15 results on '"Sepúlveda Sánchez JM"'
Search Results
2. NKG2C/ KLRC2 tumor cell expression enhances immunotherapeutic efficacy against glioblastoma.
- Author
-
de Dios O, Ramírez-González MA, Gómez-Soria I, Segura-Collar B, Manosalva J, Megías D, De Andrea CE, Fernández-Rubio L, Hernández-Laín A, Sepúlveda-Sánchez JM, Rodriguez-Ruiz ME, Pérez-Núñez Á, Wainwright DA, Gargini R, and Sánchez-Gómez P
- Subjects
- Humans, Mice, Animals, Brain Neoplasms immunology, Brain Neoplasms drug therapy, Brain Neoplasms metabolism, Cell Line, Tumor, Killer Cells, Natural immunology, Killer Cells, Natural metabolism, Female, Tumor Microenvironment, Glioblastoma immunology, Glioblastoma drug therapy, Glioblastoma metabolism, NK Cell Lectin-Like Receptor Subfamily C metabolism, Immunotherapy methods
- Abstract
Background: Activating and inhibitory receptors of natural killer (NK) cells such as NKp, NKG2, or CLEC are highly relevant to cold tumors including glioblastoma (GBM). Here, we aimed to characterize the expression of these receptors in GBM to gain insight into their potential role as modulators of the intratumoral microenvironment., Methods: We performed a transcriptomic analysis of several NK receptors with a focus on the activating receptor encoded by KLRC2, NKG2C, among bulk and single-cell RNA sequencing GBM data sets. We also evaluated the effects of KLRC2-overexpressing GL261 cells in mice treated with or without programmed cell death protein-1 (PD-1) monoclonal antibody (mAb). Finally, we analyzed samples from two clinical trials evaluating PD-1 mAb effects in patients with GBM to determine the potential of NKG2C to serve as a biomarker of response., Results: We observed significant expression of several inhibitory NK receptors on GBM-infiltrating NK and T cells, which contrasts with the strong expression of KLRC2 on tumor cells, mainly at the infiltrative margin. Neoplastic KLRC2 expression was associated with a reduction in the number of myeloid-derived suppressor cells and with a higher level of tumor-resident lymphocytes. A stronger antitumor activity after PD-1 mAb treatment was observed in NKG2C
high -expressing tumors both in mouse models and patients with GBM whereas the expression of inhibitory NK receptors showed an inverse association., Conclusions: This study explored the role of neoplastic NKG2C/ KLRC2 expression in shaping the immune profile of GBM and suggests that it is a predictive biomarker for positive responses to immune checkpoint inhibitor treatment in patients with GBM. Future studies could further validate this finding in prospective trials., Competing Interests: Competing interests: MER-R reports personal fees from BMS and grants from Highlight-Therapeutics and Roche outside the submitted work. She also has received speaker’s bureau honoraria from BMS and ROCHE. The rest of the authors declare that they have no competing interests., (© Author(s) (or their employer(s)) 2024. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2024
- Full Text
- View/download PDF
3. On optimal temozolomide scheduling for slowly growing glioblastomas.
- Author
-
Segura-Collar B, Jiménez-Sánchez J, Gargini R, Dragoj M, Sepúlveda-Sánchez JM, Pešić M, Ramírez MA, Ayala-Hernández LE, Sánchez-Gómez P, and Pérez-García VM
- Abstract
Background: Temozolomide (TMZ) is an oral alkylating agent active against gliomas with a favorable toxicity profile. It is part of the standard of care in the management of glioblastoma (GBM), and is commonly used in low-grade gliomas (LGG). In-silico mathematical models can potentially be used to personalize treatments and to accelerate the discovery of optimal drug delivery schemes., Methods: Agent-based mathematical models fed with either mouse or patient data were developed for the in-silico studies. The experimental test beds used to confirm the results were: mouse glioma models obtained by retroviral expression of EGFR-wt/EGFR-vIII in primary progenitors from p16/p19 ko mice and grown in-vitro and in-vivo in orthotopic allografts, and human GBM U251 cells immobilized in alginate microfibers. The patient data used to parametrize the model were obtained from the TCGA/TCIA databases and the TOG clinical study., Results: Slow-growth "virtual" murine GBMs benefited from increasing TMZ dose separation in-silico . In line with the simulation results, improved survival, reduced toxicity, lower expression of resistance factors, and reduction of the tumor mesenchymal component were observed in experimental models subject to long-cycle treatment, particularly in slowly growing tumors. Tissue analysis after long-cycle TMZ treatments revealed epigenetically driven changes in tumor phenotype, which could explain the reduction in GBM growth speed. In-silico trials provided support for implementation methods in human patients., Conclusions: In-silico simulations, in-vitro and in-vivo studies show that TMZ administration schedules with increased time between doses may reduce toxicity, delay the appearance of resistances and lead to survival benefits mediated by changes in the tumor phenotype in slowly-growing GBMs., (© The Author(s) 2022. Published by Oxford University Press, the Society for Neuro-Oncology and the European Association of Neuro-Oncology.)
- Published
- 2022
- Full Text
- View/download PDF
4. Infigratinib in Patients with Recurrent Gliomas and FGFR Alterations: A Multicenter Phase II Study.
- Author
-
Lassman AB, Sepúlveda-Sánchez JM, Cloughesy TF, Gil-Gil MJ, Puduvalli VK, Raizer JJ, De Vos FYF, Wen PY, Butowski NA, Clement PMJ, Groves MD, Belda-Iniesta C, Giglio P, Soifer HS, Rowsey S, Xu C, Avogadri F, Wei G, Moran S, and Roth P
- Subjects
- Adult, Follow-Up Studies, Humans, Microtubule-Associated Proteins, Phenylurea Compounds, Protein Kinase Inhibitors adverse effects, Pyrimidines, Receptor, Fibroblast Growth Factor, Type 3 genetics, Glioma drug therapy, Glioma genetics, Neoplasm Recurrence, Local drug therapy, Neoplasm Recurrence, Local genetics
- Abstract
Purpose: FGFR genomic alterations (amplification, mutations, and/or fusions) occur in ∼8% of gliomas, particularly FGFR1 and FGFR3. We conducted a multicenter open-label, single-arm, phase II study of a selective FGFR1-3 inhibitor, infigratinib (BGJ398), in patients with FGFR-altered recurrent gliomas., Patients and Methods: Adults with recurrent/progressive gliomas harboring FGFR alterations received oral infigratinib 125 mg on days 1 to 21 of 28-day cycles. The primary endpoint was investigator-assessed 6-month progression-free survival (PFS) rate by Response Assessment in Neuro-Oncology criteria. Comprehensive genomic profiling was performed on available pretreatment archival tissue to explore additional molecular correlations with efficacy., Results: Among 26 patients, the 6-month PFS rate was 16.0% [95% confidence interval (CI), 5.0-32.5], median PFS was 1.7 months (95% CI, 1.1-2.8), and objective response rate was 3.8%. However, 4 patients had durable disease control lasting longer than 1 year. Among these, 3 had tumors harboring activating point mutations at analogous positions of FGFR1 (K656E; n = 2) or FGFR3 (K650E; n = 1) in pretreatment tissue; an FGFR3-TACC3 fusion was detected in the other. Hyperphosphatemia was the most frequently reported treatment-related adverse event (all-grade, 76.9%; grade 3, 3.8%) and is a known on-target toxicity of FGFR inhibitors., Conclusions: FGFR inhibitor monotherapy with infigratinib had limited efficacy in a population of patients with recurrent gliomas and different FGFR genetic alterations, but durable disease control lasting more than 1 year was observed in patients with tumors harboring FGFR1 or FGFR3 point mutations or FGFR3-TACC3 fusions. A follow-up study with refined biomarker inclusion criteria and centralized FGFR testing is warranted., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
5. Safety and Efficacy of Crizotinib in Combination with Temozolomide and Radiotherapy in Patients with Newly Diagnosed Glioblastoma: Phase Ib GEINO 1402 Trial.
- Author
-
Martínez-García M, Velasco G, Pineda E, Gil-Gil M, Alameda F, Capellades J, Martín-Soberón MC, López-Valero I, Tovar Ambel E, Foro P, Taus Á, Arumi M, Hernández-Laín A, and Sepúlveda-Sánchez JM
- Abstract
Background: MET-signaling and midkine (ALK ligand) promote glioma cell maintenance and resistance against anticancer therapies. ALK and c-MET inhibition with crizotinib have a preclinical therapeutic rationale to be tested in newly diagnosed GBM., Methods: Eligible patients received crizotinib with standard radiotherapy (RT)/temozolomide (TMZ) followed by maintenance with crizotinib. The primary objective was to determine the recommended phase 2 dose (RP2D) in a 3 + 3 dose escalation (DE) strategy and safety evaluation in the expansion cohort (EC). Secondary objectives included progression-free (PFS) and overall survival (OS) and exploratory biomarker analysis., Results: The study enrolled 38 patients. The median age was 52 years (33-76), 44% were male, 44% were MGMT methylated, and three patients had IDH1/2 mutation. In DE, DLTs were reported in 1/6 in the second cohort (250 mg/QD), declaring 250 mg/QD of crizotinib as the RP2D for the EC. In the EC, 9/25 patients (32%) presented grade ≥3 adverse events. The median follow up was 18.7 months (m) and the median PFS was 10.7 m (95% CI, 7.7-13.8), with a 6 m PFS and 12 m PFS of 71.5% and 38.8%, respectively. At the time of this analysis, 1 died without progression and 24 had progressed. The median OS was 22.6 m (95% CI, 14.1-31.1) with a 24 m OS of 44.5%. Molecular biomarkers showed no correlation with efficacy., Conclusions: The addition of crizotinib to standard RT and TMZ for newly diagnosed GBM was safe and the efficacy was encouraging, warranting prospective validation in an adequately powered, randomized controlled study.
- Published
- 2022
- Full Text
- View/download PDF
6. IDP-410: a Novel Therapeutic Peptide that Alters N-MYC Stability and Reduces Angiogenesis and Tumor Progression in Glioblastomas.
- Author
-
Gargini R, Segura-Collar B, Garranzo-Asensio M, Hortigüela R, Iglesias-Hernández P, Lobato-Alonso D, Moreno-Raja M, Esteban-Martin S, Sepúlveda-Sánchez JM, Nevola L, and Sánchez-Gómez P
- Subjects
- Cell Line, Tumor, Cell Proliferation, Gene Expression Regulation, Neoplastic, Humans, N-Myc Proto-Oncogene Protein genetics, N-Myc Proto-Oncogene Protein metabolism, N-Myc Proto-Oncogene Protein therapeutic use, Neovascularization, Pathologic drug therapy, Peptides genetics, Peptides pharmacology, Peptides therapeutic use, Tumor Microenvironment, Brain Neoplasms genetics, Glioblastoma genetics
- Abstract
Glioblastomas (GBMs) are the most frequent and highly aggressive brain tumors, being resistant to all cytotoxic and molecularly targeted agents tested so far. There is, therefore, an urgent need to find novel therapeutic approaches and/or alternative targets to bring treatment options to patients. Here, we first show that GBMs express high levels of N-MYC protein, a transcription factor involved in normal brain development. A novel stapled peptide designed to specifically target N-MYC protein monomer, IDP-410, is able to impair the formation of N-MYC/MAX complex and reduce the stability of N-MYC itself. As a result, the viability of GBM cells is compromised. Moreover, the efficacy is found dependent on the levels of expression of N-MYC. Finally, we demonstrate that IDP-410 reduces GBM growth in vivo when administered systemically, both in subcutaneous and intracranial xenografts, reducing the vascularization of the tumors, highlighting a potential relationship between the function of N-MYC and the expression of mesenchymal/angiogenic genes. Overall, our results strengthen the view of N-MYC as a therapeutic target in GBM and strongly suggest that IDP-410 could be further developed to become a first-in-class inhibitor of N-MYC protein, affecting not only tumor cell proliferation and survival, but also the interplay between GBM cells and their microenvironment., (© 2022. The American Society for Experimental NeuroTherapeutics, Inc.)
- Published
- 2022
- Full Text
- View/download PDF
7. First-In-Human Phase I Study of a Next-Generation, Oral, TGFβ Receptor 1 Inhibitor, LY3200882, in Patients with Advanced Cancer.
- Author
-
Yap TA, Vieito M, Baldini C, Sepúlveda-Sánchez JM, Kondo S, Simonelli M, Cosman R, van der Westhuizen A, Atkinson V, Carpentier AF, Löhr M, Redman R, Mason W, Cervantes A, Le Rhun E, Ochsenreither S, Warren L, Zhao Y, Callies S, Estrem ST, Man M, Gandhi L, Avsar E, and Melisi D
- Subjects
- Antineoplastic Combined Chemotherapy Protocols adverse effects, Humans, Maximum Tolerated Dose, Paclitaxel therapeutic use, Transforming Growth Factor beta, Antineoplastic Agents adverse effects, Head and Neck Neoplasms, Pancreatic Neoplasms drug therapy
- Abstract
Purpose: A novel, selective, next-generation transforming growth factor beta (TGFβ) receptor type-1 small molecule inhibitor, LY3200882, demonstrated promising preclinical data. This first-in-human trial evaluated safety, tolerability, recommended phase II dose (RP2D), pharmacokinetics, pharmacodynamics, and preliminary antitumor activity of LY3200882 as monotherapy or with other anticancer agents in patients with advanced cancer., Patients and Methods: This phase I multicenter study of oral LY3200882 (NCT02937272) comprised dose escalation, monotherapy expansion in grade 4 glioma, and combination therapy in solid tumors (LY3200882 and PD-L1 inhibitor LY3300054), pancreatic cancer (LY3200882, gemcitabine, and nab-paclitaxel), and head and neck squamous cell cancer (LY3200882, cisplatin, and radiation)., Results: Overall, 139 patients with advanced cancer were treated. The majority (93.5%) of patients experienced ≥1 treatment-emergent adverse events (TEAE), with 39.6% LY3200882-related. Grade 3 LY3200882-related toxicities were only observed in combination therapy arms. One patient in the pancreatic cancer arm experienced cardiovascular toxicity. The LY3200882 monotherapy RP2Ds were established in two schedules: 50 mg twice a day 2-weeks-on/2-weeks-off and 35 mg twice a day 3-weeks-on/1-week-off. Four patients with grade 4 glioma had durable Revised Assessment in Neuro Oncology (RANO) partial responses (PR) with LY3200882 monotherapy ( n = 3) or LY3200882-LY3300054 combination therapy ( n = 1). In treatment-naïve patients with advanced pancreatic cancer, 6 of 12 patients achieved Response Evaluation Criteria in Solid Tumors (RECIST) v1.1 PR and 3 of 12 patients demonstrated stable disease, for an overall 75% disease-control rate with the combination of LY3200882, gemcitabine, and nab-paclitaxel., Conclusions: LY3200882 as monotherapy and combination therapy was safe and well tolerated with preliminary antitumor activity observed in pancreatic cancer. Further studies to evaluate the efficacy of LY3200882 with gemcitabine and nab-paclitaxel in advanced pancreatic cancer are warranted., (©2021 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
8. Tumor-Derived Pericytes Driven by EGFR Mutations Govern the Vascular and Immune Microenvironment of Gliomas.
- Author
-
Segura-Collar B, Garranzo-Asensio M, Herranz B, Hernández-SanMiguel E, Cejalvo T, Casas BS, Matheu A, Pérez-Núñez Á, Sepúlveda-Sánchez JM, Hernández-Laín A, Palma V, Gargini R, and Sánchez-Gómez P
- Subjects
- Adenine analogs & derivatives, Adenine pharmacology, Angiogenesis Inhibitors pharmacology, Animals, Blood Vessels metabolism, Blood Vessels pathology, Blood-Brain Barrier metabolism, Bone Marrow, Brain Neoplasms immunology, Brain Neoplasms pathology, Cell Line, Tumor, Chromosomes, Human, X, ErbB Receptors genetics, Glioma immunology, Glioma pathology, Humans, Immunity, Cellular, Isocitrate Dehydrogenase genetics, Mice, Pericytes drug effects, Pericytes metabolism, Piperidines pharmacology, Receptor, Platelet-Derived Growth Factor beta metabolism, SOX9 Transcription Factor, Sunitinib pharmacology, Tumor Hypoxia, Tumor Microenvironment, Brain Neoplasms blood supply, Cell Transdifferentiation, Cellular Microenvironment, Glioma blood supply, Mutation, Pericytes physiology
- Abstract
The extraordinary plasticity of glioma cells allows them to contribute to different cellular compartments in tumor vessels, reinforcing the vascular architecture. It was recently revealed that targeting glioma-derived pericytes, which represent a big percentage of the mural cell population in aggressive tumors, increases the permeability of the vessels and improves the efficiency of chemotherapy. However, the molecular determinants of this transdifferentiation process have not been elucidated. Here we show that mutations in EGFR stimulate the capacity of glioma cells to function as pericytes in a BMX- (bone marrow and X-linked) and SOX9-dependent manner. Subsequent activation of platelet-derived growth factor receptor beta in the vessel walls of EGFR-mutant gliomas stabilized the vasculature and facilitated the recruitment of immune cells. These changes in the tumor microenvironment conferred a growth advantage to the tumors but also rendered them sensitive to pericyte-targeting molecules such as ibrutinib or sunitinib. In the absence of EGFR mutations, high-grade gliomas were enriched in blood vessels, but showed a highly disrupted blood-brain barrier due to the decreased BMX/SOX9 activation and pericyte coverage, which led to poor oxygenation, necrosis, and hypoxia. Overall, these findings identify EGFR mutations as key regulators of the glioma-to-pericyte transdifferentiation, highlighting the intricate relationship between the tumor cells and their vascular and immune milieu. Our results lay the foundations for a vascular-dependent stratification of gliomas and suggest different therapeutic vulnerabilities determined by the genetic status of EGFR . SIGNIFICANCE: This study identifies the EGFR-related mechanisms that govern the capacity of glioma cells to transdifferentiate into pericytes, regulating the vascular and immune phenotypes of the tumors. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/8/2142/F1.large.jpg., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
9. Pneumatosis intestinalis in a radioactive iodine-refractory metastasic thyroid papillary carcinoma with BRAF V600E mutation treated with dabrafenib-trametinib: a case report.
- Author
-
Martín-Soberón MC, Ruiz S, De Velasco G, Yarza R, Carretero A, Castellano D, and Sepúlveda-Sánchez JM
- Subjects
- Emphysema chemically induced, Female, Humans, Intestinal Diseases chemically induced, Iodine Radioisotopes therapeutic use, Middle Aged, Positron Emission Tomography Computed Tomography, Proto-Oncogene Proteins B-raf genetics, Skull Base Neoplasms diagnostic imaging, Skull Base Neoplasms genetics, Skull Base Neoplasms secondary, Thyroid Cancer, Papillary diagnostic imaging, Thyroid Cancer, Papillary genetics, Thyroid Cancer, Papillary secondary, Emphysema diagnostic imaging, Imidazoles adverse effects, Intestinal Diseases diagnostic imaging, Oximes adverse effects, Protein Kinase Inhibitors adverse effects, Pyridones adverse effects, Pyrimidinones adverse effects, Skull Base Neoplasms drug therapy, Thyroid Cancer, Papillary drug therapy, Thyroid Neoplasms pathology
- Abstract
Background: Pneumatosis intestinalis (PI) is a rare entity which refers to the presence of gas within the wall of the small bowel or colon which is a radiographic sign. The etiology and clinical presentation are variable. Patients with PI may present either with chronic mild non-specific symptoms or with acute abdominal pain with peritonitis. Some cases of intestinal pneumatosis have been reported as adverse events of new oncological treatments such as targeted therapies that are widely used in multiple tumors., Case Presentation: A 59-year-old caucasian female with radioactive iodine-refractory metastatic thyroid papillary carcinoma with BRAF
V600E mutation was treated with dabrafenib and trametinib as a compassionate use. After 4 months treatment, positron emission tomography-computed tomography (PET-CT) showed PI. At the time of diagnosis, the patient was asymptomatic without signs of peritonitis. The initial treatment was conservative and no specific treatment for PI was needed. Unfortunately, after dabrafenib-trametinib withdrawal, the patient developed tumor progression with significant clinical worsening., Conclusions: This case report is, in our knowledge, the first description of PI in a patient treated with dabrafenib-trametinib. Conservative treatment is feasible if there are no abdominal symptoms.- Published
- 2021
- Full Text
- View/download PDF
10. The ESMO-EANO clinical practice guidelines for neurological and vascular complications of primary and secondary brain tumours: a valuable tool for clinicians.
- Author
-
Sepúlveda-Sánchez JM and Pérez-Núñez A
- Subjects
- Follow-Up Studies, Humans, Brain Neoplasms complications, Brain Neoplasms epidemiology, Brain Neoplasms therapy, Medical Oncology
- Abstract
Competing Interests: Disclosure The authors have declared no conflicts of interest.
- Published
- 2021
- Full Text
- View/download PDF
11. Immune Profiling of Gliomas Reveals a Connection with IDH1/2 Mutations, Tau Function and the Vascular Phenotype.
- Author
-
Cejalvo T, Gargini R, Segura-Collar B, Mata-Martínez P, Herranz B, Cantero D, Ruano Y, García-Pérez D, Pérez-Núñez Á, Ramos A, Hernández-Laín A, Martín-Soberón MC, Sánchez-Gómez P, and Sepúlveda-Sánchez JM
- Abstract
Background: Gliomas remain refractory to all attempted treatments, including those using immune checkpoint inhibitors. The characterization of the tumor (immune) microenvironment has been recognized as an important challenge to explain this lack of response and to improve the therapy of glial tumors., Methods: We designed a prospective analysis of the immune cells of gliomas by flow cytometry. Tumors with or without isocitrate dehydrogenase 1/2 ( IDH1/2 ) mutations were included in the study. The genetic profile and the presence of different molecular and cellular features of the gliomas were analyzed in parallel. The findings were validated in syngeneic mouse models., Results: We observed that few immune cells infiltrate mutant IDH1/2 gliomas whereas the immune content of IDH1/2 wild-type tumors was more heterogeneous. Some of them contained an important immune infiltrate, particularly enriched in myeloid cells with immunosuppressive features, but others were more similar to mutant IDH1/2 gliomas, with few immune cells and a less immunosuppressive profile. Notably, we observed a direct correlation between the percentage of leukocytes and the presence of vascular alterations, which were associated with a reduced expression of Tau, a microtubule-binding protein that controls the formation of tumor vessels in gliomas. Furthermore, overexpression of Tau was able to reduce the immune content in orthotopic allografts of GL261 cells, delaying tumor growth., Conclusions: We have confirmed the reduced infiltration of immune cells in IDH1/2 mutant gliomas. By contrast, in IDH1/2 wild-type gliomas, we have found a direct correlation between the presence of vascular alterations and the entrance of leukocytes into the tumors. Interestingly, high levels of Tau inversely correlated with the vascular and the immune content of gliomas. Altogether, our results could be exploited for the design of more successful clinical trials with immunomodulatory molecules.
- Published
- 2020
- Full Text
- View/download PDF
12. Ocoxin Modulates Cancer Stem Cells and M2 Macrophage Polarization in Glioblastoma.
- Author
-
Hernández-SanMiguel E, Gargini R, Cejalvo T, Segura-Collar B, Núñez-Hervada P, Hortigüela R, Sepúlveda-Sánchez JM, Hernández-Laín A, Pérez-Núñez A, Sanz E, and Sánchez-Gómez P
- Subjects
- Animals, Ascorbic Acid pharmacology, Folic Acid, Glioblastoma pathology, Humans, Mice, Mice, Nude, Pantothenic Acid, Plant Extracts pharmacology, Vitamin B 12 pharmacology, Vitamin B 6 pharmacology, Zinc Sulfate, Ascorbic Acid therapeutic use, Glioblastoma drug therapy, Macrophages metabolism, Neoplastic Stem Cells metabolism, Plant Extracts therapeutic use, Vitamin B 12 therapeutic use, Vitamin B 6 therapeutic use
- Abstract
Glioblastoma (GBM) is the most common and devastating primary brain tumor. The presence of cancer stem cells (CSCs) has been linked to their therapy resistance. Molecular and cellular components of the tumor microenvironment also play a fundamental role in the aggressiveness of these tumors. In particular, high levels of hypoxia and reactive oxygen species participate in several aspects of GBM biology. Moreover, GBM contains a large number of macrophages, which normally behave as immunosuppressive tumor-supportive cells. In fact, the presence of both, hypoxia and M2-like macrophages, correlates with malignancy and poor prognosis in gliomas. Antioxidant agents, as nutritional supplements, might have antitumor activity. Ocoxin® oral solution (OOS), in particular, has anti-inflammatory and antioxidant properties, as well as antitumor properties in several neoplasia, without known side effects. Here, we describe how OOS affects stem cell properties in certain GBMs, slowing down their tumor growth. In parallel, OOS has a direct effect on macrophage polarization in vitro and in vivo , inhibiting the protumoral features of M2 macrophages. Therefore, OOS could be a feasible candidate to be used in combination therapies during GBM treatment because it can target the highly resilient CSCs as well as their supportive immune microenvironment, without adding toxicity to conventional treatments., Competing Interests: The authors declare that this work was partially supported by Catalysis S.L. ES is a current employee of Catalysis S.L. The authors declare that there are no other nonfinancial competing interests.
- Published
- 2019
- Full Text
- View/download PDF
13. Phase II trial of dacomitinib, a pan-human EGFR tyrosine kinase inhibitor, in recurrent glioblastoma patients with EGFR amplification.
- Author
-
Sepúlveda-Sánchez JM, Vaz MÁ, Balañá C, Gil-Gil M, Reynés G, Gallego Ó, Martínez-García M, Vicente E, Quindós M, Luque R, Ramos A, Ruano Y, Pérez-Segura P, Benavides M, Sánchez-Gómez P, and Hernández-Laín A
- Subjects
- Adult, Aged, Aged, 80 and over, Brain Neoplasms genetics, Brain Neoplasms pathology, ErbB Receptors genetics, Female, Follow-Up Studies, Gene Expression Regulation, Neoplastic drug effects, Glioblastoma genetics, Glioblastoma pathology, Humans, Male, Middle Aged, Mutation, Prognosis, Signal Transduction, Survival Rate, Brain Neoplasms drug therapy, ErbB Receptors antagonists & inhibitors, Gene Amplification, Glioblastoma drug therapy, Quinazolinones therapeutic use
- Abstract
Background: We conducted a multicenter, 2-stage, open-label, phase II trial to assess the efficacy and safety of dacomitinib in adult patients with recurrent glioblastoma (GB) and epidermal growth factor receptor gene (EGFR) amplification with or without variant III (EGFRvIII) deletion., Methods: Patients with first recurrence were enrolled in 2 cohorts. Cohort A included patients with EGFR gene amplification without EGFRvIII mutation. Cohort B included patients with EGFR gene amplification and EGFRvIII mutation. Dacomitinib was administered (45 mg/day) until disease progression/unacceptable adverse events (AEs). Primary endpoint was progression-free survival (PFS; RANO criteria) at 6 months (PFS6)., Results: Thirty patients in Cohort A and 19 in Cohort B were enrolled. Median age was 59 years (range 39-81), 65.3% were male, and Eastern Cooperative Oncology Group Performance Status 0/1/2 were 10.2%/65.3%/24.5%, respectively. PFS6 was 10.6% (Cohort A: 13.3%; Cohort B: 5.9%) with a median PFS of 2.7 months (Cohort A: 2.7 mo; Cohort B: 2.6 mo). Four patients were progression free at 6 months and 3 patients were so at 12 months. Median overall survival was 7.4 months (Cohort A: 7.8 mo; Cohort B: 6.7 mo). The best overall response included 1 complete response and 2 partial responses (4.1%). Stable disease was observed in 12 patients (24.5%: eight in Cohort A and four in Cohort B). Diarrhea and rash were the most common AEs; 20 (40.8%) patients experienced grade 3-4 drug-related AEs., Conclusions: Dacomitinib has a limited single-agent activity in recurrent GB with EGFR amplification. The detailed molecular characterization of the 4 patients with response in this trial can be useful to select patients who could benefit from dacomitinib., (© The Author(s) 2017. Published by Oxford University Press on behalf of the Society for Neuro-Oncology. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com)
- Published
- 2017
- Full Text
- View/download PDF
14. Erratum: Efficacy and tolerability of lacosamide for secondary epileptic seizures in patients with brain tumor: A multicenter, observational retrospective study.
- Author
-
Sepúlveda-Sánchez JM, Conde-Moreno A, Barón M, Pardo J, Reynés G, and Belenguer A
- Abstract
[This corrects the article DOI: 10.3892/ol.2017.5988.].
- Published
- 2017
- Full Text
- View/download PDF
15. Efficacy and tolerability of lacosamide for secondary epileptic seizures in patients with brain tumor: A multicenter, observational retrospective study.
- Author
-
Sepúlveda-Sánchez JM, Conde-Moreno A, Barón M, Pardo J, Reynés G, and Belenguer A
- Abstract
The present observational, multicenter, retrospective study investigated the efficacy and tolerability of lacosamide in controlling secondary epileptic seizures in patients with brain tumors in Spain. Data from the medical records of patients ≥18 years of age with brain tumors, who had received at least one dose of lacosamide for seizure management between July 2013 and November 2013, were collected. The primary and secondary objectives of the present study were to assess the effectiveness and tolerability of lacosamide. Data from 39 patients (mean age, 54.1 years; 66.7% male) were collected, where the two main reasons for initiation of lacosamide treatment were the lack of efficacy of other antiepileptic drugs (in 76.9% of patients) and the presence of adverse events (12.8%) associated with other antiepileptic drugs. At the initiation of treatment, patients received a mean lacosamide dose of 138.5±68.3 mg/day. At 6 months, lacosamide had significantly reduced the mean number of seizures from 26.4 (standard deviation [SD], 50.4) seizures for the 6 months prior to lacosamide initiation to a mean of 9.4 (SD, 22.8) seizures during the 6 months subsequent to lacosamide initiation; P<0.001. Lacosamide was generally well tolerated; of the 25 patients who had complete safety data available at a 6-month follow-up, 3 patients (12%) reported an adverse event, including dizziness, asthenia, instability and irritability. The present retrospective analysis suggested that lacosamide is an effective and well-tolerated treatment in patients experiencing seizures due to brain tumors. Additional prospective studies with a larger patient population and randomized trial design are warranted.
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.