Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 183
Filtrar
1.
Anal Sci ; 40(3): 445-460, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38112961

RESUMEN

Diabetes mellitus can cause diabetic foot infection (DFI) complications. DFI is generally caused by infection from bacteria and Methicillin-Resistant Staphylococcus aureus (MRSA) which is resistant to several antibiotics. Application therapy of clindamycin (CLY) administration with the oral route has low bioavailability and non-selective distribution of antibiotics towards bacteria intravenously. In this research, CLY was developed into bacterially sensitive microparticles (MPs) which were further incorporated into a separable effervescent microarray patch (SEMAP) system to increase the selective and responsive to DFI-causing bacteria of CLY. To support this formulation, we explore the potential of silver nanoparticles (AgNPs) towards the UV-Vis spectrophotometry method. The analytical method was validated in phosphate-buffered saline (PBS), tryptic soy broth (TSB), and skin tissue to quantify CLY, CLY loaded in microparticle, and SEMAP system. The developed analytical method was suitable for the acceptance criteria of ICH guidelines. The results showed that the correlation coefficients were linear ≥ 0.999. The values of LLOQ towards PBS, TSB, and skin tissue were 2.02 µg/mL, 4.29 µg/mL, and 2.31 µg/mL, respectively. These approaching methods were also found to be accurate and precise without being affected by dilution integrity. The presence of Staphylococcus aureus bacteria culture can produce lipase enzymes that can lysing the microparticle matrix. Drug release studies showed that bacterial infection in the high drug release microparticle sensitive bacteria and high drug retention in ex vivo dermatokinetic in rat skin tissue media. In addition, in vivo studies were required to quantify the CLY inside in further analytical validation methods.


Asunto(s)
Nanopartículas del Metal , Staphylococcus aureus Resistente a Meticilina , Animales , Ratas , Clindamicina , Colorimetría , Penfluridol , Plata , Antibacterianos/farmacología , Espectrofotometría , Pruebas de Sensibilidad Microbiana
2.
Diabetes ; 72(1): 126-134, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36256885

RESUMEN

Despite significant progress in understanding the pathogenesis of type 2 diabetes (T2D), the condition remains difficult to manage. Hence, new therapeutic options targeting unique mechanisms of action are required. We have previously observed that elevated skeletal muscle succinyl CoA:3-ketoacid CoA transferase (SCOT) activity, the rate-limiting enzyme of ketone oxidation, contributes to the hyperglycemia characterizing obesity and T2D. Moreover, we identified that the typical antipsychotic agent pimozide is a SCOT inhibitor that can alleviate obesity-induced hyperglycemia. We now extend those observations here, using computer-assisted in silico modeling and in vivo pharmacology studies that highlight SCOT as a noncanonical target shared among the diphenylbutylpiperidine (DPBP) drug class, which includes penfluridol and fluspirilene. All three DPBPs tested (pimozide, penfluridol, and fluspirilene) improved glycemia in obese mice. While the canonical target of the DPBPs is the dopamine 2 receptor, studies in obese mice demonstrated that acute or chronic treatment with a structurally unrelated antipsychotic dopamine 2 receptor antagonist, lurasidone, was devoid of glucose-lowering actions. We further observed that the DPBPs improved glycemia in a SCOT-dependent manner in skeletal muscle, suggesting that this older class of antipsychotic agents may have utility in being repurposed for the treatment of T2D.


Asunto(s)
Antipsicóticos , Diabetes Mellitus Tipo 2 , Hiperglucemia , Animales , Ratones , Antipsicóticos/farmacología , Antipsicóticos/uso terapéutico , Coenzima A Transferasas , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Dopamina , Fluspirileno/farmacología , Hiperglucemia/tratamiento farmacológico , Ratones Obesos , Penfluridol/farmacología , Pimozida/farmacología , Receptores Dopaminérgicos/metabolismo
3.
Cell Commun Signal ; 20(1): 105, 2022 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-35842652

RESUMEN

BACKGROUND: Penfluridol (PF) is an FDA-approved antipsychotic drug that has recently been shown to have anticancer activity. However, the anticancer effects and underlying mechanisms of PF are not well-established in gallbladder cancer (GBC). METHODS: The anticancer efficacy of PF on GBC was investigated via a series of cell functions experiments, including cell viability, colony formation, apoptosis assays, and so on. The corresponding signaling changes after PF treatment were explored by western blotting. Then, nude mice were utilized to study and test the anticancer activity of PF in vivo. Besides, glucose consumption and lactic production assays were used to detect the glycolysis alteration. RESULTS: In this study, we discovered that PF greatly inhibited the proliferation and invasion ability of GBC cells (GBCs). The glucose consumption and lactic generation ability of GBCs were dramatically elevated following PF treatment. Additionally, we discovered that inhibiting glycolysis could improve PF's anticancer efficacy. Further studies established that the activation of the AMPK/PFKFB3 signaling pathway medicated glycolysis after PF treatment. We proved mechanistically that inhibition of AMPK/PFKFB3 singling pathway mediated glycolysis was a potential synergetic strategy to improve the anticancer efficacy of PF on GBC. CONCLUSIONS: By inhibiting AMPK, the anticancer effects of PF on GBCs were amplified. As a result, our investigations shed new light on the possibility of repurposing PF as an anticancer drug for GBC, and AMPK inhibition in combination with PF may represent a novel therapeutic strategy for GBC. Video abstract.


Asunto(s)
Neoplasias de la Vesícula Biliar , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Apoptosis , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Neoplasias de la Vesícula Biliar/diagnóstico , Neoplasias de la Vesícula Biliar/tratamiento farmacológico , Neoplasias de la Vesícula Biliar/metabolismo , Glucosa/metabolismo , Glucólisis , Ratones , Ratones Desnudos , Penfluridol/farmacología
4.
Cell Death Dis ; 13(4): 400, 2022 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-35461314

RESUMEN

Renal cell carcinoma (RCC) is one of the most lethal genitourinary malignancies with poor prognoses, since it is largely resistant to chemotherapy, radiotherapy, and targeted therapy. The persistence of cancer stem cells (CSCs) is the major cause of treatment failure with RCC. Recent evidence showed that dopamine receptor D2 (DRD2)-targeting antipsychotic drugs such as penfluridol exert oncostatic effects on several cancer types, but the effect of penfluridol on RCC remains unknown. Here, we uncovered penfluridol suppressed in vitro cell growth and in vivo tumorigenicity of various RCC cell lines (Caki-1, 786-O, A498, and ACHN) and enhanced the Sutent (sunitinib)-triggered growth inhibition on clear cell (cc)RCC cell lines. Mechanistically, upregulation of endoplasmic reticulum (ER) stress-induced unfolded protein response (UPR) was critical for autophagy-mediated apoptosis induced by penfluridol. Transcriptional inhibition of OCT4 and Nanog via inhibiting GLI1 was important for penfluridol-induced stemness and proliferation inhibition. The anticancer activities of penfluridol on ccRCC partially occurred through DRD2. In clinical ccRCC specimens, positive correlations of DRD2 with GLI1, OCT4, and Nanog were observed and their expressions were correlated with worse prognoses. Summarizing, DRD2 antagonists such as penfluridol induce UPR signaling and suppress the GLI1/OCT4/Nanog axis in ccRCC cells to reduce their growth through inducing autophagy-mediated apoptosis and stemness inhibition. These drugs can be repurposed as potential agents to treat ccRCC patients.


Asunto(s)
Antipsicóticos , Muerte Celular Autofágica , Carcinoma de Células Renales , Neoplasias Renales , Receptores de Dopamina D2 , Antipsicóticos/farmacología , Apoptosis , Carcinoma de Células Renales/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Neoplasias Renales/tratamiento farmacológico , Masculino , Penfluridol/farmacología , Penfluridol/uso terapéutico , Proteína con Dedos de Zinc GLI1
5.
Arthritis Res Ther ; 24(1): 27, 2022 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-35045889

RESUMEN

BACKGROUND: Penfluridol, isolated from an FDA-approved small-molecule drug library as an inhibitor of tumor necrosis factor α (TNFα)-stimulated NF-κB activation, is clinically used to treat chronic schizophrenia and related disorders. This study is aimed to investigate the therapeutic effect of penfluridol on TNFα-stimulated inflammatory autoimmune diseases, particularly inflammatory arthritis. METHODS: Various in vitro studies to confirm the inhibitory effect of penfluridol on TNFα-induced NF-κB activity in bone marrow-derived macrophages or Raw 264.7 macrophage cell line. In vivo studies assessed the therapeutic effects of penfluridol in various disease models, including TNFα transgenic mice, collagen-induced arthritis, DSS-induced colitis, and TNBS-induced colitis. Identification and characterization of the binding of penfluridol to acid sphingomyelinase using bioinformatics and drug affinity responsive target stability assay. Acid sphingomyelinase activity assays to reveal penfluridol-mediated inhibition of acid sphingomyelinase activity. siRNA knockdown experiments to illustrate the dependence of penfluridol's anti-TNF activity on acid sphingomyelinase. RESULTS: Penfluridol effectively inhibited TNFα-induced NF-κB activation in vitro and alleviated the severity of arthritis and colitis in vivo. Mechanistic studies revealed that penfluridol bound to acid sphingomyelinase and inhibited its activation. In addition, knockdown of acid sphingomyelinase largely abolished the inhibitory effects of penfluridol on TNFα-induced inflammatory cytokine production. Furthermore, penfluridol suppressed the differentiation of spleen naive CD4+T cells to TH1 and TH17 and inhibited M1 macrophage polarization. CONCLUSION: This study provides the rationale for the possible innovative use of penfluridol as a newly identified small-molecule drug for TNFα-driven diseases, such as inflammatory arthritis and colitis.


Asunto(s)
Enfermedades Autoinmunes , Penfluridol , Animales , Enfermedades Autoinmunes/tratamiento farmacológico , Ratones , FN-kappa B/metabolismo , Esfingomielina Fosfodiesterasa , Inhibidores del Factor de Necrosis Tumoral , Factor de Necrosis Tumoral alfa/metabolismo
6.
J Psychopharmacol ; 36(2): 223-231, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34702106

RESUMEN

AIM: In this study, we sought to determine clinical outcomes at 1 year for patients prescribed penfluridol in an inner London National Health Service Trust. Using noninterventional data, we describe the use, effectiveness and safety of this treatment modality. RESULTS: We retrospectively followed up 17 patients prescribed penfluridol as part of routine clinical practice. All patients took penfluridol once weekly. Of these patients, 12 (70.6%) were considered treatment resistant. The average duration of illness for this cohort was 10 years (SD = 6.7). At 1 year, nine (53%) patients remained on treatment. Median survival time was not reached at 1-year follow-up; mean time on penfluridol was 251 days (95% confidence interval (CI), 184-318). The mean number of admissions to hospital in the year following penfluridol initiation was 0.6 compared with 0.8, 1 year before initiation (p = 0.465). The median number of bed days 1 year before penfluridol initiation was 24, whereas in the year following penfluridol initiation, it was 0 (p = 0.514). CLINICAL IMPLICATIONS: Although penfluridol is unlicensed in the United Kingdom, limited data suggest that this long-acting oral therapy has the potential to be used safely and effectively for the treatment of psychotic disorders. However, more data are required to establish the place of penfluridol and other potential long-acting oral antipsychotic formulations in the treatment of psychotic disorders.


Asunto(s)
Antipsicóticos/administración & dosificación , Hospitalización/estadística & datos numéricos , Penfluridol/administración & dosificación , Trastornos Psicóticos/tratamiento farmacológico , Administración Oral , Adulto , Anciano , Antipsicóticos/efectos adversos , Preparaciones de Acción Retardada , Femenino , Estudios de Seguimiento , Humanos , Londres , Masculino , Persona de Mediana Edad , Penfluridol/efectos adversos , Estudios Retrospectivos , Resultado del Tratamiento
7.
Psychiatry Res ; 304: 114147, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34371297

RESUMEN

Psychiatric disorders, including schizophrenic spectrum disorders, are common in People Who Use Drugs (PWUD). Promoting adherence to medication among PWUD with dual diagnosis is challenging. We present the case of a treatment-disrupted patient suffering from schizophrenia with co-occuring multiple drug dependence to whom penfluridol - an oral long acting typical antipsychotic - was proposed at the Supervised Drug Consumption Room (SDCR) of Paris. Penfluridol quickly improved patient's psychotic symptoms, increased engagement in addiction care and helped maintaining the patient in "Housing First" program. In harm reduction structure, penfluridol can be seen as a "hook treatment" while maintaining therapeutic alliance and favoring patients' engagement in specific care.


Asunto(s)
Antipsicóticos , Esquizofrenia , Trastornos Relacionados con Sustancias , Antipsicóticos/uso terapéutico , Reducción del Daño , Humanos , Penfluridol/uso terapéutico , Esquizofrenia/complicaciones , Esquizofrenia/tratamiento farmacológico , Trastornos Relacionados con Sustancias/tratamiento farmacológico
8.
Cell Oncol (Dordr) ; 44(5): 1087-1103, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34319576

RESUMEN

PURPOSE: Metastasis of lung adenocarcinoma (LADC) is a crucial factor determining patient survival. Repurposing of the antipsychotic agent penfluridol has been found to be effective in the inhibition of growth of various cancers. As yet, however, the anti-metastatic effect of penfluridol on LADC has rarely been investigated. Herein, we addressed the therapeutic potential of penfluridol on the invasion/metastasis of LADC cells harboring different epidermal growth factor receptor (EGFR) mutation statuses. METHODS: MTS viability, transwell migration and invasion, and tumor endothelium adhesion assays were employed to determine cytotoxic and anti-metastatic effects of penfluridol on LADC cells. Protease array, Western blot, immunohistochemistry (IHC), immunofluorescence (IF) staining, and expression knockdown by shRNA or exogenous overexpression by DNA plasmid transfection were performed to explore the underlying mechanisms, both in vitro and in vivo. RESULTS: We found that nontoxic concentrations of penfluridol reduced the migration, invasion and adhesion of LADC cells. Protease array screening identified matrix metalloproteinase-12 (MMP-12) as a potential target of penfluridol to modulate the motility and adhesion of LADC cells. In addition, we found that MMP-12 exhibited the most significantly adverse prognostic effect in LADC among 39 cancer types. Mechanistic investigations revealed that penfluridol inhibited the urokinase plasminogen activator (uPA)/uPA receptor/transforming growth factor-ß/Akt axis to downregulate MMP-12 expression and, subsequently, reverse MMP-12-induced epithelial-mesenchymal transition (EMT). Subsequent analysis of clinical LADC samples revealed a positive correlation between MMP12 and mesenchymal-related gene expression levels. A lower survival rate was found in LADC patients with a SNAl1high/MMP12high profile compared to those with a SNAl1low/MMP12low profile. CONCLUSIONS: Our results indicate that MMP-12 may serve as a useful biomarker for predicting LADC progression and as a promising penfluridol target for treating metastatic LADC.


Asunto(s)
Adenocarcinoma del Pulmón/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Neoplasias Pulmonares/metabolismo , Metaloproteinasa 12 de la Matriz/metabolismo , Penfluridol/farmacología , Proteínas/metabolismo , Transducción de Señal/efectos de los fármacos , Células A549 , Adenocarcinoma del Pulmón/tratamiento farmacológico , Adenocarcinoma del Pulmón/genética , Animales , Línea Celular Tumoral , Reposicionamiento de Medicamentos/métodos , Transición Epitelial-Mesenquimal/genética , Humanos , Estimación de Kaplan-Meier , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Masculino , Metaloproteinasa 12 de la Matriz/genética , Ratones Endogámicos NOD , Ratones SCID , Metástasis de la Neoplasia , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
9.
CNS Drugs ; 35(4): 451-460, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33837915

RESUMEN

BACKGROUND: Many patients with schizophrenia discontinue antipsychotic medication, frequently with adverse outcomes. Although different antipsychotic formulations are associated with different times to discontinuation, not much is known about discontinuation rates with oral-weekly formulations. Such a formulation of penfluridol is available in both the Netherlands and several other countries. OBJECTIVES: We aimed to investigate the impact of antipsychotic formulations on time to discontinuation, especially the oral-weekly formulation. METHODS: In a large, registry-based, retrospective cohort study from 1 January 2013 to 31 December 2016, we determined the time to medication discontinuation during the follow-up period with antipsychotic formulations, including oral-daily, oral-weekly, depot, or a combination of these. Patients with schizophrenia aged between 18 and 69 years were included and stratified according to the duration of recent antipsychotic use (taking the same formulation for ≤ 60 days or > 60 days before follow-up: short-term or long-term recent antipsychotic use). Medication discontinuation was defined as discontinuation of current antipsychotic formulation. RESULTS: Overall, 8257 patients were included for analyses, with 80% of patients discontinuing antipsychotic medication. Time to discontinuation was longer in those with long-term recent antipsychotic use before the follow-up period and longest for oral-daily formulations. Patterns for discontinuation of oral-weekly and depot formulations were similar, regardless of the duration of recent antipsychotic use before follow-up. More prior discontinuations were associated with shorter time to discontinuation. CONCLUSIONS: Time to discontinuation differed considerably between formulations. The duration of recent antipsychotic use was a strong predictor of time to discontinuation. While oral-daily formulations had the longest time to discontinuation in the long-term recent antipsychotic use group, discontinuation trends were similar for oral-weekly and depot formulations. An oral-weekly formulation, whose administration route is noninvasive, might therefore be considered an alternative to depot formulations.


Asunto(s)
Preparaciones de Acción Retardada , Duración de la Terapia , Selección de Paciente , Penfluridol , Esquizofrenia , Administración Oral , Adulto , Antipsicóticos/administración & dosificación , Antipsicóticos/farmacología , Preparaciones de Acción Retardada/administración & dosificación , Preparaciones de Acción Retardada/farmacología , Femenino , Humanos , Masculino , Cumplimiento de la Medicación/psicología , Cumplimiento de la Medicación/estadística & datos numéricos , Administración del Tratamiento Farmacológico/normas , Administración del Tratamiento Farmacológico/estadística & datos numéricos , Persona de Mediana Edad , Países Bajos/epidemiología , Penfluridol/administración & dosificación , Penfluridol/farmacología , Pronóstico , Sistema de Registros/estadística & datos numéricos , Esquizofrenia/diagnóstico , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/epidemiología , Psicología del Esquizofrénico
10.
Microbiologyopen ; 10(1): e1148, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33345466

RESUMEN

The bacterium Enterococcus faecalis has increasingly attracted global attention as an important opportunistic pathogen due to its ability to form biofilms that are known to increase drug resistance. However, there are still no effective antibiofilm drugs in clinical settings. Here, by drug repurposing, we investigated the antibacterial activity of penfluridol (PF), an oral long-acting antipsychotic approved by the FDA, against E. faecalis type strain and its clinical isolates. It was found that PF inhibited the growth of E. faecalis planktonic cells with the MIC and MBC of 7.81 µg/ml and 15.63 ~ 62.50 µg/ml, respectively. Moreover, PF could significantly prevent the biofilm formation of E. faecalis at the concentration of 1 × MIC. Furthermore, PF significantly eradicated 24 h pre-formed biofilms of E. faecalis in a dose-dependent manner, with a concentration range of 1 × MIC to 8 × MIC. Here, through the checkerboard method with other tested conventional antibiotics, we also determined that gentamycin, penicillin G, and amikacin showed partial synergistic antibacterial effects with PF. Also, PF showed almost no hemolysis on human erythrocytes. In a mouse peritonitis model, a single dose of 20 mg/kg of PF treatment could significantly reduce the bacterial colonization in the liver (~5-fold reduction) and spleen (~3-fold reduction). In conclusion, these findings indicated that after structural optimization, PF has the potential as a new antibacterial agent against E. faecalis.


Asunto(s)
Antibacterianos/farmacología , Reposicionamiento de Medicamentos/métodos , Enterococcus faecalis/efectos de los fármacos , Infecciones por Bacterias Grampositivas/tratamiento farmacológico , Penfluridol/farmacología , Animales , Biopelículas/crecimiento & desarrollo , Modelos Animales de Enfermedad , Enterococcus faecalis/crecimiento & desarrollo , Eritrocitos/efectos de los fármacos , Femenino , Humanos , Ratones , Ratones Endogámicos ICR , Pruebas de Sensibilidad Microbiana , Viabilidad Microbiana/efectos de los fármacos , Infecciones Oportunistas/tratamiento farmacológico , Peritonitis/tratamiento farmacológico , Peritonitis/microbiología
11.
Anat Rec (Hoboken) ; 304(3): 520-530, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32470200

RESUMEN

Penfluridol, a commonly used antipsychotic agent in a clinical setting, exhibits potential anticancer properties against various human malignancies. Here, we investigated the effect of penfluridol on the biological behavior of colorectal cancer (CRC) cells. Cell viability and clonogenic potential were detected by the cell counting kit-8 and colony formation assay. The cell apoptosis and cell cycle distribution were quantified through flow cytometry. Caspase-3 activity, glucose consumption, lactate production, and intracellular ATP levels were evaluated using the corresponding commercial detection kits. The protein levels of related genes were detected through western blotting. Mitochondrial membrane potential was detected using JC-1 staining. A CRC xenograft tumor model was used to validate the antitumor activity of penfluridol in vivo. Penfluridol reduced cell survival and promoted apoptotic cell death effectively through the mitochondria-mediated intrinsic pathway in a dose-dependent manner. Furthermore, the process of glycolysis in HCT-116 and HT-29 cells was inhibited upon penfluridol treatment, as evidenced by the decrease in glucose consumption, lactate production, and intracellular ATP levels. Further mechanistic studies revealed that penfluridol influenced cell apoptosis and glycolysis in CRC cells by downregulating hexokinase-2 (HK-2). The proapoptotic effect and glycolytic inhibition-induced by penfluridol were effectively reversed by HK-2 overexpression. Consistent with in vitro results, penfluridol could also suppress tumor growth and trigger apoptosis in vivo. Penfluridol triggers mitochondrial-mediated apoptosis and induces glycolysis inhibition via modulating HK-2 in CRC and provides a theoretical basis to support penfluridol as a repurposed drug for CRC patients.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias Colorrectales/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Glucólisis/efectos de los fármacos , Hexoquinasa/metabolismo , Mitocondrias/efectos de los fármacos , Penfluridol/farmacología , Animales , Antineoplásicos/farmacología , Caspasa 3/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Neoplasias Colorrectales/patología , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Mitocondrias/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Mol Oncol ; 14(12): 3121-3134, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32896947

RESUMEN

More effective therapy for patients with either muscle-invasive or high-risk non-muscle-invasive urothelial carcinoma of the bladder (UCB) is an unmet clinical need. For this, drug repositioning of clinically approved drugs represents an interesting approach. By repurposing existing drugs, alternative anticancer therapies can be introduced in the clinic relatively fast, because the safety and dosing of these clinically approved pharmacological agents are generally well known. Cationic amphiphilic drugs (CADs) dose-dependently decreased the viability of a panel of human UCB lines in vitro. CADs induced lysosomal puncta formation, a hallmark of lysosomal leakage. Intravesical instillation of the CAD penfluridol in an orthotopic mouse xenograft model of human UCB resulted in significantly reduced intravesical tumor growth and metastatic progression. Furthermore, treatment of patient-derived ex vivo cultured human UCB tissue caused significant partial or complete antitumor responses in 97% of the explanted tumor tissues. In conclusion, penfluridol represents a promising treatment option for bladder cancer patients and warrants further clinical evaluation.


Asunto(s)
Antineoplásicos/uso terapéutico , Tensoactivos/uso terapéutico , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Cationes , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Clonales , Humanos , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , Metástasis de la Neoplasia , Penfluridol/farmacología , Penfluridol/uso terapéutico , Tensoactivos/farmacología , Neoplasias de la Vejiga Urinaria/patología , Urotelio/efectos de los fármacos , Urotelio/patología , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Int J Mol Sci ; 21(3)2020 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-31979394

RESUMEN

: Metastasis is considered a major burden in cancer, being responsible for more than 90% of cancer-related deaths. Tumor angiogenesis is one of the main processes that lead to tumor metastasis. Penfluridol is a classic and commonly used antipsychotic drug, which has a great ability to cross the blood-brain barrier. Recent studies have revealed that penfluridol has significant anti-cancer activity in diverse tumors, such as metastatic breast cancer and glioblastoma. Here, we aim to identify the effect of low doses of penfluridol on tumor microenvironment and compare it with its effect on tumor cells. Although low concentration of penfluridol was not toxic for endothelial cells, it blocked angiogenesis in vitro and in vivo. In vitro, penfluridol inhibited VEGF-induced primary endothelial cell migration and tube formation, and in vivo, it blocked VEGF- and FGF-induced angiogenesis in the matrigel plug assay. VEGF-induced VEGFR2 phosphorylation and the downstream p38 and ERK signaling pathways were not affected in endothelial cells, although VEGF-induced Src and Akt activation were abrogated by penfluridol treatment. When cancer cells were treated with the same low concentration of penfluridol, basal Src activation levels were mildly impaired, thus impacting their cell migration and wound healing efficiency. The potential of cancer-induced paracrine effect on endothelial cells was explored, although that did not seem to be a player for angiogenesis. Overall, our data demonstrates that low penfluridol levels, similar to the ones clinically used for anti-psychotic conditions, suppress angiogenic efficiency in the tumor microenvironment.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Neoplasias de la Mama/metabolismo , Penfluridol/farmacología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Antipsicóticos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Colágeno , Combinación de Medicamentos , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Laminina , Ratones , Ratones Endogámicos C57BL , Penfluridol/uso terapéutico , Proteoglicanos , Transducción de Señal/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
14.
Gastroenterology ; 158(5): 1433-1449.e27, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31786131

RESUMEN

BACKGROUND & AIMS: Prolactin (PRL) signaling is up-regulated in hormone-responsive cancers. The PRL receptor (PRLR) is a class I cytokine receptor that signals via the Janus kinase (JAK)-signal transducer and activator of transcription and mitogen-activated protein kinase pathways to regulate cell proliferation, migration, stem cell features, and apoptosis. Patients with pancreatic ductal adenocarcinoma (PDAC) have high plasma levels of PRL. We investigated whether PRLR signaling contributes to the growth of pancreatic tumors in mice. METHODS: We used immunohistochemical analyses to compare levels of PRL and PRLR in multitumor tissue microarrays. We used structure-based virtual screening and fragment-based drug discovery to identify compounds likely to bind PRLR and interfere with its signaling. Human pancreatic cell lines (AsPC-1, BxPC-3, Panc-1, and MiaPaCa-2), with or without knockdown of PRLR (clustered regularly interspaced short palindromic repeats or small hairpin RNA), were incubated with PRL or penfluridol and analyzed in proliferation and spheroid formation. C57BL/6 mice were given injections of UNKC-6141 cells, with or without knockdown of PRLR, into pancreas, and tumor development was monitored for 4 weeks, with some mice receiving penfluridol treatment for 21 days. Human pancreatic tumor tissues were implanted into interscapular fat pads of NSG mice, and mice were given injections of penfluridol daily for 28 days. Nude mice were given injections of Panc-1 cells, xenograft tumors were grown for 2 weeks, and mice were then given intraperitoneal penfluridol for 35 days. Tumors were collected from mice and analyzed by histology, immunohistochemistry, and immunoblots. RESULTS: Levels of PRLR were increased in PDAC compared with nontumor pancreatic tissues. Incubation of pancreatic cell lines with PRL activated signaling via JAK2-signal transducer and activator of transcription 3 and extracellular signal-regulated kinase, as well as formation of pancospheres and cell migration; these activities were not observed in cells with PRLR knockdown. Pancreatic cancer cells with PRLR knockdown formed significantly smaller tumors in mice. We identified several diphenylbutylpiperidine-class antipsychotic drugs as agents that decreased PRL-induced JAK2 signaling; incubation of pancreatic cancer cells with these compounds reduced their proliferation and formation of panco spheres. Injections of 1 of these compounds, penfluridol, slowed the growth of xenograft tumors in the different mouse models, reducing proliferation and inducing autophagy of the tumor cells. CONCLUSIONS: Levels of PRLR are increased in PDAC, and exposure to PRL increases proliferation and migration of pancreatic cancer cells. Antipsychotic drugs, such as penfluridol, block PRL signaling in pancreatic cancer cells to reduce their proliferation, induce autophagy, and slow the growth of xenograft tumors in mice. These drugs might be tested in patients with PDAC.


Asunto(s)
Antipsicóticos/farmacología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Neoplasias Pancreáticas/tratamiento farmacológico , Penfluridol/farmacología , Prolactina/metabolismo , Receptores de Prolactina/antagonistas & inhibidores , Animales , Antipsicóticos/uso terapéutico , Autofagia/efectos de los fármacos , Carcinoma Ductal Pancreático/sangre , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Descubrimiento de Drogas , Técnicas de Silenciamiento del Gen , Humanos , Inyecciones Intraperitoneales , Janus Quinasa 2/metabolismo , Masculino , Ratones , Páncreas/patología , Neoplasias Pancreáticas/sangre , Neoplasias Pancreáticas/patología , Penfluridol/uso terapéutico , Prolactina/sangre , Receptores de Prolactina/genética , Receptores de Prolactina/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Esferoides Celulares , Análisis de Matrices Tisulares , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Biomed Pharmacother ; 121: 109598, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31733572

RESUMEN

Lung cancer remains the leading cause of cancer mortality because of highly malignant and metastatic potential. The current status of lung cancer treatment is limited, and more treatment options are needed. Interesting, antipsychotic drugs have been reported to show anti-cancer effects. In this present study, we investigated the anticancer potential of penfluridol (PF), an anti-schizophrenic drug, in lung cancer and its underlying mechanism in vitro and in vivo. In vitro, it could inhibit the viability of various lung cancer cells with G0/G1 phase arrest via increasing the expression level of p21/p27 and decreasing the expression levels of cyclin-CDK complex. Meanwhile, cell-cycle arrest causes DNA repair in the nucleus, which was associated with the upregulation of H2A.X and p-H2A.X. Moreover, PF could also decrease mitochondrial membrane potential and increase reactive oxygen species levels in the lung cancer cells. These results implied that PF might induce the mitochondria-mediated intrinsic apoptosis. In addition, PF inhibits the migration and invasion of lung cancer cells via downregulation of FAK-MMP signaling. In vivo, oral administration of PF at concentration of 10 mg/kg inhibited tumor growth in A549 xenograft model. Notably, PF is an approved drug and the price is exceedingly cheap, so this study demonstrates the potential of PF to treat lung cancer.


Asunto(s)
Antipsicóticos/uso terapéutico , Apoptosis/efectos de los fármacos , Fase G1/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Penfluridol/uso terapéutico , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Células A549 , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Antipsicóticos/farmacología , Apoptosis/fisiología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/fisiología , Relación Dosis-Respuesta a Droga , Femenino , Fase G1/fisiología , Inhibidores de Crecimiento/farmacología , Inhibidores de Crecimiento/uso terapéutico , Humanos , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica/patología , Penfluridol/farmacología , Fase de Descanso del Ciclo Celular/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
16.
Molecules ; 24(20)2019 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-31614431

RESUMEN

Penfluridol has robust antipsychotic efficacy and is a first-generation diphenylbutylpiperidine. Its effects last for several days after a single oral dose and it can be administered once a week to provide better compliance and symptom control. Recently; strong antitumour effects for penfluridol were discovered in various cancer cell lines; such as breast; pancreatic; glioblastoma; and lung cancer cells via several distinct mechanisms. Therefore; penfluridol has drawn much attention as a potentially novel anti-tumour agent. In addition; the anti-cancer effects of penfluridol have been demonstrated in vivo: results showed slight changes in the volume and weight of organs at doses tested in animals. This paper outlines the potential for penfluridol to be developed as a next-generation anticancer drug.


Asunto(s)
Antineoplásicos/uso terapéutico , Reposicionamiento de Medicamentos , Neoplasias/tratamiento farmacológico , Penfluridol/uso terapéutico , Línea Celular Tumoral , Humanos
17.
J Biomed Sci ; 26(1): 63, 2019 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-31470848

RESUMEN

BACKGROUND: Chemotherapy is the main treatment for acute myeloid leukemia (AML), but the cure rates for AML patients remain low, and the notorious adverse effects of chemotherapeutic drugs drastically reduce the life quality of patients. Penfluridol, a long-acting oral antipsychotic drug, has an outstanding safety record and exerts oncostatic effects on various solid tumors. Until now, the effect of penfluridol on AML remains unknown. METHODS: AML cell lines harboring wild-type (WT) Fms-like tyrosine kinase 3 (FLT3) and internal tandem duplication (ITD)-mutated FLT3 were used to evaluate the cytotoxic effects of penfluridol by an MTS assay. A flow cytometric analysis and immunofluorescence staining were employed to determine the cell-death phenotype, cell cycle profile, and reactive oxygen species (ROS) and acidic vesicular organelle (AVO) formation. Western blotting and chemical inhibitors were used to explore the underlying mechanisms involved in penfluridol-mediated cell death. RESULTS: We observed that penfluridol concentration-dependently suppressed the cell viability of AML cells with FLT3-WT (HL-60 and U937) and FLT3-ITD (MV4-11). We found that penfluridol treatment not only induced apoptosis as evidenced by increases of nuclear fragmentation, the sub-G1 populations, poly (ADP ribose) polymerase (PARP) cleavage, and caspase-3 activation, but also triggered autophagic responses, such as the light chain 3 (LC3) turnover and AVO formation. Interestingly, blocking autophagy by the pharmacological inhibitors, 3-methyladenine and chloroquine, dramatically enhanced penfluridol-induced apoptosis, indicating the cytoprotective role of autophagy in penfluridol-treated AML cells. Mechanistically, penfluridol-induced apoptosis occurred through activating protein phosphatase 2A (PP2A) to suppress Akt and mitogen-activated protein kinase (MAPK) activities. Moreover, penfluridol's augmentation of intracellular ROS levels was critical for the penfluridol-induced autophagic response. In the clinic, we observed that patients with AML expressing high PP2A had favorable prognoses. CONCLUSIONS: These findings provide a rationale for penfluridol being used as a PP2A activator for AML treatment, and the combination of penfluridol with an autophagy inhibitor may be a novel strategy for AML harboring FLT3-WT and FLT3-ITD.


Asunto(s)
Antipsicóticos/farmacología , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Leucemia Mieloide Aguda/tratamiento farmacológico , Penfluridol/farmacología , Especies Reactivas de Oxígeno/metabolismo , Tirosina Quinasa 3 Similar a fms/metabolismo , Línea Celular Tumoral , Citoprotección/efectos de los fármacos , Células HL-60 , Humanos , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteína Fosfatasa 2/genética , Proteína Fosfatasa 2/metabolismo , Transducción de Señal , Células U937
18.
Cell Death Dis ; 10(8): 538, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31308361

RESUMEN

Anticancer chemotherapeutic drugs mainly trigger apoptosis induction to eliminate malignant cells. However, many cancer cells are chemoresistant because of defective apoptosis induction. Targeting the autophagic pathway is currently regarded as an alternative strategy for cancer drug discovery. Penfluridol, an antipsychotic drug, has been reported to exert oncostatic effects, but the effect of penfluridol on lung cancer remains unknown. Herein, the antitumor activity of penfluridol was determined in vitro in non-small-cell lung cancer (NSCLC) cell lines using MTS, plate clonogenic, and transwell migration assays and in vivo in an orthotopic xenograft model. Flow cytometry, holotomographic microscopy, immunofluorescence, and immunohistochemistry were employed to determine the cell-death phenotype induced by penfluridol in vitro and in vivo. Western blotting and genetic knockdown by small interfering RNA were performed to explore the underlying mechanisms involved in penfluridol-mediated cell death. We uncovered that penfluridol inhibited the viability and motility of NSCLC cells in vitro and in vivo. Penfluridol induced nonapoptotic cell death by blocking autophagic flux and inducing accumulation of autophagosome-related protein, light chain 3 (LC3) B-II, in HCC827 and A549 NSCLC cells, and in an A549 orthotopic xenograft tumor model. Autophagosome accumulation-induced cell viability inhibition by penfluridol was mainly attributed to ATP energy deprivation. Moreover, we observed that patients with lung tumors expressing high LC3B had longer overall and disease-free survival times. Mechanistically, upregulation of endoplasmic reticulum (ER) stress-induced unfolded protein response (UPR) pathways and activation of p38 mitogen-activated protein kinase (MAPK) were critical for penfluridol-induced autophagosome accumulation. Our findings identify that penfluridol acts as an inducer of ER stress and p38 MAPK activation, which led to UPR-mediated nonapoptotic cell death via autophagosome accumulation-caused energy loss. Penfluridol is clinically used for schizophrenia, and our study results strongly support penfluridol as a repurposed drug for treating NSCLC.


Asunto(s)
Adenosina Trifosfato/metabolismo , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Autofagosomas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Neoplasias Pulmonares/metabolismo , Penfluridol/farmacología , Respuesta de Proteína Desplegada/efectos de los fármacos , Células A549 , Animales , Antineoplásicos/uso terapéutico , Autofagia/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Reposicionamiento de Medicamentos/métodos , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Penfluridol/uso terapéutico , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Int J Pharm Compd ; 23(2): 113-119, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31085776

RESUMEN

Schizophrenia is a serious, disabling, enduring, and relapsing mental illness which causes problems with the ability to think, feel, and perceive things clearly. One cause of relapse and readmission to a hospital is poor compliance with antipsychotic medication, often due to its adverse effects. Schizophrenia may also affect a person's insight, interfering with their ability to appreciate the benefit of taking medication long term. The relapse rate is significantly higher in those who have discontinued antipsychotic medication. Penfluridol is an unusual, potent, long-acting, first-generation, oral antipsychotic agent indicated for the treatment of chronic schizophrenia, acute psychosis, and Tourette syndrome. It may be considered a depot medication, as it is administered once a week. Despite this positive analysis, and the unique added value of this medication to psychotic, non-compliant patients, Janssen-Cilag, the sole manufacturer of penfluridol worldwide, decided to stop production in 2009. This decision forced many psychotic patients worldwide to abandon the once-a-week convenient treatment and to replace it with a daily, oral treatment or a depot injection. Because penfluridol is no longer commercially available, it has created an opportunity for compounding pharmacists worldwide to accept this challenge and offer this medication to psychiatrists because of its clinical therapeutics. For the past 5 years, penfluridol has been available to compounding pharmacists in Israel and has received favorable feedback from physicians and patients.


Asunto(s)
Antipsicóticos/farmacología , Penfluridol , Esquizofrenia , Administración Oral , Antipsicóticos/química , Humanos , Inyecciones , Penfluridol/farmacología
20.
Sci Rep ; 9(1): 5066, 2019 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-30911062

RESUMEN

Paclitaxel is a first line chemotherapeutic agent for the patients with metastatic breast cancer. But inherited or acquired resistance to paclitaxel leads to poor response rates in a majority of these patients. To identify mechanisms of paclitaxel resistance, we developed paclitaxel resistant breast cancer cell lines, MCF-7 and 4T1 by continuous exposure to paclitaxel for several months. Western blot analysis showed increased expression of HER2 and ß-catenin pathway in resistant cell lines as compared to parent cells. Hence, we hypothesized that HER2/ß-catenin mediates paclitaxel resistance in breast cancer and suppression of HER2/ß-catenin signaling could overcome paclitaxel resistance. Our data showed that penfluridol (PFL) treatment significantly reduced the survival of paclitaxel-resistant cells. Western blot analysis revealed that PFL treatment suppressed HER2, as well as, ß-catenin pathway. In vivo data confirmed that PFL significantly potentiated tumor growth suppressive effects of paclitaxel in an orthotropic breast cancer model. In addition, tumors from paclitaxel and PFL-treated mice showed reduced HER2 and ß-catenin expression, along with increased apoptosis. Taken together our results demonstrate a novel role of HER2/ß-catenin in paclitaxel resistance and open up new avenues for application of PFL as a therapeutic option for overcoming paclitaxel resistance.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Resistencia a Antineoplásicos/efectos de los fármacos , Penfluridol/uso terapéutico , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Apoptosis/efectos de los fármacos , Apoptosis/genética , Neoplasias de la Mama/metabolismo , Femenino , Humanos , Ratones , Estadificación de Neoplasias , Paclitaxel/uso terapéutico , Penfluridol/administración & dosificación , Penfluridol/efectos adversos , Receptor ErbB-2/metabolismo , Transducción de Señal , beta Catenina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA