necessary for the smart design of theranostic mNPs, where 'smart design' refers to the process involving advanced consideration of the physicochemical features of the mNPs, targeting motifs, and physiological barriers that must be overcome for successful tumor targeting and/or tumor cell targeting.


Subject(s)
Metal Nanoparticles , Neoplasms , Theranostic Nanomedicine , Humans , Neoplasms/therapy , Neoplasms/drug therapy , Neoplasms/diagnosis , Neoplasms/pathology , Theranostic Nanomedicine/methods , Animals , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Drug Delivery Systems/methods
2.
EBioMedicine ; 90: 104484, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36907105

ABSTRACT

BACKGROUND: Glioblastoma is one of the most devastating and incurable cancers due to its aggressive behaviour and lack of available therapies, being its overall-survival from diagnosis ∼14-months. Thus, identification of new therapeutic tools is urgently needed. Interestingly, metabolism-related drugs (e.g., metformin/statins) are emerging as efficient antitumour agents for several cancers. Herein, we evaluated the in vitro/in vivo effects of metformin and/or statins on key clinical/functional/molecular/signalling parameters in glioblastoma patients/cells. METHODS: An exploratory-observational-randomized retrospective glioblastoma patient cohort (n = 85), human glioblastoma/non-tumour brain human cells (cell lines/patient-derived cell cultures), mouse astrocytes progenitor cell cultures, and a preclinical xenograft glioblastoma mouse model were used to measure key functional parameters, signalling-pathways and/or antitumour progression in response to metformin and/or simvastatin. FINDINGS: Metformin and simvastatin exerted strong antitumour actions in glioblastoma cell cultures (i.e., proliferation/migration/tumoursphere/colony-formation/VEGF-secretion inhibition and apoptosis/senescence induction). Notably, their combination additively altered these functional parameters vs. individual treatments. These actions were mediated by the modulation of key oncogenic signalling-pathways (i.e., AKT/JAK-STAT/NF-κB/TGFß-pathways). Interestingly, an enrichment analysis uncovered a TGFß-pathway activation, together with AKT inactivation, in response to metformin + simvastatin combination, which might be linked to an induction of the senescence-state, the associated secretory-phenotype, and to the dysregulation of spliceosome components. Remarkably, the antitumour actions of metformin + simvastatin combination were also observed in vivo [i.e., association with longer overall-survival in human, and reduction in tumour-progression in a mouse model (reduced tumour-size/weight/mitosis-number, and increased apoptosis)]. INTERPRETATION: Altogether, metformin and simvastatin reduce aggressiveness features in glioblastomas, being this effect significantly more effective (in vitro/in vivo) when both drugs are combined, offering a clinically relevant opportunity that should be tested for their use in humans. FUNDING: Spanish Ministry of Science, Innovation and Universities; Junta de Andalucía; CIBERobn (CIBER is an initiative of Instituto de Salud Carlos III, Spanish Ministry of Health, Social Services and Equality).


Subject(s)
Glioblastoma , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Metformin , Humans , Mice , Animals , Metformin/pharmacology , Metformin/therapeutic use , Glioblastoma/drug therapy , Glioblastoma/pathology , Proto-Oncogene Proteins c-akt , Simvastatin/pharmacology , Simvastatin/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Retrospective Studies , Transforming Growth Factor beta/pharmacology , Cell Line, Tumor , Cell Proliferation
3.
Laryngoscope Investig Otolaryngol ; 7(3): 684-691, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35734050

ABSTRACT

Background: Computer-assisted navigated piezoelectric surgery (CANPS) is a surgical technique that combines the surgical navigation with a piezoelectric device. This association multiplies the advantages of both technologies, taking the best of each one providing a synergistic association. Objective: To describe and assess the indications, advantages, disadvantages, and complications of this association of surgical techniques. Methods: CANPS was used in 32 patients. The clinical diagnosis was facial trauma, tumors, orthognathic surgeries, temporomandibular joint ankylosis, pathology of the frontal sinus, and alveolar distraction. Nineteen patients were men and 13 were women. Planning software iPlan 3.05 of Brainlab, and Elements of Brainlab were used for planning and the Kolibri and Kurve of Brainlab for surgical navigation. The piezoelectric device used was a "Vercelotti" type in all patients. Results: CAPNS could be performed successfully in all cases without complications and reduced the surgeon's uncertainty during the osteotomies. There is continuous control of the position of the surgical instrument. The use of the navigated piezoelectric device allowed the surgeon's uncertainty to be reduced during the performance of the osteotomies in depth, in poorly visible areas, with little access or reduced visibility. It also increases the safety of bone resections near important anatomical structures. Conclusions: CANPS combines the advantages of piezoelectric surgery and navigation. CANPS affords real-time control of the position of the cutting tip and allows semiburied approaches. CANPS allows surgery to be precise, safer, and minimally invasive.

4.
J Clin Endocrinol Metab ; 107(7): e2938-e2951, 2022 06 16.
Article in English | MEDLINE | ID: mdl-35312002

ABSTRACT

CONTEXT: Adrenocorticotropin (ACTH)-secreting pituitary tumors (ACTHomas) are associated with severe comorbidities and increased mortality. Current treatments mainly focus on remission and prevention of persistent disease and recurrence. However, there are still no useful biomarkers to accurately predict the clinical outcome after surgery, long-term remission, or disease relapse. OBJECTIVES: This work aimed to identify clinical, biochemical, and molecular markers for predicting long-term clinical outcome and remission in ACTHomas. METHODS: A retrospective multicenter study was performed with 60 ACTHomas patients diagnosed between 2004 and 2018 with at least 2 years' follow-up. Clinical/biochemical variables were evaluated yearly. Molecular expression profile of the somatostatin/ghrelin/dopamine regulatory systems components and of key pituitary factors and proliferation markers were evaluated in tumor samples after the first surgery. RESULTS: Clinical variables including tumor size, time until diagnosis/first surgery, serum prolactin, and postsurgery cortisol levels were associated with tumor remission and relapsed disease. The molecular markers analyzed were distinctly expressed in ACTHomas, with some components (ie, SSTR1, CRHR1, and MKI67) showing instructive associations with recurrence and/or remission. Notably, an integrative model including selected clinical variables (tumor size/postsurgery serum cortisol), and molecular markers (SSTR1/CRHR1) can accurately predict the clinical evolution and remission of patients with ACTHomas, generating a receiver operating characteristic curve with an area under the curve of 1 (P < .001). CONCLUSION: This study demonstrates that the combination of a set of clinical and molecular biomarkers in ACTHomas is able to accurately predict the clinical evolution and remission of patients. Consequently, the postsurgery molecular profile represents a valuable tool for clinical evaluation and follow-up of patients with ACTHomas.


Subject(s)
Pituitary ACTH Hypersecretion , Pituitary Diseases , Pituitary Neoplasms , Humans , Hydrocortisone , Pituitary ACTH Hypersecretion/diagnosis , Pituitary ACTH Hypersecretion/genetics , Pituitary ACTH Hypersecretion/surgery , Pituitary Gland/pathology , Pituitary Neoplasms/diagnosis , Pituitary Neoplasms/genetics , Pituitary Neoplasms/surgery , Recurrence , Remission Induction , Retrospective Studies , Treatment Outcome
5.
Int J Mol Sci ; 23(3)2022 Jan 20.
Article in English | MEDLINE | ID: mdl-35163067

ABSTRACT

Glioblastoma (GBM) is the most malignant and lethal brain tumor. Current standard treatment consists of surgery followed by radiotherapy/chemotherapy; however, this is only a palliative approach with a mean post-operative survival of scarcely ~12-15 months. Thus, the identification of novel therapeutic targets to treat this devastating pathology is urgently needed. In this context, the truncated splicing variant of the somatostatin receptor subtype 5 (sst5TMD4), which is produced by aberrant alternative splicing, has been demonstrated to be overexpressed and associated with increased aggressiveness features in several tumors. However, the presence, functional role, and associated molecular mechanisms of sst5TMD4 in GBM have not been yet explored. Therefore, we performed a comprehensive analysis to characterize the expression and pathophysiological role of sst5TMD4 in human GBM. sst5TMD4 was significantly overexpressed (at mRNA and protein levels) in human GBM tissue compared to non-tumor (control) brain tissue. Remarkably, sst5TMD4 expression was significantly associated with poor overall survival and recurrent tumors in GBM patients. Moreover, in vitro sst5TMD4 overexpression (by specific plasmid) increased, whereas sst5TMD4 silencing (by specific siRNA) decreased, key malignant features (i.e., proliferation and migration capacity) of GBM cells (U-87 MG/U-118 MG models). Furthermore, sst5TMD4 overexpression in GBM cells altered the activity of multiple key signaling pathways associated with tumor aggressiveness/progression (AKT/JAK-STAT/NF-κB/TGF-ß), and its silencing sensitized GBM cells to the antitumor effect of pasireotide (a somatostatin analog). Altogether, these results demonstrate that sst5TMD4 is overexpressed and associated with enhanced malignancy features in human GBMs and reveal its potential utility as a novel diagnostic/prognostic biomarker and putative therapeutic target in GBMs.


Subject(s)
Alternative Splicing , Brain Neoplasms/mortality , Drug Resistance, Neoplasm , Glioblastoma/mortality , Receptors, Somatostatin/genetics , Somatostatin/analogs & derivatives , Up-Regulation , Adult , Aged , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Survival , Female , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/genetics , Humans , Male , Middle Aged , Neoplasm Recurrence, Local , Prognosis , Signal Transduction
6.
J Exp Clin Cancer Res ; 41(1): 39, 2022 Jan 27.
Article in English | MEDLINE | ID: mdl-35086552

ABSTRACT

BACKGROUND: Glioblastoma is one of the most devastating cancer worldwide based on its locally aggressive behavior and because it cannot be cured by current therapies. Defects in alternative splicing process are frequent in cancer. Recently, we demonstrated that dysregulation of the spliceosome is directly associated with glioma development, progression, and aggressiveness. METHODS: Different human cohorts and a dataset from different glioma mouse models were analyzed to determine the mutation frequency as well as the gene and protein expression levels between tumor and control samples of the splicing-factor-3B-subunit-1 (SF3B1), an essential and druggable spliceosome component. SF3B1 expression was also explored at the single-cell level across all cell subpopulations and transcriptomic programs. The association of SF3B1 expression with relevant clinical data (e.g., overall survival) in different human cohorts was also analyzed. Different functional (proliferation/migration/tumorspheres and colonies formation/VEGF secretion/apoptosis) and mechanistic (gene expression/signaling pathways) assays were performed in three different glioblastomas cell models (human primary cultures and cell lines) in response to SF3B1 blockade (using pladienolide B treatment). Moreover, tumor progression and formation were monitored in response to SF3B1 blockade in two preclinical xenograft glioblastoma mouse models. RESULTS: Our data provide novel evidence demonstrating that the splicing-factor-3B-subunit-1 (SF3B1, an essential and druggable spliceosome component) is low-frequency mutated in human gliomas (~ 1 %) but widely overexpressed in glioblastoma compared with control samples from the different human cohorts and mouse models included in the present study, wherein SF3B1 levels are associated with key molecular and clinical features (e.g., overall survival, poor prognosis and/or drug resistance). Remarkably, in vitro and in vivo blockade of SF3B1 activity with pladienolide B drastically altered multiple glioblastoma pathophysiological processes (i.e., reduction in proliferation, migration, tumorspheres formation, VEGF secretion, tumor initiation and increased apoptosis) likely by suppressing AKT/mTOR/ß-catenin pathways, and an imbalance of BCL2L1 splicing. CONCLUSIONS: Together, we highlight SF3B1 as a potential diagnostic and prognostic biomarker and an efficient pharmacological target in glioblastoma, offering a clinically relevant opportunity worth to be explored in humans.


Subject(s)
Glioblastoma/genetics , Phosphoproteins/metabolism , RNA Splicing Factors/metabolism , TOR Serine-Threonine Kinases/metabolism , bcl-X Protein/metabolism , beta Catenin/metabolism , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Glioblastoma/mortality , Humans , Mice , Survival Analysis , Transfection , Xenograft Model Antitumor Assays
9.
Microsurgery ; 41(3): 250-257, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33332622

ABSTRACT

BACKGROUND: The possibility of harvesting the profunda artery perforator (PAP) flap in a chimeric configuration together with the innervated gracilis muscle may be a good option for dynamic reconstruction following total glossectomies. In this paper, we present a retrospective radiological study, which evaluates the presence and characteristics of the anatomical variation of the chimeric PAP - gracilis flap. METHODS: The study sample comprised 66 patients (132 legs), 38 men, and 28 women with an average age of 56 ± 2 years old, who underwent head and neck reconstruction with a free flap. Preoperative computed tomographic angiography (CTA) was used for morphologic analysis. We present a case report of a patient with a total glossectomy reconstructed with this flap. RESULTS: A perforator in the PAP area joining with the main pedicle of the gracilis muscle was found in 38 legs (28.8%). Mean length of the pedicle from the profunda femoral artery to the point where the perforator in the PAP area and the pedicle of the gracilis joined was 3.0 ± 0.3 cm. Differences in the existence of this vascular configuration were not significant comparing groups by sex, BMI, height, side or source vessel. Motion of the new tongue was documented clinically and with electromyography. The patient achieved an intelligible speech and normal diet. CONCLUSIONS: Preoperative evaluation is necessary to assess the existence of a perforator in the PAP area joining with the main pedicle of the gracilis muscle. This chimeric flap has been demonstrated useful for dynamic reconstruction of a patient with a total glossectomy.


Subject(s)
Gracilis Muscle , Perforator Flap , Angiography , Female , Femoral Artery , Humans , Male , Retrospective Studies
10.
Brain ; 143(11): 3273-3293, 2020 12 05.
Article in English | MEDLINE | ID: mdl-33141183

ABSTRACT

Glioblastomas remain the deadliest brain tumour, with a dismal ∼12-16-month survival from diagnosis. Therefore, identification of new diagnostic, prognostic and therapeutic tools to tackle glioblastomas is urgently needed. Emerging evidence indicates that the cellular machinery controlling the splicing process (spliceosome) is altered in tumours, leading to oncogenic splicing events associated with tumour progression and aggressiveness. Here, we identify for the first time a profound dysregulation in the expression of relevant spliceosome components and splicing factors (at mRNA and protein levels) in well characterized cohorts of human high-grade astrocytomas, mostly glioblastomas, compared to healthy brain control samples, being SRSF3, RBM22, PTBP1 and RBM3 able to perfectly discriminate between tumours and control samples, and between proneural-like or mesenchymal-like tumours versus control samples from different mouse models with gliomas. Results were confirmed in four additional and independent human cohorts. Silencing of SRSF3, RBM22, PTBP1 and RBM3 decreased aggressiveness parameters in vitro (e.g. proliferation, migration, tumorsphere-formation, etc.) and induced apoptosis, especially SRSF3. Remarkably, SRSF3 was correlated with patient survival and relevant tumour markers, and its silencing in vivo drastically decreased tumour development and progression, likely through a molecular/cellular mechanism involving PDGFRB and associated oncogenic signalling pathways (PI3K-AKT/ERK), which may also involve the distinct alteration of alternative splicing events of specific transcription factors controlling PDGFRB (i.e. TP73). Altogether, our results demonstrate a drastic splicing machinery-associated molecular dysregulation in glioblastomas, which could potentially be considered as a source of novel diagnostic and prognostic biomarkers as well as therapeutic targets for glioblastomas. Remarkably, SRSF3 is directly associated with glioblastoma development, progression, aggressiveness and patient survival and represents a novel potential therapeutic target to tackle this devastating pathology.


Subject(s)
Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , Glioblastoma/genetics , Serine-Arginine Splicing Factors/genetics , Alternative Splicing , Apoptosis , Biomarkers, Tumor/genetics , Brain Neoplasms/mortality , Cell Movement , Cell Proliferation , Gene Silencing , Glioblastoma/mortality , Humans , Neoplasm Invasiveness/genetics , Receptor, Platelet-Derived Growth Factor beta/genetics , Signal Transduction/genetics , Survival Analysis , Xenograft Model Antitumor Assays
11.
Neuroendocrinology ; 110(11-12): 1028-1041, 2020.
Article in English | MEDLINE | ID: mdl-31940630

ABSTRACT

INTRODUCTION: Pituitary neuroendocrine tumors (PitNETs), the most abundant of all intracranial tumors, entail severe comorbidities. First-line therapy is transsphenoidal surgery, but subsequent pharmacological therapy is often required. Unfortunately, many patients are/become unresponsive to available drugs (somatostatin analogues [SSAs]/dopamine agonists), underscoring the need for new therapies. Statins are well-known drugs commonly prescribed to treat hyperlipidemia/cardiovascular diseases, but can convey additional beneficial effects, including antitumor actions. The direct effects of statins on normal human pituitary or PitNETs are poorly known. Thus, we aimed to explore the direct effects of statins, especially simvastatin, on key functional parameters in normal and tumoral pituitary cells, and to evaluate the combined effects of simvastatin with metformin (MF) or SSAs. METHODS: Effects of statins in cell proliferation/viability, hormone secretion, and signaling pathways were evaluated in normal pituitary cells from a primate model (Papio anubis), tumor cells from corticotropinomas, somatotropinomas, nonfunctioning pituitary tumors, and PitNET cell-lines (AtT20/GH3-cells). RESULTS: All statins decreased AtT20-cell proliferation, simvastatin showing stronger effects. Indeed, simvastatin reduced cell viability and/or hormone secretion in all PitNETs subtypes and cell-lines, and ACTH/GH/PRL/FSH/LH secretion (but not expression), in primate cell cultures, by modulating MAPK/PI3K/mTOR pathways and expression of key receptors (GH-releasing hormone-receptor/ghrelin-R/Kiss1-R) regulating pituitary function. Addition of MF or SSAs did not enhance simvastatin antitumor effects. CONCLUSION: Our data reveal direct antitumor effects of simvastatin on PitNET-cells, paving the way to explore these compounds as a possible tool to treat PitNETs.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Neuroendocrine Tumors/drug therapy , Pituitary Gland/drug effects , Pituitary Neoplasms/drug therapy , Simvastatin/pharmacology , Adult , Aged , Animals , Cell Line, Tumor , Disease Models, Animal , Drug Therapy, Combination , Female , Humans , Hypoglycemic Agents/pharmacology , Male , Metformin/pharmacology , Mice , Middle Aged , Papio anubis , Rats , Somatostatin/pharmacology , Young Adult
12.
Neuroendocrinology ; 110(1-2): 70-82, 2020.
Article in English | MEDLINE | ID: mdl-31272096

ABSTRACT

BACKGROUND: Pituitary neuroendocrine tumors (PitNETs) represent approximately 15% of all intracranial tumors and usually are associated with severe comorbidities. Unfortunately, a relevant number of patients do not respond to currently available pharmacological treatments, that is, somatostatin analogs (SSAs) or dopamine-agonists (DA). Thus, novel, chimeric somatostatin/dopamine compounds (dopastatins) that could improve medical treatment of PitNETs have been designed. OBJECTIVE: This study aims to determine the direct therapeutic effects of a new-generation dopastatin, BIM-065, on primary cell cultures from different PitNETs subtypes. METHODS: Thirty-one PitNET-derived cell cultures (9 corticotropinomas, 9 somatotropinomas, 11 nonfunctioning pituitary adenomas [NFPAs], and 2 prolactinomas), were treated with BIM-065, and key functional endpoints were assessed (cell viability, apoptosis, hormone secretion, expression levels of key genes, free cytosolic [Ca2+]i dynamics, etc.). AtT-20 cell line was used to evaluate signaling pathways in response to BIM-065. RESULTS: This chimeric compound decreased cell viability in all corticotropinomas and somatotropinomas tested, but not in NFPAs. BIM-065 reduced ACTH, GH, chromogranin-A and PRL secretion, and increased apoptosis in corticotropinomas, somatotropinomas, and NFPAs. These effects were possibly mediated through modulation of pivotal signaling cascades like [Ca2+]i kinetic and Akt- or ERK1/2-phosphorylation. CONCLUSIONS: Our results unveil a robust antitumoral effect in vitro of the novel chimeric compound BIM-065 on the main PitNET subtypes, inform on the mechanisms involved, and suggest that BIM-065 could be an efficacious therapeutic option to be considered in the treatment of PitNETs.


Subject(s)
Dopamine Agents/pharmacology , Dopamine/analogs & derivatives , Neuroendocrine Tumors/drug therapy , Pituitary Neoplasms/drug therapy , Somatostatin/analogs & derivatives , Somatostatin/pharmacology , Dopamine/pharmacology , Humans , Somatostatin/analysis , Tumor Cells, Cultured
13.
Clin Cancer Res ; 26(4): 957-969, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31624102

ABSTRACT

PURPOSE: Somatostatin analogues (SSA) are efficacious and safe treatments for a variety of neuroendocrine tumors, especially pituitary neuroendocrine tumors (PitNET). Their therapeutic effects are mainly mediated by somatostatin receptors SST2 and SST5. Most SSAs, such as octreotide/lanreotide/pasireotide, are either nonselective or activate mainly SST2. However, nonfunctioning pituitary tumors (NFPTs), the most common PitNET type, mainly express SST3 and finding peptides that activate this particular somatostatin receptor has been very challenging. Therefore, the main objective of this study was to identify SST3-agonists and characterize their effects on experimental NFPT models. EXPERIMENTAL DESIGN: Binding to SSTs and cAMP level determinations were used to screen a peptide library and identify SST3-agonists. Key functional parameters (cell viability/caspase activity/chromogranin-A secretion/mRNA expression/intracellular signaling pathways) were assessed on NFPT primary cell cultures in response to SST3-agonists. Tumor growth was assessed in a preclinical PitNET mouse model treated with a SST3-agonist. RESULTS: We successfully identified the first SST3-agonist peptides. SST3-agonists lowered cell viability and chromogranin-A secretion, increased apoptosis in vitro, and reduced tumor growth in a preclinical PitNET model. As expected, inhibition of cell viability in response to SST3-agonists defined two NFPT populations: responsive and unresponsive, wherein responsive NFPTs expressed more SST3 than unresponsive NFPTs and exhibited a profound reduction of MAPK, PI3K-AKT/mTOR, and JAK/STAT signaling pathways upon SST3-agonist treatments. Concurrently, SSTR3 silencing increased cell viability in a subset of NFPTs. CONCLUSIONS: This study demonstrates that SST3-agonists activate signaling mechanisms that reduce NFPT cell viability and inhibit pituitary tumor growth in experimental models that expresses SST3, suggesting that targeting this receptor could be an efficacious treatment for NFPTs.


Subject(s)
Neuroendocrine Tumors/drug therapy , Peptides/pharmacology , Pituitary Neoplasms/drug therapy , Receptors, Somatostatin/agonists , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Humans , Janus Kinases/metabolism , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Knockout , Middle Aged , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Peptides/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/pathology , Signal Transduction , Tumor Cells, Cultured , Young Adult
14.
J Oral Maxillofac Surg ; 78(2): 284.e1-284.e4, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31705863

ABSTRACT

Several surgical procedures have been described to correct the deformities associated with craniosynostosis. To simplify the prediction of results, virtual planning techniques and image-guided surgery have been used. Digital planning can be transferred to the operating room using osteotomy and cutting guides or surgical navigation. We describe a novel bone fixation method that allows for anchoring of a cranial dynamic reference frame (DRF) in a steady manner. DRF can be used for registration and as a reference for surgical navigation in an infant's skull. We describe this novel technique to overcome the problems of DRF fixation on an infant's thin and weak calvarium. We fixed the DRF to the cranium using this new system. A 6-hole X-shaped miniplate was placed using 5 screws, leaving 1 of the central holes free. The self-drilling screw that anchors the DRF in position was placed in the free central hole, avoiding calvarial bone breakage and allowing for surgical navigation. To the best of our knowledge, the present study is the first report of this DRF anchorage modification for surgical navigation during surgery of craniosynostosis in an infant.


Subject(s)
Imaging, Three-Dimensional , Surgery, Computer-Assisted , Bone Screws , Skull , Tomography, X-Ray Computed
15.
Cancers (Basel) ; 11(10)2019 09 26.
Article in English | MEDLINE | ID: mdl-31561558

ABSTRACT

Pituitary neuroendocrine tumors (PitNETs) constitute approximately 15% of all brain tumors, and most have a sporadic origin. Recent studies suggest that altered alternative splicing and, consequently, appearance of aberrant splicing variants, is a common feature of most tumor pathologies. Moreover, spliceosome is considered an attractive therapeutic target in tumor pathologies, and the inhibition of SF3B1 (e.g., using pladienolide-B) has been shown to exert antitumor effects. Therefore, we aimed to analyze the expression levels of selected splicing-machinery components in 261 PitNETs (somatotropinomas/non-functioning PitNETS/corticotropinomas/prolactinomas) and evaluated the direct effects of pladienolide-B in cell proliferation/viability/hormone secretion in human PitNETs cell cultures and pituitary cell lines (AtT-20/GH3). Results revealed a severe dysregulation of splicing-machinery components in all the PitNET subtypes compared to normal pituitaries and a unique fingerprint of splicing-machinery components that accurately discriminate between normal and tumor tissue in each PitNET subtype. Moreover, expression of specific components was associated with key clinical parameters. Interestingly, certain components were commonly dysregulated throughout all PitNET subtypes. Finally, pladienolide-B reduced cell proliferation/viability/hormone secretion in PitNET cell cultures and cell lines. Altogether, our data demonstrate a drastic dysregulation of the splicing-machinery in PitNETs that might be associated to their tumorigenesis, paving the way to explore the use of specific splicing-machinery components as novel diagnostic/prognostic and therapeutic targets in PitNETs.

16.
Front Oncol ; 9: 328, 2019.
Article in English | MEDLINE | ID: mdl-31134147

ABSTRACT

Objective: We assess the efficacy of the metabolomic profile from glioma biopsies in providing estimates of postsurgical Overall Survival in glioma patients. Methods: Tumor biopsies from 46 patients bearing gliomas, obtained neurosurgically in the period 1992-1998, were analyzed by high resolution 1H magnetic resonance spectroscopy (HR- 1H MRS), following retrospectively individual postsurgical Overall Survival up to 720 weeks. Results: The Overall Survival profile could be resolved in three groups; Short (shorter than 52 weeks, n = 19), Intermediate (between 53 and 364 weeks, n = 19) or Long (longer than 365 weeks, n = 8), respectively. Classical histopathological analysis assigned WHO grades II-IV to every biopsy but notably, some patients with low grade glioma depicted unexpectedly Short Overall Survival, while some patients with high grade glioma, presented unpredictably Long Overall Survival. To explore the reasons underlying these different responses, we analyzed HR-1H MRS spectra from acid extracts of the same biopsies, to characterize the metabolite patterns associated to OS predictions. Poor prognosis was found in biopsies with higher contents of alanine, acetate, glutamate, total choline, phosphorylcholine, and glycine, while more favorable prognosis was achieved in biopsies with larger contents of total creatine, glycerol-phosphorylcholine, and myo-inositol. We then implemented a multivariate analysis to identify hierarchically the influence of metabolomic biomarkers on OS predictions, using a Classification Regression Tree (CRT) approach. The CRT based in metabolomic biomarkers grew up to three branches and split into eight nodes, predicting correctly the outcome of 94.7% of the patients in the Short Overall Survival group, 78.9% of the patients in the Intermediate Overall Survival group, and 75% of the patients in the Long Overall Survival group, respectively. Conclusion: Present results indicate that metabolic profiling by HR-1H MRS improves the Overall Survival predictions derived exclusively from classical histopathological gradings, thus favoring more precise therapeutic decisions.

17.
J Clin Endocrinol Metab ; 104(8): 3501-3513, 2019 08 01.
Article in English | MEDLINE | ID: mdl-30860580

ABSTRACT

CONTEXT: Pituitary neuroendocrine tumors (PitNETs) are a commonly underestimated pathology in terms of incidence and associated morbimortality. Currently, an appreciable subset of patients are resistant or poorly responsive to the main current medical treatments [i.e., synthetic somatostatin analogs (SSAs) and dopamine agonists]. Thus, development and optimization of novel and available medical therapies is necessary. Biguanides (metformin, buformin, and phenformin) are antidiabetic drugs that exert antitumoral actions in several tumor types, but their pharmacological effects on PitNETs are poorly known. OBJECTIVE: We aimed to explore the direct effects of biguanides on key functions (cell viability, hormone release, apoptosis, and signaling pathways) in primary cell cultures from human PitNETs and cell lines. Additionally, we evaluated the effect of combined metformin with SSAs on cell viability and hormone secretion. DESIGN: A total of 13 corticotropinomas, 13 somatotropinomas, 13 nonfunctioning PitNETs, 3 prolactinomas, and 2 tumoral pituitary cell lines (AtT-20 and GH3) were used to evaluate the direct effects of biguanides on cell viability, hormone release, apoptosis, and signaling pathways. RESULTS: Biguanides reduced cell viability in all PitNETs and cell lines (with phenformin being the most effective biguanide) and increased apoptosis in somatotropinomas. Moreover, buformin and phenformin, but not metformin, reduced hormone secretion in a cell type-specific manner. Combination metformin/SSA therapy did not increase SSA monotherapy effectiveness. Effects of biguanides on PitNETs could involve the modulation of AMP-activated protein kinase-dependent ([Ca2+]i, PI3K/Akt) and independent (MAPK) mechanisms. CONCLUSION: Altogether, our data unveil clear antitumoral effects of biguanides on PitNET cells, opening avenues to explore their potential as drugs to treat these pathologies.


Subject(s)
Antineoplastic Agents/pharmacology , Biguanides/pharmacology , Hypoglycemic Agents/pharmacology , Neuroendocrine Tumors/drug therapy , Pituitary Neoplasms/drug therapy , AMP-Activated Protein Kinases/drug effects , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Humans , Signal Transduction/drug effects
19.
J Clin Endocrinol Metab ; 99(2): E384-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24203067

ABSTRACT

RATIONALE: Metabolic syndrome (MetS) is a high-prevalence condition characterized by altered energy metabolism, insulin resistance, and elevated cardiovascular risk. OBJECTIVES: Although many individual single nucleotide polymorphisms (SNPs) have been linked to certain MetS features, there are few studies analyzing the influence of SNPs on carbohydrate metabolism in MetS. METHODS: A total of 904 SNPs (tag SNPs and functional SNPs) were tested for influence on 8 fasting and dynamic markers of carbohydrate metabolism, by performance of an intravenous glucose tolerance test in 450 participants in the LIPGENE study. FINDINGS: From 382 initial gene-phenotype associations between SNPs and any phenotypic variables, 61 (16% of the preselected variables) remained significant after bootstrapping. Top SNPs affecting glucose metabolism variables were as follows: fasting glucose, rs26125 (PPARGC1B); fasting insulin, rs4759277 (LRP1); C-peptide, rs4759277 (LRP1); homeostasis assessment of insulin resistance, rs4759277 (LRP1); quantitative insulin sensitivity check index, rs184003 (AGER); sensitivity index, rs7301876 (ABCC9), acute insulin response to glucose, rs290481 (TCF7L2); and disposition index, rs12691 (CEBPA). CONCLUSIONS: We describe here the top SNPs linked to phenotypic features in carbohydrate metabolism among approximately 1000 candidate gene variations in fasting and postprandial samples of 450 patients with MetS from the LIPGENE study.


Subject(s)
Carbohydrate Metabolism/genetics , Metabolic Syndrome/genetics , Adult , Aged , Alleles , Cardiovascular Diseases/genetics , Female , Genetic Association Studies , Genotype , Humans , Male , Metabolic Syndrome/metabolism , Middle Aged , Polymorphism, Single Nucleotide , Risk Factors
20.
J Neurol Surg A Cent Eur Neurosurg ; 74 Suppl 1: e155-8, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23427029

ABSTRACT

We present a patient with an otogenic cerebellar abscess caused by Enterococcus avium, a microorganism that is a rare cause of infection in humans. The patient experienced full recovery after early needle aspiration of the abscess and surgical treatment of the primary focus. Linezolid was selected as a first-line antimicrobial drug.


Subject(s)
Brain Abscess/microbiology , Cerebellar Diseases/microbiology , Enterococcus , Gram-Negative Bacterial Infections/microbiology , Acetamides/therapeutic use , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/therapeutic use , Brain Abscess/drug therapy , Cerebellar Diseases/drug therapy , Ciprofloxacin/therapeutic use , Female , Gram-Negative Bacterial Infections/drug therapy , Humans , Injections, Intravenous , Linezolid , Magnetic Resonance Imaging , Metronidazole/therapeutic use , Middle Aged , Nervous System Diseases/etiology , Oxazolidinones/therapeutic use , Tomography, X-Ray Computed
SELECTION OF CITATIONS
SEARCH DETAIL
...