Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Microorganisms ; 9(3)2021 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-33801811

RESUMEN

Outbreaks of Ebola ebolavirus (EBOV) have been associated with high morbidity and mortality. Milestones have been reached recently in the management of EBOV disease (EVD) with licensure of an EBOV vaccine and two monoclonal antibody therapies. However, neither vaccines nor therapies are available for other disease-causing filoviruses. In preparation for such outbreaks, and for more facile and cost-effective management of EVD, we seek a cocktail containing orally available and room temperature stable drugs with strong activity against multiple filoviruses. We previously showed that (bepridil + sertraline) and (sertraline + toremifene) synergistically suppress EBOV in cell cultures. Here, we describe steps towards testing these combinations in a mouse model of EVD. We identified a vehicle suitable for oral delivery of the component drugs and determined that, thus formulated the drugs are equally active against EBOV as preparations in DMSO, and they maintain activity upon storage in solution for up to seven days. Pharmacokinetic (PK) studies indicated that the drugs in the oral delivery vehicle are well tolerated in mice at the highest doses tested. Collectively the data support advancement of these combinations to tests for synergy in a mouse model of EVD. Moreover, mathematical modeling based on human oral PK projects that the combinations would be more active in humans than their component single drugs.

3.
Artículo en Inglés | MEDLINE | ID: mdl-33468464

RESUMEN

Neglected diseases caused by arenaviruses such as Lassa virus (LASV) and filoviruses like Ebola virus (EBOV) primarily afflict resource-limited countries, where antiviral drug development is often minimal. Previous studies have shown that many approved drugs developed for other clinical indications inhibit EBOV and LASV and that combinations of these drugs provide synergistic suppression of EBOV, often by blocking discrete steps in virus entry. We hypothesize that repurposing of combinations of orally administered approved drugs provides effective suppression of arenaviruses. In this report, we demonstrate that arbidol, an approved influenza antiviral previously shown to inhibit EBOV, LASV, and many other viruses, inhibits murine leukemia virus (MLV) reporter viruses pseudotyped with the fusion glycoproteins (GPs) of other arenaviruses (Junin virus [JUNV], lymphocytic choriomeningitis virus [LCMV], and Pichinde virus [PICV]). Arbidol and other approved drugs, including aripiprazole, amodiaquine, sertraline, and niclosamide, also inhibit infection of cells by infectious PICV, and arbidol, sertraline, and niclosamide inhibit infectious LASV. Combining arbidol with aripiprazole or sertraline results in the synergistic suppression of LASV and JUNV GP-bearing pseudoviruses. This proof-of-concept study shows that arenavirus infection in vitro can be synergistically inhibited by combinations of approved drugs. This approach may lead to a proactive strategy with which to prepare for and control known and new arenavirus outbreaks.


Asunto(s)
Antivirales/uso terapéutico , Infecciones por Arenaviridae/tratamiento farmacológico , Arenavirus/efectos de los fármacos , Administración Oral , Animales , Infecciones por Arenaviridae/virología , Línea Celular , Chlorocebus aethiops , Sinergismo Farmacológico , Quimioterapia Combinada/métodos , Células HEK293 , Humanos , Ratones , Prueba de Estudio Conceptual , Células Vero
4.
J Infect Dis ; 218(suppl_5): S672-S678, 2018 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-29939303

RESUMEN

Background: A need to develop therapeutics to treat Ebola virus disease patients in remote and resource-challenged settings remains in the wake of the 2013-2016 epidemic in West Africa. Toward this goal, we screened drugs under consideration as treatment options and other drugs of interest, most being small molecules approved by the Food and Drug Administration. Drugs demonstrating in vitro antiviral activity were advanced for evaluation in combinations because of advantages often provided by drug cocktails. Methods: Drugs were screened for blockade of Ebola virus infection in cultured cells. Twelve drugs were tested in all (78 pair-wise) combinations, and 3 were tested in a subset of combinations. Results: Multiple synergistic drug pairs emerged, with the majority comprising 2 entry inhibitors. For the pairs of entry inhibitors studied, synergy was demonstrated at the level of virus entry into host cells. Highly synergistic pairs included aripiprazole/piperacetazine, sertraline/toremifene, sertraline/bepridil, and amodiaquine/clomiphene. Conclusions: Our study shows the feasibility of identifying pairs of approved drugs that synergistically block Ebola virus infection in cell cultures. We discuss our findings in terms of the theoretic ability of these or alternate combinations to reach therapeutic levels. Future research will assess selected combinations in small-animal models of Ebola virus disease.


Asunto(s)
Antivirales/administración & dosificación , Ebolavirus/efectos de los fármacos , Animales , Antivirales/uso terapéutico , Chlorocebus aethiops , Aprobación de Drogas , Sinergismo Farmacológico , Quimioterapia Combinada , Células Vero , Virión/efectos de los fármacos , Internalización del Virus/efectos de los fármacos
5.
Sci Transl Med ; 7(290): 290ra89, 2015 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-26041706

RESUMEN

Currently, no approved therapeutics exist to treat or prevent infections induced by Ebola viruses, and recent events have demonstrated an urgent need for rapid discovery of new treatments. Repurposing approved drugs for emerging infections remains a critical resource for potential antiviral therapies. We tested ~2600 approved drugs and molecular probes in an in vitro infection assay using the type species, Zaire ebolavirus. Selective antiviral activity was found for 80 U.S. Food and Drug Administration-approved drugs spanning multiple mechanistic classes, including selective estrogen receptor modulators, antihistamines, calcium channel blockers, and antidepressants. Results using an in vivo murine Ebola virus infection model confirmed the protective ability of several drugs, such as bepridil and sertraline. Viral entry assays indicated that most of these antiviral drugs block a late stage of viral entry. By nature of their approved status, these drugs have the potential to be rapidly advanced to clinical settings and used as therapeutic countermeasures for Ebola virus infections.


Asunto(s)
Antivirales/uso terapéutico , Aprobación de Drogas , Fiebre Hemorrágica Ebola/terapia , Sondas Moleculares , Animales , Bepridil/farmacología , Ebolavirus/efectos de los fármacos , Humanos , Ratones , Sertralina/farmacología
6.
Antimicrob Agents Chemother ; 58(8): 4885-93, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24841273

RESUMEN

Outbreaks of emerging infections present health professionals with the unique challenge of trying to select appropriate pharmacologic treatments in the clinic with little time available for drug testing and development. Typically, clinicians are left with general supportive care and often untested convalescent-phase plasma as available treatment options. Repurposing of approved pharmaceutical drugs for new indications presents an attractive alternative to clinicians, researchers, public health agencies, drug developers, and funding agencies. Given the development times and manufacturing requirements for new products, repurposing of existing drugs is likely the only solution for outbreaks due to emerging viruses. In the studies described here, a library of 290 compounds was screened for antiviral activity against Middle East respiratory syndrome coronavirus (MERS-CoV) and severe acute respiratory syndrome coronavirus (SARS-CoV). Selection of compounds for inclusion in the library was dependent on current or previous FDA approval or advanced clinical development. Some drugs that had a well-defined cellular pathway as target were included. In total, 27 compounds with activity against both MERS-CoV and SARS-CoV were identified. The compounds belong to 13 different classes of pharmaceuticals, including inhibitors of estrogen receptors used for cancer treatment and inhibitors of dopamine receptor used as antipsychotics. The drugs identified in these screens provide new targets for in vivo studies as well as incorporation into ongoing clinical studies.


Asunto(s)
Antivirales/farmacología , Reposicionamiento de Medicamentos , Coronavirus del Síndrome Respiratorio de Oriente Medio/efectos de los fármacos , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Antipsicóticos/farmacología , Chlorocebus aethiops , Aprobación de Drogas , Antagonistas de Estrógenos/farmacología , Ensayos Analíticos de Alto Rendimiento , Humanos , Concentración 50 Inhibidora , Coronavirus del Síndrome Respiratorio de Oriente Medio/fisiología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/fisiología , Células Vero , Replicación Viral/efectos de los fármacos
7.
Sci Transl Med ; 5(190): 190ra79, 2013 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-23785035

RESUMEN

Ebola viruses remain a substantial threat to both civilian and military populations as bioweapons, during sporadic outbreaks, and from the possibility of accidental importation from endemic regions by infected individuals. Currently, no approved therapeutics exist to treat or prevent infection by Ebola viruses. Therefore, we performed an in vitro screen of Food and Drug Administration (FDA)- and ex-US-approved drugs and selected molecular probes to identify drugs with antiviral activity against the type species Zaire ebolavirus (EBOV). From this screen, we identified a set of selective estrogen receptor modulators (SERMs), including clomiphene and toremifene, which act as potent inhibitors of EBOV infection. Anti-EBOV activity was confirmed for both of these SERMs in an in vivo mouse infection model. This anti-EBOV activity occurred even in the absence of detectable estrogen receptor expression, and both SERMs inhibited virus entry after internalization, suggesting that clomiphene and toremifene are not working through classical pathways associated with the estrogen receptor. Instead, the response appeared to be an off-target effect where the compounds interfere with a step late in viral entry and likely affect the triggering of fusion. These data support the screening of readily available approved drugs to identify therapeutics for the Ebola viruses and other infectious diseases. The SERM compounds described in this report are an immediately actionable class of approved drugs that can be repurposed for treatment of filovirus infections.


Asunto(s)
Aprobación de Drogas , Ebolavirus/fisiología , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Moduladores Selectivos de los Receptores de Estrógeno/uso terapéutico , United States Food and Drug Administration , Animales , Catepsinas/metabolismo , Chlorocebus aethiops , Clomifeno/farmacología , Clomifeno/uso terapéutico , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Ebolavirus/efectos de los fármacos , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Fiebre Hemorrágica Ebola/virología , Células Hep G2 , Humanos , Concentración de Iones de Hidrógeno/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Receptores de Estrógenos/metabolismo , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Análisis de Supervivencia , Toremifeno/farmacología , Toremifeno/uso terapéutico , Estados Unidos , Células Vero , Virión/efectos de los fármacos , Internalización del Virus/efectos de los fármacos
8.
PLoS One ; 8(2): e56265, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23441171

RESUMEN

Ebola virus (EBOV) is an enveloped RNA virus that causes hemorrhagic fever in humans and non-human primates. Infection requires internalization from the cell surface and trafficking to a late endocytic compartment, where viral fusion occurs, providing a conduit for the viral genome to enter the cytoplasm and initiate replication. In a concurrent study, we identified clomiphene as a potent inhibitor of EBOV entry. Here, we screened eleven inhibitors that target the same biosynthetic pathway as clomiphene. From this screen we identified six compounds, including U18666A, that block EBOV infection (IC(50) 1.6 to 8.0 µM) at a late stage of entry. Intriguingly, all six are cationic amphiphiles that share additional chemical features. U18666A induces phenotypes, including cholesterol accumulation in endosomes, associated with defects in Niemann-Pick C1 protein (NPC1), a late endosomal and lysosomal protein required for EBOV entry. We tested and found that all six EBOV entry inhibitors from our screen induced cholesterol accumulation. We further showed that higher concentrations of cationic amphiphiles are required to inhibit EBOV entry into cells that overexpress NPC1 than parental cells, supporting the contention that they inhibit EBOV entry in an NPC1-dependent manner. A previously reported inhibitor, compound 3.47, inhibits EBOV entry by blocking binding of the EBOV glycoprotein to NPC1. None of the cationic amphiphiles tested had this effect. Hence, multiple cationic amphiphiles (including several FDA approved agents) inhibit EBOV entry in an NPC1-dependent fashion, but by a mechanism distinct from that of compound 3.47. Our findings suggest that there are minimally two ways of perturbing NPC1-dependent pathways that can block EBOV entry, increasing the attractiveness of NPC1 as an anti-filoviral therapeutic target.


Asunto(s)
Proteínas Portadoras/metabolismo , Cationes , Ebolavirus/efectos de los fármacos , Ebolavirus/fisiología , Glicoproteínas de Membrana/metabolismo , Tensoactivos/farmacología , Internalización del Virus/efectos de los fármacos , Animales , Antivirales/química , Antivirales/farmacología , Vías Biosintéticas/efectos de los fármacos , Cationes/química , Línea Celular , Fiebre Hemorrágica Ebola , Humanos , Péptidos y Proteínas de Señalización Intracelular , Proteína Niemann-Pick C1 , Fenotipo , Esteroides/biosíntesis , Tensoactivos/química
9.
Antimicrob Agents Chemother ; 55(3): 1028-35, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21149629

RESUMEN

Anthrax lethal toxin (LT) is the major virulence factor for Bacillus anthracis. The lethal factor (LF) component of this bipartite toxin is a protease which, when transported into the cellular cytoplasm, cleaves mitogen-activated protein kinase kinase (MEK) family proteins and induces rapid toxicity in mouse macrophages through activation of the Nlrp1b inflammasome. A high-throughput screen was performed to identify synergistic LT-inhibitory drug combinations from within a library of approved drugs and molecular probes. From this screen we discovered that auranofin, an organogold compound with anti-inflammatory activity, strongly inhibited LT-mediated toxicity in mouse macrophages. Auranofin did not inhibit toxin transport into cells or MEK cleavage but inhibited both LT-mediated caspase-1 activation and caspase-1 catalytic activity. Thus, auranofin inhibited LT-mediated toxicity by preventing activation of the Nlrp1b inflammasome and the downstream actions that occur in response to the toxin. Idebenone, an analog of coenzyme Q, synergized with auranofin to increase its protective effect. We found that idebenone functions as an inhibitor of voltage-gated potassium channels and thus likely mediates synergy through inhibition of the potassium fluxes which have been shown to be required for Nlrp1b inflammasome activation.


Asunto(s)
Antígenos Bacterianos/toxicidad , Proteínas Reguladoras de la Apoptosis/metabolismo , Auranofina/farmacología , Toxinas Bacterianas/toxicidad , Animales , Caspasa 1/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Interleucina-1beta/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Ratas , Ratas Endogámicas F344
10.
Mol Syst Biol ; 6: 375, 2010 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-20531405

RESUMEN

The search for effective Hepatitis C antiviral therapies has recently focused on host sterol metabolism and protein prenylation pathways that indirectly affect viral replication. However, inhibition of the sterol pathway with statin drugs has not yielded consistent results in patients. Here, we present a combination chemical genetic study to explore how the sterol and protein prenylation pathways work together to affect hepatitis C viral replication in a replicon assay. In addition to finding novel targets affecting viral replication, our data suggest that the viral replication is strongly affected by sterol pathway regulation. There is a marked transition from antagonistic to synergistic antiviral effects as the combination targets shift downstream along the sterol pathway. We also show how pathway regulation frustrates potential hepatitis C therapies based on the sterol pathway, and reveal novel synergies that selectively inhibit hepatitis C replication over host toxicity. In particular, combinations targeting the downstream sterol pathway enzymes produced robust and selective synergistic inhibition of hepatitis C replication. Our findings show how combination chemical genetics can reveal critical pathway connections relevant to viral replication, and can identify potential treatments with an increased therapeutic window.


Asunto(s)
Antivirales/farmacología , Hepacivirus/efectos de los fármacos , Hepacivirus/fisiología , Redes y Vías Metabólicas/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Línea Celular Tumoral , Sinergismo Farmacológico , Regulación Viral de la Expresión Génica/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento , Humanos , Hidroximetilglutaril-CoA Reductasas/metabolismo , ARN Viral/genética , Replicón/genética , Reproducibilidad de los Resultados , Esteroles/biosíntesis
11.
Nat Biotechnol ; 27(7): 659-66, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19581876

RESUMEN

Drug combinations are a promising strategy to overcome the compensatory mechanisms and unwanted off-target effects that limit the utility of many potential drugs. However, enthusiasm for this approach is tempered by concerns that the therapeutic synergy of a combination will be accompanied by synergistic side effects. Using large scale simulations of bacterial metabolism and 94,110 multi-dose experiments relevant to diverse diseases, we provide evidence that synergistic drug combinations are generally more specific to particular cellular contexts than are single agent activities. We highlight six combinations whose selective synergy depends on multitarget drug activity. For one anti-inflammatory example, we show how such selectivity is achieved through differential expression of the drugs' targets in cell types associated with therapeutic, but not toxic, effects and validate its therapeutic relevance in a rat model of asthma. The context specificity of synergistic combinations creates many opportunities for therapeutically relevant selectivity and enables improved control of complex biological systems.


Asunto(s)
Sinergismo Farmacológico , Quimioterapia Combinada , Preparaciones Farmacéuticas/administración & dosificación , Farmacología , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Descubrimiento de Drogas , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Escherichia coli/efectos de los fármacos , Escherichia coli/crecimiento & desarrollo , Humanos , Masculino , Modelos Biológicos , Ratas , Ratas Sprague-Dawley , Reproducibilidad de los Resultados
12.
Blood ; 102(9): 3163-71, 2003 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-12869508

RESUMEN

The transcription factor C/EBP alpha (CCAAT/enhancer binding protein alpha) is critical for granulopoiesis. Gene disruption in mice blocks early granulocyte differentiation and disruption of C/EBP alpha function has been implicated in human acute myeloid leukemia (AML), but no systematic structure-function analysis has been undertaken to identify the mechanisms involved in C/EBP alpha-mediated granulocyte differentiation. Here we demonstrate that loss of either of 2 key regions results in disruption of C/EBP alpha granulocytic development: the amino terminus and specific residues residing on the non-DNA binding face of the basic region. Mutation of either results in loss of C/EBP alpha inhibition of E2F and down-regulation of c-Myc, but only mutation of the basic region results in loss of physical interaction with E2F. In contrast, while the amino terminal mutant retains the ability to interact with E2F, this mutant fails to bind a C/EBP alpha site efficiently, fails to activate C/EBP alpha target genes, and is also defective in inhibition of E2F activity. These results further emphasize the importance of inhibition of proliferative pathways in granulopoiesis and demonstrate that several regions of the C/EBP alpha protein are involved in this mechanism.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/química , Proteína alfa Potenciadora de Unión a CCAAT/fisiología , Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Granulocitos/citología , Hematopoyesis , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Proteína alfa Potenciadora de Unión a CCAAT/genética , Factores de Transcripción E2F , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/citología , Humanos , Células K562 , Mutación , Estructura Terciaria de Proteína/fisiología , Factores de Transcripción/genética , Factores de Transcripción/fisiología
13.
J Virol ; 77(10): 6029-40, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12719594

RESUMEN

The immortalization of human B lymphocytes by Epstein-Barr virus (EBV) requires the virus-encoded transactivator EBNA2 and the products of both viral and cellular genes which serve as EBNA2 targets. In this study, we identified BATF as a cellular gene that is up-regulated dramatically within 24 h following the infection of established and primary human B cells with EBV. The transactivation of BATF is mediated by EBNA2 in a B-cell-specific manner and is duplicated in non-EBV-infected B cells by the expression of mammalian Notch proteins. In contrast to other target genes activated by EBNA2, the BATF gene encodes a member of the AP-1 family of transcription factors that functions as a negative regulator of AP-1 activity and as an antagonist of cell growth. A potential role for BATF in promoting EBV latency is supported by studies in which BATF was shown to negatively impact the expression of a BZLF1 reporter gene and to reduce the frequency of lytic replication in latently infected cells. The identification of BATF as a cellular target of EBV provides important new information on how programs of viral and cellular gene expression may be coordinated to promote viral latency and control lytic-cycle entry.


Asunto(s)
Antígenos Nucleares del Virus de Epstein-Barr/metabolismo , Herpesvirus Humano 4/patogenicidad , Proteínas de la Membrana/metabolismo , Factores de Transcripción/metabolismo , Activación Transcripcional , Linfocitos B/virología , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico , Línea Celular , Células Cultivadas , Antígenos Nucleares del Virus de Epstein-Barr/genética , Regulación Viral de la Expresión Génica , Células HeLa , Herpesvirus Humano 4/fisiología , Humanos , Proteínas de la Membrana/genética , Receptores Notch , Factores de Transcripción/genética , Transcripción Genética , Proteínas Virales , Latencia del Virus
15.
Blood ; 100(3): 998-1007, 2002 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-12130514

RESUMEN

The transcription factor PU.1 is required for normal blood cell development. PU.1 regulates the expression of a number of crucial myeloid genes, such as the macrophage colony-stimulating factor (M-CSF) receptor, the granulocyte colony-stimulating factor (G-CSF) receptor, and the granulocyte-macrophage colony-stimulating factor (GM-CSF) receptor. Myeloid cells derived from PU.1(-/-) mice are blocked at the earliest stage of myeloid differentiation, similar to the blast cells that are the hallmark of human acute myeloid leukemia (AML). These facts led us to hypothesize that molecular abnormalities involving the PU.1 gene could contribute to the development of AML. We identified 10 mutant alleles of the PU.1 gene in 9 of 126 AML patients. The PU.1 mutations comprised 5 deletions affecting the DNA-binding domain, and 5 point mutations in 1) the DNA-binding domain (2 patients), 2) the PEST domain (2 patients), and 3) the transactivation domain (one patient). DNA binding to and transactivation of the M-CSF receptor promoter, a direct PU.1 target gene, were deficient in the 7 PU.1 mutants that affected the DNA-binding domain. In addition, these mutations decreased the ability of PU.1 to synergize with PU.1-interacting proteins such as AML1 or c-Jun in the activation of PU.1 target genes. This is the first report of mutations in the PU.1 gene in human neoplasia and suggests that disruption of PU.1 function contributes to the block in differentiation found in AML patients.


Asunto(s)
Leucemia Mieloide/genética , Mutación , Proteínas Proto-Oncogénicas/genética , Transactivadores/genética , Enfermedad Aguda , Sitios de Unión/genética , Diferenciación Celular/genética , Análisis Mutacional de ADN , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Granulocitos/citología , Heterocigoto , Humanos , Leucemia Mieloide/etiología , Leucemia Mieloide/patología , Leucocitos Mononucleares/patología , Unión Proteica/genética , Proteínas Proto-Oncogénicas/farmacología , Proteínas Proto-Oncogénicas/fisiología , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Transactivadores/farmacología , Transactivadores/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...