Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Mol Pharm ; 20(5): 2702-2713, 2023 05 01.
Article in English | MEDLINE | ID: mdl-37013916

ABSTRACT

Glioblastoma (GBM), as the most central nervous system (CNS) intractable disease, has spoiled millions of lives due to its high mortality. Even though several efforts have been made, the existing treatments have had limited success. In this sense, we studied a lead compound, the boron-rich selective epidermal growth factor receptor (EGFR)-inhibitor hybrid 1, as a potential drug for GBM treatment. For this end, we analyzed the in vitro activity of hybrid 1 in a glioma/primary astrocytes coculture, studying cellular death types triggered by treatment with this compound and its cellular localizations. Additionally, hybrid 1 concentrated boron in glioma cells selectively and more effectively than the boron neutron capture therapy (BNCT)-clinical agent 10B-l-boronophenylalanine and thus displayed a better in vitro-BNCT effect. This encouraged us to analyze hybrid 1 in vivo. Therefore, immunosuppressed mice bearing U87 MG human GBM were treated with both 1 and 1 encapsulated in a modified liposome (recognized by brain-blood barrier peptide transporters), and we observed a potent in vivo per se antitumor activity (tumor size decrease and animal survival increase). These data demonstrate that 1 could be a promising new targeted therapy for GBM.


Subject(s)
Boron Neutron Capture Therapy , Brain Neoplasms , Glioblastoma , Glioma , Mice , Humans , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Brain Neoplasms/metabolism , Boron , Boron Compounds/pharmacology , Boron Compounds/therapeutic use , Glioma/drug therapy , Glioma/radiotherapy , Glioma/metabolism , Glioblastoma/drug therapy
2.
Chem Biol Drug Des ; 101(6): 1435-1445, 2023 06.
Article in English | MEDLINE | ID: mdl-36746676

ABSTRACT

Moving towards high-grade glioma drug discovery, this study aimed to detect the mechanism of cellular death (apoptosis, necrosis and/or autophagy) induced by three carboranyl-based lead compounds. For that, we performed in U87 MG cells, flow cytometry experiments, as the gold standard technique, as well as confocal microscopy and 1 H-NMR experiments as non-invasive assays. We selected three hybrid leads (1-3) from the in-house-library and the corresponding parent compounds, and recognized tyrosine kinase inhibitors (lapatinib, sunitinib and erlotinib) to put to the test in these experiments. Flow cytometry with Annexin V-FITC/DAPI staining showed that leads 1 and 3 and lapatinib mainly induced necrosis in U87 MG upon a 24 h treatment at IC50 dose; meanwhile, hybrid 2, sunitinib and erlotinib seem to induce apoptosis in such cells. In general, confocal microscopy studies were in agreement with flow cytometry observing loss of cell membrane integrity in necrotic cells and features of apoptosis, that is, chromatin condensation, in apoptotic cells. Finally, NMR results showed that glioblastoma cells treated with hybrid 1, 3 or lapatinib displayed changes in CH2 /CH3 signal ratio and choline signals that could indicate necrotic cell death mechanism: meanwhile, 2-, sunitinib- or erlotinib-treated cells showed apoptotic characteristic behaviors. Additionally, carboranyl-hybrid 2 also produced autophagy in U87 MG cells.


Subject(s)
Antineoplastic Agents , Glioblastoma , Humans , Glioblastoma/drug therapy , Glioblastoma/metabolism , Lapatinib/pharmacology , Lapatinib/therapeutic use , Sunitinib , Erlotinib Hydrochloride/pharmacology , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis , Necrosis/drug therapy , Cell Proliferation
3.
Int J Mol Sci ; 23(24)2022 Dec 16.
Article in English | MEDLINE | ID: mdl-36555682

ABSTRACT

Through a combination of comparative modeling, site-directed and classical random mutagenesis approaches, we previously identified critical residues for binding, recognition, and translocation of urea, and its inhibition by 2-thiourea and acetamide in the Aspergillus nidulans urea transporter, UreA. To deepen the structural characterization of UreA, we employed the artificial intelligence (AI) based AlphaFold2 (AF2) program. In this analysis, the resulting AF2 models lacked inward- and outward-facing cavities, suggesting a structural intermediate state of UreA. Moreover, the orientation of the W82, W84, N279, and T282 side chains showed a large variability, which in the case of W82 and W84, may operate as a gating mechanism in the ligand pathway. To test this hypothesis non-conservative and conservative substitutions of these amino acids were introduced, and binding and transport assessed for urea and its toxic analogue 2-thiourea, as well as binding of the structural analogue acetamide. As a result, residues W82, W84, N279, and T282 were implicated in substrate identification, selection, and translocation. Using molecular docking with Autodock Vina with flexible side chains, we corroborated the AF2 theoretical intermediate model, showing a remarkable correlation between docking scores and experimental affinities determined in wild-type and UreA mutants. The combination of AI-based modeling with classical docking, validated by comprehensive mutational analysis at the binding region, would suggest an unforeseen option to determine structural level details on a challenging family of proteins.


Subject(s)
Artificial Intelligence , Furylfuramide , Molecular Docking Simulation , Urea/metabolism , Thiourea , Acetamides , Urea Transporters
4.
Sci Adv ; 8(38): eabn6545, 2022 Sep 23.
Article in English | MEDLINE | ID: mdl-36129987

ABSTRACT

Severe COVID-19 is associated with hyperinflammation and weak T cell responses against SARS-CoV-2. However, the links between those processes remain partially characterized. Moreover, whether and how therapeutically manipulating T cells may benefit patients are unknown. Our genetic and pharmacological evidence demonstrates that the ion channel TMEM176B inhibited inflammasome activation triggered by SARS-CoV-2 and SARS-CoV-2-related murine ß-coronavirus. Tmem176b-/- mice infected with murine ß-coronavirus developed inflammasome-dependent T cell dysfunction and critical disease, which was controlled by modulating dysfunctional T cells with PD-1 blockers. In critical COVID-19, inflammasome activation correlated with dysfunctional T cells and low monocytic TMEM176B expression, whereas PD-L1 blockade rescued T cell functionality. Here, we mechanistically link T cell dysfunction and inflammation, supporting a cancer immunotherapy to reinforce T cell immunity in critical ß-coronavirus disease.

5.
Acta Neuropathol Commun ; 9(1): 136, 2021 08 13.
Article in English | MEDLINE | ID: mdl-34389060

ABSTRACT

Degeneration of motor neurons, glial cell reactivity, and vascular alterations in the CNS are important neuropathological features of amyotrophic lateral sclerosis (ALS). Immune cells trafficking from the blood also infiltrate the affected CNS parenchyma and contribute to neuroinflammation. Mast cells (MCs) are hematopoietic-derived immune cells whose precursors differentiate upon migration into tissues. Upon activation, MCs undergo degranulation with the ability to increase vascular permeability, orchestrate neuroinflammation and modulate the neuroimmune response. However, the prevalence, pathological significance, and pharmacology of MCs in the CNS of ALS patients remain largely unknown. In autopsy ALS spinal cords, we identified for the first time that MCs express c-Kit together with chymase, tryptase, and Cox-2 and display granular or degranulating morphology, as compared with scarce MCs in control cords. In ALS, MCs were mainly found in the niche between spinal motor neuron somas and nearby microvascular elements, and they displayed remarkable pathological abnormalities. Similarly, MCs accumulated in the motor neuron-vascular niche of ALS murine models, in the vicinity of astrocytes and motor neurons expressing the c-Kit ligand stem cell factor (SCF), suggesting an SCF/c-Kit-dependent mechanism of MC differentiation from precursors. Mechanistically, we provide evidence that fully differentiated MCs in cell cultures can be generated from the murine ALS spinal cord tissue, further supporting the presence of c-Kit+ MC precursors. Moreover, intravenous administration of bone marrow-derived c-Kit+ MC precursors infiltrated the spinal cord in ALS mice but not in controls, consistent with aberrant trafficking through a defective microvasculature. Pharmacological inhibition of c-Kit with masitinib in ALS mice reduced the MC number and the influx of MC precursors from the periphery. Our results suggest a previously unknown pathogenic mechanism triggered by MCs in the ALS motor neuron-vascular niche that might be targeted pharmacologically.


Subject(s)
Amyotrophic Lateral Sclerosis/immunology , Mast Cells/immunology , Microvessels/pathology , Motor Neurons/pathology , Neuroinflammatory Diseases/immunology , Proto-Oncogene Proteins c-kit/metabolism , Spinal Cord/immunology , Aged , Aged, 80 and over , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Astrocytes/metabolism , Astrocytes/pathology , Benzamides/pharmacology , Case-Control Studies , Chymases/metabolism , Cyclooxygenase 2/metabolism , Female , Humans , Male , Mast Cells/drug effects , Mast Cells/metabolism , Mice , Microvessels/metabolism , Middle Aged , Motor Neurons/metabolism , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Pyridines/pharmacology , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord/pathology , Stem Cell Factor/metabolism , Thiazoles/pharmacology , Tryptases/metabolism
6.
Cancers (Basel) ; 12(11)2020 Nov 18.
Article in English | MEDLINE | ID: mdl-33218150

ABSTRACT

Malignant gliomas are the most common malignant and aggressive primary brain tumors in adults, the prognosis being-especially for glioblastomas-extremely poor. There are no effective treatments yet. However, tyrosine kinase receptor (TKR) inhibitors and boron neutron capture therapy (BNCT), together, have been proposed as future therapeutic strategies. In this sense in our ongoing project of developing new anti-glioblastoma drugs, we identified a sunitinib-carborane hybrid agent, 1, with both in vitro selective cytotoxicity and excellent BNCT-behavior. Consequently, we studied the ability of compound 1 to inhibit TKRs, its promotion of cellular death processes, and its effects on the cell cycle. Moreover, we analyzed some relevant drug-like properties of 1, i.e., mutagenicity and ability to cross the blood-brain barrier. These results encouraged us to perform an in vivo anti-glioblastoma proof of concept assay. It turned out to be a selective FLT3, KIT, and PDGFR-ß inhibitor and increased the apoptotic glioma-cell numbers and arrested sub-G1-phase cell cycle. Its in vivo activity in immunosuppressed mice bearing U87 MG human glioblastoma evidenced excellent anti-tumor behavior.

7.
Chemistry ; 26(63): 14335-14340, 2020 Nov 11.
Article in English | MEDLINE | ID: mdl-32738078

ABSTRACT

About 95 % of people diagnosed with glioblastoma die within five years. Glioblastoma is the most aggressive central nervous system tumour. It is necessary to make progress in the glioblastoma treatment so that advanced chemotherapy drugs or radiation therapy or, ideally, two-in-one hybrid systems should be implemented. Tyrosine kinase receptors-inhibitors and boron neutron capture therapy (BNCT), together, could provide a therapeutic strategy. In this work, sunitinib decorated-carborane hybrids were prepared and biologically evaluated identifying excellent antitumoral- and BNCT-agents. One of the selected hybrids was studied against glioma-cells and found to be 4 times more cytotoxic than sunitinib and 1.7 times more effective than 10 B-boronophenylalanine fructose complex when the cells were irradiated with neutrons.


Subject(s)
Antineoplastic Agents , Boron Neutron Capture Therapy , Brain Neoplasms , Cell Survival/drug effects , Glioblastoma , Pharmaceutical Preparations , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Boron Compounds , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Cell Line, Tumor , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Humans , Mice , Phenylalanine
8.
Cells ; 9(6)2020 06 05.
Article in English | MEDLINE | ID: mdl-32517054

ABSTRACT

One of the driving forces of carcinogenesis in humans is the aberrant activation of receptors; consequently, one of the most promising mechanisms for cancer treatment is receptor inhibition by chemotherapy. Although a variety of cancers are initially susceptible to chemotherapy, they eventually develop multi-drug resistance. Anti-tumor agents overcoming resistance and acting through two or more ways offer greater therapeutic benefits over single-mechanism entities. In this study, we report on a new family of bifunctional compounds that, offering the possibility of dual action (drug + radiotherapy combinations), may result in significant clinical benefits. This new family of compounds combines two fragments: the drug fragment is a lapatinib group, which inhibits the tyrosine kinase receptor activity, and an icosahedral boron cluster used as agents for neutron capture therapy (BNCT). The developed compounds were evaluated in vitro against different tyrosine kinase receptors (TKRs)-expressing tumoral cells, and in vitro-BNCT experiments were performed for two of the most promising hybrids, 19 and 22. We identified hybrid 19 with excellent selectivity to inhibit cell proliferation and ability to induce necrosis/apoptosis of glioblastoma U87 MG cell line. Furthermore, derivative 22, bearing a water-solubility-enhancing moiety, showed moderate inhibition of cell proliferation in both U87 MG and colorectal HT-29 cell lines. Additionally, the HT-29 cells accumulated adequate levels of boron after hybrids 19 and 22 incubations rendering, and after neutron irradiation, higher BNCT-effects than BPA. The attractive profile of developed hybrids makes them interesting agents for combined therapy.


Subject(s)
Boron Neutron Capture Therapy , Lapatinib/therapeutic use , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Animals , Animals, Newborn , Boron Compounds/chemical synthesis , Boron Compounds/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Humans , Inhibitory Concentration 50 , Lapatinib/chemistry , Lapatinib/pharmacology , Mice , Protein Kinase Inhibitors/pharmacology , Triazines/chemical synthesis , Triazines/chemistry
9.
Future Med Chem ; 11(17): 2273-2285, 2019 09.
Article in English | MEDLINE | ID: mdl-31581913

ABSTRACT

Background: Carboranylanilinoquinazoline-hybrids, developed for boron neutron capture therapy, have demonstrated cytotoxicity against murine-glioma cells with EGFR-inhibition ability. In addition, their adequate aqueous/metabolic stabilities and ability to cross blood-brain barrier make them good leads as to become antiglioma drugs. Aim: Analyze drug-like properties of representative carboranylanilinoquinazolines. Materials & methods: To expand carboranylanilinoquinazolines therapeutic spectrum, we studied their ability to act against glioma-mammal cells, U-87 MG and other tyrosine kinase-overexpress cells, HT-29. Additionally, we predicted theoretically and studied experimentally drug-like properties, in other words, organization for economic cooperation and development-recommended toxicity-studies and, due to some aqueous-solubility problems, and vehicularization for oral and intravenous administrations. Conclusion: We have identified a promising drug-candidate with broad activity spectrum, appropriate drug-like properties, adequate toxicological behavior and able ability to be loaded in suitable vehicles.


Subject(s)
Aniline Compounds/chemistry , Antineoplastic Agents/chemistry , Brain Neoplasms/radiotherapy , ErbB Receptors/antagonists & inhibitors , Glioma/radiotherapy , Protein Kinase Inhibitors/chemistry , Quinazolines/chemistry , Aniline Compounds/pharmacology , Animals , Antineoplastic Agents/pharmacology , Blood-Brain Barrier/metabolism , Boron Neutron Capture Therapy/methods , Cell Line, Tumor , Cell Survival , Cholesterol/chemistry , Drug Compounding/methods , Drug Development , Drug Liberation , Female , Humans , Liposomes/chemistry , Mice , Mice, Inbred BALB C , Models, Molecular , Phosphatidylcholines/chemistry , Polyamines/chemistry , Polyethylenes/chemistry , Polypropylenes/chemistry , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/metabolism , Quinazolines/pharmacology , Solubility , Water
10.
Chemistry ; 24(13): 3122-3126, 2018 Mar 02.
Article in English | MEDLINE | ID: mdl-29194843

ABSTRACT

New 1,7-closo-carboranylanilinoquinazoline hybrids have been identified as EGFR inhibitors, one of them with higher affinity than the parent compound erlotinib. The comparative docking analysis with compounds bearing bioisoster-substructures, demonstrated the relevance of the 3D aromatic-boron-rich moiety for interacting into the EGFR ATP binding region. The capability to accumulate in glioma cells, the ability to cross the blood-brain barrier and the stability on simulated biological conditions, render these molecules as lead compounds for further structural modifications to obtain dual action drugs to treat glioblastoma.


Subject(s)
Boron/analysis , ErbB Receptors/antagonists & inhibitors , Glioma/drug therapy , Quinazolines/therapeutic use , Aniline Compounds , Blood-Brain Barrier/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Protein Kinase Inhibitors/chemistry , Quinazolines/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...