Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Sci Rep ; 10(1): 11713, 2020 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-32678217

RESUMEN

The ovalbumin-induced (OVA) chronic allergic airways murine model is a well-established model for investigating pre-clinical therapies for chronic allergic airways diseases, such as asthma. Here, we examined the effects of several experimental compounds with potential anti-asthmatic effects including resveratrol (RV), relaxin (RLN), L-sulforaphane (LSF), valproic acid (VPA), and trichostatin A (TSA) using both a prevention and reversal model of chronic allergic airways disease. We undertook a novel analytical approach using focal plane array (FPA) and synchrotron Fourier-transform infrared (S-FTIR) microspectroscopic techniques to provide new insights into the mechanisms of action of these experimental compounds. Apart from the typical biological effects, S-FTIR microspectroscopy was able to detect changes in nucleic acids and protein acetylation. Further, we validated the reduction in collagen deposition induced by each experimental compound evaluated. Although this has previously been observed with conventional histological methods, the S-FTIR technique has the advantage of allowing identification of the type of collagen present. More generally, our findings highlight the potential utility of S-FTIR and FPA-FTIR imaging techniques in enabling a better mechanistic understanding of novel asthma therapeutics.


Asunto(s)
Antiasmáticos/administración & dosificación , Asma/tratamiento farmacológico , Ácidos Hidroxámicos/administración & dosificación , Isotiocianatos/administración & dosificación , Relaxina/administración & dosificación , Resveratrol/administración & dosificación , Ácido Valproico/administración & dosificación , Animales , Asma/inducido químicamente , Enfermedad Crónica/tratamiento farmacológico , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos/métodos , Femenino , Ratones , Ratones Endogámicos BALB C , Ovalbúmina/efectos adversos , Espectroscopía Infrarroja por Transformada de Fourier/métodos , Sulfóxidos , Sincrotrones , Resultado del Tratamiento
2.
J Med Chem ; 63(1): 88-102, 2020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31804829

RESUMEN

LpxC inhibitors were optimized starting from lead compounds with limited efficacy and solubility and with the goal to provide new options for the treatment of serious infections caused by Gram-negative pathogens in hospital settings. To enable the development of an aqueous formulation for intravenous administration of the drug at high dose, improvements in both solubility and antibacterial activity in vivo were prioritized early on. This lead optimization program resulted in the discovery of compounds such as 13 and 30, which exhibited high solubility and potent efficacy against Gram-negative pathogens in animal infection models.


Asunto(s)
Amidohidrolasas/antagonistas & inhibidores , Antibacterianos/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Infecciones por Escherichia coli/tratamiento farmacológico , Ácidos Hidroxámicos/uso terapéutico , Administración Intravenosa , Animales , Antibacterianos/administración & dosificación , Antibacterianos/síntesis química , Antibacterianos/farmacocinética , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacocinética , Bacterias Gramnegativas/efectos de los fármacos , Hepatocitos/metabolismo , Ácidos Hidroxámicos/administración & dosificación , Ácidos Hidroxámicos/síntesis química , Ácidos Hidroxámicos/farmacocinética , Ratones , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Ratas , Solubilidad
3.
Adv Gerontol ; 32(4): 627-632, 2019.
Artículo en Ruso | MEDLINE | ID: mdl-31800193

RESUMEN

51 patients with operated colorectal cancer T1N0M0, T3N1M0 and T4N0M1 at the age of 67±2,3 years receiving adjuvant chemotherapy and nutritional support (NS) were examined. Nutritional status was assessed using alimentazione-volume diagnosis to the points on L.N.Kostyuchenko, nutritional risk - with NRI, body composition - with bioimpedance method, the iron metabolism - with basic markers (serum ferritin, transferrin saturation with iron, erythrocyte indices: erythrocyte saturation of iron, the average concentration of hemoglobin in the erythrocyte, mean corpuscular volume, hemoglobin, etc.), traditional settings, the staging of tumors - according to TNM. Iron deficiency before the development of anemia can be corrected with NS. Upon the occurrence of anemia requires additional pharmacological treatment iron supplementation, preferable with iron III hydroxide olygoisomaltazat 1000 + NS) for the prevention of toxic-metabolic complications.


Asunto(s)
Anemia Ferropénica , Neoplasias Colorrectales , Apoyo Nutricional , Anciano , Anemia Ferropénica/complicaciones , Anemia Ferropénica/tratamiento farmacológico , Neoplasias Colorrectales/complicaciones , Neoplasias Colorrectales/tratamiento farmacológico , Compuestos Férricos/administración & dosificación , Ferritinas , Humanos , Ácidos Hidroxámicos/administración & dosificación , Apoyo Nutricional/normas , Síndrome
4.
Invest New Drugs ; 36(6): 1072-1084, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30198057

RESUMEN

PURPOSE: Resminostat is an oral inhibitor of class I, IIB, and IV histone deacetylases. This phase I/II study compared the safety and efficacy of resminostat plus sorafenib versus sorafenib monotherapy as first-line therapy for advanced hepatocellular carcinoma (HCC). EXPERIMENTAL DESIGN: In phase I, resminostat (400 mg or 600 mg/day on days 1 to 5 every 14 days) was administered with sorafenib (800 mg/day for 14 days) to determine the recommended dose for phase II. In phase II, patients were randomized (1:1) to sorafenib monotherapy or resminostat plus sorafenib. The primary endpoint was time-to-progression (TTP). RESULTS: Nine patients (3: 400 mg, 6: 600 mg) were enrolled in phase I, and the recommended dose of resminostat was determined to be 400 mg/day. Then 170 patients were enrolled in phase II. Median TTP/overall survival (OS) were 2.8/14.1 months with monotherapy versus 2.8/11.8 months with combination therapy (Hazard Ratio [HR]: 0.984, p = 0.925/HR: 1.046, p = 0.824). The overall incidence of adverse events was similar in both groups (98.8% versus 100.0%). However, thrombocytopenia ≥ Grade 3 was significantly more frequent in the combination therapy group (34.5% versus 2.4%, p < 0.001). Subgroup analysis revealed that median TTP/OS was 1.5/6.9 months for monotherapy versus 2.8/13.1 months for combination therapy (HR: 0.795, p = 0.392/HR: 0.567, p = 0.065) among patients with a normal-to-high baseline platelet count (≥ 150 × 103/mm3). CONCLUSIONS: In patients with advanced HCC, first-line therapy with resminostat at the recommended dose plus sorafenib showed no significant efficacy advantage over sorafenib monotherapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Pueblo Asiatico , Carcinoma Hepatocelular/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/uso terapéutico , Ácidos Hidroxámicos/administración & dosificación , Ácidos Hidroxámicos/uso terapéutico , Neoplasias Hepáticas/tratamiento farmacológico , Sorafenib/uso terapéutico , Sulfonamidas/administración & dosificación , Sulfonamidas/uso terapéutico , Administración Oral , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Carcinoma Hepatocelular/patología , Femenino , Inhibidores de Histona Desacetilasas/administración & dosificación , Humanos , Ácidos Hidroxámicos/efectos adversos , Estimación de Kaplan-Meier , Neoplasias Hepáticas/patología , Masculino , Estadificación de Neoplasias , Sorafenib/efectos adversos , Sulfonamidas/efectos adversos , Resultado del Tratamiento
5.
Adv Exp Med Biol ; 1074: 367-373, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29721965

RESUMEN

Photoreceptor cell death in inherited retinal degeneration is accompanied by over-activation of histone deacetylases (HDAC). Excessive HDAC activity is found both in primary rod degeneration (such as in the rd10 mouse) and in primary cone death, including the cone photoreceptor function loss 1 (cpfl1) mouse. We evaluated the potential of pharmacological HDAC inhibition to prevent photoreceptor degeneration in primary rod and cone degeneration. We show that a single in vivo treatment of cpfl1 mice with the HDAC inhibitor trichostatin A (TSA) resulted in a significant protection of cpfl1 mutant cones. Similarly, HDAC inhibition with the clinically approved HDAC inhibitor vorinostat (SAHA) resulted in a significant improvement of rod survival in rd10 retinal explant cultures. Altogether, these results highlight the feasibility of targeted neuroprotection in vivo and create hope to maintain vision in patients suffering from both rod and cone dystrophies.


Asunto(s)
Distrofias de Conos y Bastones/prevención & control , Inhibidores de Histona Desacetilasas/uso terapéutico , Ácidos Hidroxámicos/uso terapéutico , Vorinostat/uso terapéutico , Animales , Animales Congénicos , Muerte Celular , Distrofias de Conos y Bastones/tratamiento farmacológico , Distrofias de Conos y Bastones/enzimología , Distrofias de Conos y Bastones/genética , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Inhibidores de Histona Desacetilasas/administración & dosificación , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/administración & dosificación , Ácidos Hidroxámicos/farmacología , Inyecciones Intravítreas , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Técnicas de Cultivo de Órganos , Células Fotorreceptoras Retinianas Conos/efectos de los fármacos , Células Fotorreceptoras Retinianas Conos/patología , Células Fotorreceptoras Retinianas Bastones/efectos de los fármacos , Células Fotorreceptoras Retinianas Bastones/patología , Vorinostat/administración & dosificación , Vorinostat/farmacología
6.
Int J Nanomedicine ; 12: 6503-6520, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28919751

RESUMEN

BACKGROUND: Breast cancer is the most common malignant disease that occurs in women. Histone deacetylase (HDAC) inhibition has recently emerged as an effective and attractive target for the treatment of cancer. The aim of this study was to investigate the efficacy of a combined treatment of tubastatin A (TUB-A) and palladium nanoparticles (PdNPs) against MDA-MB-231 human breast cancer cells using two different cytotoxic agents that work by two different mechanisms, thereby decreasing the probability of chemoresistance in cancer cells and increasing the efficacy of toxicity, to provide efficient therapy for advanced stage of cancer without any undesired side effects. METHODS: PdNPs were synthesized using a novel biomolecule called R-phycoerythrin and characterized using various analytical techniques. The combinatorial effect of TUB-A and PdNPs was assessed by various cellular and biochemical assays and also by gene expression analysis. RESULTS: The biologically synthesized PdNPs had an average size of 25 nm and were spherical in shape. Treatment of MDA-MB-231 human breast cancer cells with TUB-A or PdNPs showed a dose-dependent effect on cell viability. The combination of 4 µM TUB-A and 4 µM PdNPs had a significant inhibitory effect on cell viability compared with either TUB-A or PdNPs alone. The combinatorial treatment also had a more pronounced effect on the inhibition of HDAC activity and enhanced apoptosis by regulating various cellular and biochemical changes. CONCLUSION: Our results suggest that there was a strong synergistic interaction between TUB-A and PdNPs in increasing apoptosis in human breast cancer cells. These data provide an important preclinical basis for future clinical trials on this drug combination. This combinatorial treatment increased therapeutic potentials, thereby demonstrating a relevant targeted therapy for breast cancer. Furthermore, we have provided the first evidence for the combinatorial effect and mechanism of toxicity of TUB-A and PdNPs in human breast cancer cells. The novelties of the study were identification of a combination therapy that consists of suitable therapeutic molecules that kill cancer cells and also exploration of two different possible mechanisms involved to reduce chemoresistance in cancer cells.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Nanopartículas del Metal/administración & dosificación , Paladio/química , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/química , Neoplasias de la Mama/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Sistemas de Liberación de Medicamentos/métodos , Femenino , Inhibidores de Histona Desacetilasas/administración & dosificación , Inhibidores de Histona Desacetilasas/química , Humanos , Ácidos Hidroxámicos/administración & dosificación , Ácidos Hidroxámicos/química , Indoles/administración & dosificación , Indoles/química , Nanopartículas del Metal/química , Paladio/farmacología , Ficoeritrina/química , Extractos Vegetales/química
7.
Artículo en Inglés | MEDLINE | ID: mdl-28807913

RESUMEN

Directly testing proposed clinical dosing regimens in nonclinical studies can reduce the risk during the development of novel antibacterial agents. Optimal dosing regimens can be identified in animal models by testing recreated human pharmacokinetic profiles. An example of this approach using continuous intravenous infusions of GSK1322322 in immunocompetent rats to evaluate recreated human exposures from phase I trials in pneumonia models with Streptococcus pneumoniae and Haemophilus influenzae and an abscess model with Staphylococcus aureus is presented. GSK1322322 was administered via continuous intravenous infusion to recreate 1,000- or 1,500-mg oral doses every 12 h in humans. Significant reductions (P ≤ 0.05 for all comparisons) in bacterial numbers compared with those for the baseline controls were observed for S. pneumoniae and H. influenzae (mean log10 reductions, 1.6 to ≥2.7 and 1.8 to 3.3 CFU/lungs, respectively) with the recreated 1,000-mg oral dose. This profile was also efficacious against S. aureus (mean log10 reduction, 1.9 to 2.4 CFU/abscess). There was a nonsignificant trend for improved efficacy against S. aureus with the 1,500-mg oral dose (mean log10 reduction, 2.4 to 3.1 CFU/abscess). These results demonstrate that the human oral 1,000- or 1,500-mg exposure profiles of GSK1322322 recreated in rats were effective against representative community-associated pathogens and supported selection of the 1,500-mg oral dose given every 12 h for a phase II clinical skin infection study. Furthermore, this work exemplifies how the testing of recreated human pharmacokinetic profiles can be incorporated into the development process and serve as an aid for selecting optimal dosing regimens prior to conducting large-scale clinical studies.


Asunto(s)
Antibacterianos/administración & dosificación , Antibacterianos/uso terapéutico , Compuestos Bicíclicos Heterocíclicos con Puentes/administración & dosificación , Compuestos Bicíclicos Heterocíclicos con Puentes/uso terapéutico , Haemophilus influenzae/efectos de los fármacos , Ácidos Hidroxámicos/administración & dosificación , Ácidos Hidroxámicos/uso terapéutico , Staphylococcus aureus/efectos de los fármacos , Streptococcus pneumoniae/efectos de los fármacos , Animales , Antibacterianos/farmacocinética , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacocinética , Modelos Animales de Enfermedad , Esquema de Medicación , Infecciones por Haemophilus/tratamiento farmacológico , Humanos , Ácidos Hidroxámicos/farmacocinética , Masculino , Pruebas de Sensibilidad Microbiana , Infecciones Neumocócicas/tratamiento farmacológico , Ratas , Ratas Sprague-Dawley , Infecciones Estafilocócicas/tratamiento farmacológico
8.
BMC Microbiol ; 17(1): 98, 2017 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-28438125

RESUMEN

BACKGROUND: Chlamydia species are obligate intracellular bacteria that infect a broad range of mammalian hosts. Members of related genera are pathogens of a variety of vertebrate and invertebrate species. Despite the diversity of Chlamydia, all species contain an outer membrane lipooligosaccharide (LOS) that is comprised of a genus-conserved, and genus-defining, trisaccharide 3-deoxy-D-manno-oct-2-ulosonic acid Kdo region. Recent studies with lipopolysaccharide inhibitors demonstrate that LOS is important for the C. trachomatis developmental cycle during RB- > EB differentiation. Here, we explore the effects of one of these inhibitors, LPC-011, on the developmental cycle of five chlamydial species. RESULTS: Sensitivity to the drug varied in some of the species and was conserved between others. We observed that inhibition of LOS biosynthesis in some chlamydial species induced formation of aberrant reticulate bodies, while in other species, no change was observed to the reticulate body. However, loss of LOS production prevented completion of the chlamydial reproductive cycle in all species tested. In previous studies we found that C. trachomatis and C. caviae infection enhances MHC class I antigen presentation of a model self-peptide. We find that treatment with LPC-011 prevents enhanced host-peptide presentation induced by infection with all chlamydial-species tested. CONCLUSIONS: The data demonstrate that LOS synthesis is necessary for production of infectious progeny and inhibition of LOS synthesis induces aberrancy in certain chlamydial species, which has important implications for the use of LOS synthesis inhibitors as potential antibiotics.


Asunto(s)
Proteínas Bacterianas/efectos de los fármacos , Proteínas Bacterianas/genética , Chlamydia/efectos de los fármacos , Chlamydia/crecimiento & desarrollo , Ácidos Hidroxámicos/antagonistas & inhibidores , Treonina/análogos & derivados , Secuencia de Aminoácidos , Ampicilina/farmacología , Animales , Antibacterianos/farmacología , Línea Celular/efectos de los fármacos , Línea Celular/microbiología , Chlamydia/genética , Chlamydia/patogenicidad , Infecciones por Chlamydia/tratamiento farmacológico , Citoplasma/microbiología , Fibroblastos , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Interacciones Huésped-Patógeno , Humanos , Ácidos Hidroxámicos/administración & dosificación , Lipopolisacáridos/biosíntesis , Ratones , Pruebas de Sensibilidad Microbiana , Fenotipo , Filogenia , Biosíntesis de Proteínas/efectos de los fármacos , Alineación de Secuencia , Análisis de Secuencia de Proteína , Azúcares Ácidos , Treonina/administración & dosificación , Treonina/antagonistas & inhibidores
9.
Expert Opin Investig Drugs ; 25(9): 1117-31, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27485472

RESUMEN

INTRODUCTION: Therapeutic strategies in patients with acute myeloid leukemia (AML) have not changed significantly over the last decades. Appropriate strategies are ultimately driven by the assessment of patients' fitness to define suitability for intensive induction chemotherapy, which produces high initial remission rates but, increased likelihood of relapse. Old/unfit AML patients still represent an urgent and unmet therapeutic need. Epigenetic deregulation represents a strategic characteristic of AML pathophysiology whereby aberrant gene transcription provides an advantage to leukemic cell survival. Efforts to re-establish impaired epigenetic regulation include hypomethylating agents and histone deacetylase inhibitors (HDACi). AREAS COVERED: The review discusses the underlying mechanisms leading to disruption of lysine acetyltransferases (KAT or HAT)/deacetylase (KDAC or HDAC) balance and the rationale for using the HDACi panobinostat (LBH-589) in AML. EXPERT OPINION: Although panobinostat has produced significant results in myeloma, its efficacy remains limited in AML. Panobinostat exerts pleiotropic activity and lack of specificity, which likely contributes to its inadequate safety in elderly AML patients. Phase I-II trials, utilizing panobinostat associated with well-known chemotherapeutic agents are ongoing and combinations with other druggable targets may likely be evaluated in future trials. The clinical use of this HDACi in AML the near future does not appearing promising.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Inhibidores de Histona Desacetilasas/uso terapéutico , Ácidos Hidroxámicos/uso terapéutico , Indoles/uso terapéutico , Leucemia Mieloide Aguda/tratamiento farmacológico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Ensayos Clínicos como Asunto , Metilación de ADN/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Epigénesis Genética/efectos de los fármacos , Inhibidores de Histona Desacetilasas/administración & dosificación , Inhibidores de Histona Desacetilasas/efectos adversos , Inhibidores de Histona Desacetilasas/farmacocinética , Humanos , Ácidos Hidroxámicos/administración & dosificación , Ácidos Hidroxámicos/efectos adversos , Ácidos Hidroxámicos/farmacocinética , Indoles/administración & dosificación , Indoles/efectos adversos , Indoles/farmacocinética , Leucemia Mieloide Aguda/genética , Panobinostat , Resultado del Tratamiento
10.
J Exp Clin Cancer Res ; 35(1): 94, 2016 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-27317342

RESUMEN

BACKGROUND: Medulloblastoma (MB) is the most common pediatric brain tumor. Current treatment regimes consisting of primary surgery followed by radio- and chemotherapy, achieve 5-year overall survival rates of only about 60 %. Therapy-induced endocrine and neurocognitive deficits are common late adverse effects. Thus, improved antitumor strategies are urgently needed. In this study, we combined irradiation (IR) together with epigenetic modifiers and differentiation inducers in a multimodal approach to enhance the efficiency of tumor therapy in MB and also assessed possible late adverse effects on neurogenesis. METHODS: In three human MB cell lines (DAOY, MEB-Med8a, D283-Med) short-time survival (trypan blue exclusion assay), apoptosis, autophagy, cell cycle distribution, formation of gH2AX foci, and long-term reproductive survival (clonogenic assay) were analyzed after treatment with 5-aza-2'-deoxycytidine (5-azadC), valproic acid (VPA), suberanilohydroxamic acid (SAHA), abacavir (ABC), all-trans retinoic acid (ATRA) and resveratrol (RES) alone or combined with 5-aza-dC and/or IR. Effects of combinatorial treatments on neurogenesis were evaluated in cultured murine hippocampal slices from transgenic nestin-CFPnuc C57BL/J6 mice. Life imaging of nestin-positive neural stem cells was conducted at distinct time points for up to 28 days after treatment start. RESULTS: All tested drugs showed a radiosynergistic action on overall clonogenic survival at least in two-outof-three MB cell lines. This effect was pronounced in multimodal treatments combining IR, 5-aza-dC and a second drug. Hereby, ABC and RES induced the strongest reduction of clongenic survival in all three MB cell lines and led to the induction of apoptosis (RES, ABC) and/or autophagy (ABC). Additionally, 5-aza-dC, RES, and ABC increased the S phase cell fraction and induced the formation of gH2AX foci at least in oneout-of-three cell lines. Thereby, the multimodal treatment with 5-aza-dC, IR, and RES or ABC did not change the number of normal neural progenitor cells in murine slice cultures. CONCLUSION: In conclusion, the radiosensitizing capacities of epigenetic and differentiation-inducing drugs presented here suggest that their adjuvant administration might improve MB therapy. Thereby, the combination of 5-aza-dC/IR with ABC and RES seemed to be the most promising to enhance tumor control without affecting the normal neural precursor cells.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias Cerebelosas/tratamiento farmacológico , Neoplasias Cerebelosas/radioterapia , Meduloblastoma/tratamiento farmacológico , Meduloblastoma/radioterapia , Fármacos Sensibilizantes a Radiaciones/administración & dosificación , Animales , Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Azacitidina/administración & dosificación , Azacitidina/análogos & derivados , Azacitidina/farmacología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/efectos de la radiación , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Neoplasias Cerebelosas/genética , Terapia Combinada , Decitabina , Didesoxinucleósidos/administración & dosificación , Didesoxinucleósidos/farmacología , Epigénesis Genética/efectos de los fármacos , Epigénesis Genética/efectos de la radiación , Humanos , Ácidos Hidroxámicos/administración & dosificación , Ácidos Hidroxámicos/farmacología , Meduloblastoma/genética , Ratones , Neurogénesis/efectos de los fármacos , Neurogénesis/efectos de la radiación , Fármacos Sensibilizantes a Radiaciones/farmacología , Resveratrol , Estilbenos/administración & dosificación , Estilbenos/farmacología , Resultado del Tratamiento , Tretinoina/administración & dosificación , Tretinoina/farmacología , Ácido Valproico/administración & dosificación , Ácido Valproico/farmacología , Vorinostat , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Am J Health Syst Pharm ; 73(7): 441-50, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-27001985

RESUMEN

PURPOSE: The mechanism of action, pharmacodynamics, pharmacokinetics, clinical efficacy, interaction potential, adverse effects, and place in therapy of panobinostat are reviewed. SUMMARY: Panobinostat (Farydak, Novartis) is a novel pan-deacetylase inhibitor approved for use in combination with bortezomib and dexamethasone in patients with relapsed or refractory multiple myeloma (RRMM) who have received at least two regimens containing an immunomodulatory drug and bortezomib. National Comprehensive Cancer Network (NCCN) guidelines recommend the use of panobinostat plus bortezomib and dexamethasone as a preferred regimen for previously treated multiple myeloma (MM). A Phase III trial comparing panobinostat or placebo use in combination with bortezomib and dexamethasone demonstrated improved median progression-free survival in the panobinostat group (11.99 months [95% CI, 10.33-12.94 months] versus 8.08 months [95% CI, 7.56-9.23 months]; hazard ratio, 0.63 [95% CI, 0.52-0.76]; p < 0.0001), as well as a significantly higher rate of complete or near complete response (27.6% [95% CI, 23.2-32.4%] versus 15.7% [95% CI, 12.2-19.8%]; p = 0.00006). Common grade 3 or 4 laboratory abnormalities and adverse events associated with panobinostat include thrombocytopenia, lymphopenia, diarrhea, asthenia, fatigue, and peripheral neuropathy. CONCLUSION: Panobinostat is a promising alternative to well-studied, NCCN-recommended regimens for the treatment of RRMM. It has demonstrated efficacy when used in combination with bortezomib and dexamethasone for the treatment of patients with MM who have received at least two prior regimens including bortezomib and an immunomodulatory agent. Despite the observed benefits, concern regarding toxicity may limit panobinostat use in practice.


Asunto(s)
Inhibidores de Histona Desacetilasas/administración & dosificación , Ácidos Hidroxámicos/administración & dosificación , Indoles/administración & dosificación , Mieloma Múltiple/tratamiento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Bortezomib/administración & dosificación , Ensayos Clínicos Fase II como Asunto/métodos , Ensayos Clínicos Fase III como Asunto/métodos , Dexametasona/administración & dosificación , Humanos , Mieloma Múltiple/diagnóstico , Mieloma Múltiple/epidemiología , Panobinostat , Recurrencia , Resultado del Tratamiento
12.
Int J Clin Exp Pathol ; 8(5): 5584-90, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26191268

RESUMEN

Vorinostat (suberoylanilide hydroxamic acid, SAHA) is the first approved histone deacetylase (HDAC) inhibitor for the treatment of cutaneous T-cell lymphoma after progressive disease following two systemic therapies. The rats were randomly divided into SAHA groups (low, medium and high dosage) and control group. The SAHA group rats were given 12.3, 24.5, and 49 mg/kg SAHA, respectively, by continuous intragastric administration for 7 days. The influence of SAHA on the activities of CYP450 isoforms CYP2B6, CYP1A2, CYP2C19, CYP2D6 and CYP2C9 were evaluated by cocktail method, they were responsed by the changes of pharmacokinetic parameters of bupropion, phenacetin, tolbutamide, metroprolol and omeprazole. The five probe drugs were given to rats through intragastric administration, and the plasma concentration were determined by UPLC-MS/MS. The result of SAHA group compared to control group, there were statistical pharmacokinetics difference for bupropion, phenacetin, tolbutamide and metroprolol. Continuous intragastric administration for 7 days may induce the activities of CYP2C19 of rats, inhibit CYP1A2 and slightly inhibit CYP2B6 and CYP2D6 of rats. This may give advising for reasonable drug use after co-used with SAHA. The results indicated that drug co-administrated with SAHA may need dose adjustment. Furthermore, continuous intragastric administration of SAHA for 7 days, liver cell damaged, causing liver cell edema, in liver metabolism process.


Asunto(s)
Inhibidores del Citocromo P-450 CYP1A2/administración & dosificación , Inductores del Citocromo P-450 CYP2C19/administración & dosificación , Citocromo P-450 CYP2C19/biosíntesis , Citocromos/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/administración & dosificación , Ácidos Hidroxámicos/administración & dosificación , Hígado/efectos de los fármacos , Administración Oral , Animales , Bupropión/sangre , Bupropión/farmacocinética , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Cromatografía Liquida , Citocromo P-450 CYP1A2 , Inhibidores del Citocromo P-450 CYP1A2/toxicidad , Citocromo P-450 CYP2B6/metabolismo , Inhibidores del Citocromo P-450 CYP2B6/administración & dosificación , Inductores del Citocromo P-450 CYP2C19/toxicidad , Citocromo P-450 CYP2D6/metabolismo , Inhibidores del Citocromo P-450 CYP2D6/administración & dosificación , Citocromos/metabolismo , Interacciones Farmacológicas , Edema/inducido químicamente , Edema/patología , Inducción Enzimática , Inhibidores de Histona Desacetilasas/toxicidad , Ácidos Hidroxámicos/toxicidad , Hígado/enzimología , Hígado/patología , Masculino , Metoprolol/sangre , Metoprolol/farmacocinética , Omeprazol/sangre , Omeprazol/farmacocinética , Fenacetina/sangre , Fenacetina/farmacocinética , Ratas Sprague-Dawley , Especificidad por Sustrato , Espectrometría de Masas en Tándem , Tolbutamida/sangre , Tolbutamida/farmacocinética , Vorinostat
13.
Cancer Biol Ther ; 16(7): 1047-55, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25774632

RESUMEN

Arginine auxotrophy constitutes a weak point of several tumors, among them glioblastoma multiforme (GBM). Hence, those tumors are supposed to be sensitive for arginine-depleting substances, such as arginine deiminase (ADI). Here we elucidated the sensitivity of patient-individual GBM cell lines toward Streptococcus pyogenes-derived ADI. To improve therapy, ADI was combined with currently established and pre-clinical cytostatic drugs. Additionally, effectiveness of local ADI therapy was determined in xenopatients. Half of the GBM cell lines tested responded well toward ADI monotherapy. In those cell lines, viability decreased significantly (up to 50%). Responding cell lines were subjected to combination therapy experiments to test if any additive or even synergistic effects may be achieved. Such promising results were obtained in 2/3 cases. In cell lines HROG02, HROG05 and HROG10, ADI and Palomid 529 combinations were most effective yielding more than 70% killing after 2 rounds of treatment. Comparable boosted antitumoral effects were observed after adding chloroquine to ADI (>60% killing). Apoptosis, as well as cell cycle dysregulation were found to play a minor role. In some, but clearly not all cases, (epi-) genetic silencing of arginine synthesis pathway genes (argininosuccinate synthetase 1 and argininosuccinate lyase) explained obtained results. In vivo, ADI as well as the combination of ADI and SAHA efficiently controlled HROG05 xenograft growth, whereas adding Palomid 529 to ADI did not further increase the strong antitumoral effect of ADI. The cumulative in vitro and in vivo results proved ADI as a very promising candidate therapeutic, especially for development of adjuvant GBM combination treatments.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Arginina/metabolismo , Glioblastoma/tratamiento farmacológico , Hidrolasas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Antineoplásicos/administración & dosificación , Argininosuccinatoliasa/genética , Argininosuccinato Sintasa/genética , Benzopiranos/administración & dosificación , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Cloroquina/administración & dosificación , Metilación de ADN/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Hidrolasas/administración & dosificación , Hidrolasas/metabolismo , Ácidos Hidroxámicos/administración & dosificación , Masculino , Ratones Endogámicos , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Streptococcus pyogenes/enzimología , Resultado del Tratamiento , Carga Tumoral/efectos de los fármacos , Carga Tumoral/genética , Vorinostat
14.
PLoS One ; 9(12): e115068, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25500581

RESUMEN

Mutations in the WNT/beta-catenin pathway are present in the majority of all sporadic colorectal cancers (CRCs), and histone deacetylase inhibitors induce apoptosis in CRC cells with such mutations. This apoptosis is counteracted by (1) the signaling heterogeneity of CRC cell populations, and (2) the survival pathways induced by mitogens secreted from apoptotic cells. The phenomena of signaling heterogeneity and apoptosis-induced survival constitute the immediate mechanisms of resistance to histone deacetylase inhibitors, and probably other chemotherapeutic agents. We explored the strategy of augmenting CRC cell death by inhibiting all survival pathways induced by the pro-apoptotic agent LBH589, a histone deacetylase inhibitor: AKT, JAK/STAT, and ERK signaling. The apoptosis-enhancing ability of a cocktail of synthetic inhibitors of proliferation was compared to the effects of the natural product propolis. We utilized colorectal adenoma, drug-sensitive and drug-resistant colorectal carcinoma cells to evaluate the apoptotic potential of the combination treatments. The results suggest that an effective approach to CRC combination therapy is to combine apoptosis-inducing drugs (e.g., histone deacetylase inhibitors, such as LBH589) with agents that suppress all compensatory survival pathways induced during apoptosis (such as the cocktail of inhibitors of apoptosis-associated proliferation). The same paradigm can be applied to a CRC prevention approach, as the apoptotic effect of butyrate, a diet-derived histone deacetylase inhibitor, is augmented by other dietary agents that modulate survival pathways (e.g., propolis and coffee extract). Thus, dietary supplements composed by fermentable fiber, propolis, and coffee extract may effectively counteract neoplastic growth in the colon.


Asunto(s)
Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/tratamiento farmacológico , Vía de Señalización Wnt/efectos de los fármacos , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Suplementos Dietéticos , Inhibidores de Histona Desacetilasas/administración & dosificación , Humanos , Ácidos Hidroxámicos/administración & dosificación , Indoles/administración & dosificación , Panobinostat , Própolis/administración & dosificación
15.
PLoS One ; 9(11): e112619, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25402485

RESUMEN

Acute Myeloid Leukemia (AML) is an aggressive malignancy which leads to marrow failure, and ultimately death. There is a desperate need for new therapeutics for these patients. Ribonucleotide reductase (RR) is the rate limiting enzyme in DNA synthesis. Didox (3,4-Dihydroxybenzohydroxamic acid) is a novel RR inhibitor noted to be more potent than hydroxyurea. In this report we detail the activity and toxicity of Didox in preclinical models of AML. RR was present in all AML cell lines and primary patient samples tested. Didox was active against all human and murine AML lines tested with IC50 values in the low micromolar range (mean IC50 37 µM [range 25.89-52.70 µM]). It was active against primary patient samples at concentrations that did not affect normal hematopoietic stem cells (HSCs). Didox exposure resulted in DNA damage and p53 induction culminating in apoptosis. In syngeneic, therapy-resistant AML models, single agent Didox treatment resulted in a significant reduction in leukemia burden and a survival benefit. Didox was well tolerated, as marrow from treated animals was morphologically indistinguishable from controls. Didox exposure at levels that impaired leukemia growth did not inhibit normal HSC engraftment. In summary, Didox was well tolerated and effective against preclinical models of AML.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Ácidos Hidroxámicos/farmacología , Leucemia Mieloide Aguda/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Resistencia a Antineoplásicos , Inhibidores Enzimáticos/administración & dosificación , Femenino , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Ácidos Hidroxámicos/administración & dosificación , Cariotipo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/mortalidad , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Ribonucleótido Reductasas/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Ensayo de Tumor de Célula Madre , Proteína p53 Supresora de Tumor/metabolismo
16.
Cancer Treat Rev ; 40(8): 927-33, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24997678

RESUMEN

BACKGROUND: A variety of therapeutic options are available for mycosis fungoides, the most prevalent subtype of cutaneous T cell lymphomas, but thus far, no regimen has been proven to be curative. A combination of treatments is a well-established strategy to increase the therapeutic efficacy. However, data from clinical trials analyzing such combinations for the treatment of mycosis fungoides are scarce. OBJECTIVE: To analyze the available evidence on combination therapies with emphasis on the combination of psoralen with UVA phototherapy (PUVA), interferon-alpha and bexarotene with another treatment. METHODS: Systematic literature review of the databases Embase, Cochrane, Medline, and Medline in Process. RESULTS: Combination of PUVA with interferon-alpha or retinoids did not result in an increased overall response rate. Addition of methotrexate but not retinoids to interferon-alpha may increase the overall response rate. Bexarotene was investigated in one trial each with vorinostat, methotrexate or gemcitabine, whereby only methotrexate possibly enhanced the effect of bexarotene. CONCLUSION: For mycosis fungoides, no combination treatment has been demonstrated to be superior to monotherapy. Based on our analysis, we conclude that in certain clinical situations, patients may benefit from a combination of PUVA with interferon-alpha or a retinoid or a combination of the latter two. Furthermore, patients in advanced stages may benefit from the combination of methotrexate and interferon-alpha or bexarotene. Finally, the combination of bexarotene with either vorinostat or gemcitabine did not increase the overall response rate but resulted in more pronounced side effects and cannot be recommended.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Micosis Fungoide/terapia , Fototerapia/métodos , Neoplasias Cutáneas/terapia , Bexaroteno , Terapia Combinada , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Ficusina/administración & dosificación , Humanos , Ácidos Hidroxámicos/administración & dosificación , Interferón-alfa/administración & dosificación , Metotrexato/administración & dosificación , Micosis Fungoide/tratamiento farmacológico , Retinoides/administración & dosificación , Neoplasias Cutáneas/tratamiento farmacológico , Tetrahidronaftalenos/administración & dosificación , Vorinostat , Gemcitabina
17.
Int J Oncol ; 45(1): 177-88, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24807012

RESUMEN

Sorafenib is effective for patients with advanced hepatocellular carcinoma (HCC) and particularly for those who are unsuitable to receive life-prolonging transarterial chemo-embolization. The survival benefit of sorafenib, however, is unsatisfactory. Vorinostat also known as suberoylanilide hydroxamic acid (SAHA) is a histone deacetylase (HDAC) inhibitor with anti-HCC efficacy in preclinical studies. SAHA induces nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) activity in vitro, which may lead to cancer cell progression and jeopardize cytotoxic effect of SAHA in HCC. The goal of this study was to investigate whether sorafenib enhances SAHA cytotoxicity against HCC through inhibition of SAHA-induced NF-κB activity. The human HCC cell line Huh7 transfected with dual reporter genes, luciferase (luc) and thymidine kinase (tk) with NF-κB response elements, was co-transfected with red fluorescent protein (rfp) gene for non-invasive molecular imaging to assess NF-κB activity and living cells simultaneously. Cell viability assay, DNA fragmentation, western blotting, electrophoretic mobility shift assay (EMSA) and multiple modalities of molecular imaging were used to assess the combination efficacy and mechanism of sorafenib and SAHA. The administration of high-dose SAHA (10 µM) with long treatment time (48 h) in vitro, and 25 mg/kg/day by gavage in HCC-bearing nude mice to induce NF-κB activity were performed. Sorafenib inhibited SAHA-induced NF-κB activity and the expression of NF-κB-regulated effector proteins while it increased the efficacy of SAHA against HCC both in vitro and in vivo. The mechanism of sorafenib to enhance SAHA efficacy on HCC is through the suppression of ERK/NF-κB pathway, which induces extrinsic and intrinsic apoptosis. Combination of sorafenib and SAHA may have the potential as new strategy against HCC.


Asunto(s)
Antineoplásicos/administración & dosificación , Carcinoma Hepatocelular/patología , Ácidos Hidroxámicos/administración & dosificación , Neoplasias Hepáticas/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Niacinamida/análogos & derivados , Compuestos de Fenilurea/administración & dosificación , Animales , Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica , Línea Celular Tumoral , Sinergismo Farmacológico , Humanos , Ácidos Hidroxámicos/farmacología , Neoplasias Hepáticas Experimentales , Masculino , Ratones , Ratones Desnudos , Niacinamida/administración & dosificación , Niacinamida/farmacología , Compuestos de Fenilurea/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Sorafenib , Vorinostat , Ensayos Antitumor por Modelo de Xenoinjerto , Quinasa de Factor Nuclear kappa B
18.
Clin Exp Immunol ; 172(3): 375-82, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23600825

RESUMEN

Type 1 diabetes (T1D) results from T helper type 1 (Th1)-mediated autoimmune destruction of insulin-producing ß cells. Novel experimental therapies for T1D target immunomodulation, ß cell survival and inflammation. We examined combination therapy with the dipeptidyl peptidase-IV inhibitor MK-626 and the histone deacetylase inhibitor vorinostat in the non-obese diabetic (NOD) mouse model of T1D. We hypothesized that combination therapy would ameliorate T1D by providing protection from ß cell inflammatory destruction while simultaneously shifting the immune response towards immune-tolerizing regulatory T cells (T(regs)). Although neither mono- nor combination therapies with MK-626 and vorinostat caused disease remission in diabetic NOD mice, the combination of MK-626 and vorinostat increased ß cell area and reduced the mean insulitis score compared to diabetic control mice. In prediabetic NOD mice, MK-626 monotherapy resulted in improved glucose tolerance, a reduction in mean insulitis score and an increase in pancreatic lymph node T(reg) percentage, and combination therapy with MK-626 and vorinostat increased pancreatic lymph node T(reg) percentage. We conclude that neither single nor combination therapies using MK-626 and vorinostat induce diabetes remission in NOD mice, but combination therapy appears to have beneficial effects on ß cell area, insulitis and T(reg) populations. Combinations of vorinostat and MK-626 may serve as beneficial adjunctive therapy in clinical trials for T1D prevention or remission.


Asunto(s)
Diabetes Mellitus Tipo 1/tratamiento farmacológico , Inhibidores de la Dipeptidil-Peptidasa IV/administración & dosificación , Inhibidores de Histona Desacetilasas/administración & dosificación , Ácidos Hidroxámicos/administración & dosificación , Triazoles/administración & dosificación , Animales , Glucemia/metabolismo , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Quimioterapia Combinada , Femenino , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/patología , Ratones , Ratones Endogámicos NOD , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología , Factor de Crecimiento Transformador beta1/metabolismo , Vorinostat
19.
BMJ Case Rep ; 20122012 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-23257272

RESUMEN

Here, we report the case of a patient, who showed an antitumour response to a new combination therapy of sorafenib and the histon deacetylase inhibitor panobinostat (LBH-589). D-CEUS (Dynamic contrast-enhanced ultrasonography) was able to predict response to the new therapy regime and may be an interesting tool in the early evaluation of response to therapy. It might be especially useful to differentiate between responders and non-responders of new-targeted pharmaceuticals like multikinase inhibitors in hepatocellular carcinomas.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma Hepatocelular/diagnóstico por imagen , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/diagnóstico por imagen , Neoplasias Hepáticas/tratamiento farmacológico , Anciano , Carcinoma Hepatocelular/patología , Medios de Contraste , Resultado Fatal , Humanos , Ácidos Hidroxámicos/administración & dosificación , Indoles/administración & dosificación , Neoplasias Hepáticas/patología , Masculino , Estadificación de Neoplasias , Niacinamida/administración & dosificación , Niacinamida/análogos & derivados , Panobinostat , Compuestos de Fenilurea/administración & dosificación , Sorafenib , Ultrasonografía/métodos
20.
Clin Cancer Res ; 18(17): 4691-701, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22811583

RESUMEN

PURPOSE: Histone deacetylase inhibitors (HDACi) are actively explored as new-generation epigenetic drugs but have low efficacy in cancer monotherapy. To reveal new mechanism for combination therapy, we show that HDACi induce cell death but simultaneously activate tumor-progressive genes to ruin therapeutic efficacy. Combined treatments to target tumorigenesis and HDACi-activated metastasis with low toxic modalities could develop new strategies for long-term cancer therapy. EXPERIMENTAL DESIGN: Because metastasis is the major cause of cancer mortality, we measured cell migration activity and profiled metastasis-related gene expressions in HDACi-treated cancer cells. We developed low toxic combination modalities targeting tumorigenesis and HDACi-activated metastasis for preclinical therapies in mice. RESULTS: We showed that cell migration activity was dramatically and dose dependently enhanced by various classes of HDACi treatments in 13 of 30 examined human breast, gastric, liver, and lung cancer cell lines. Tumor metastasis was also enhanced in HDACi-treated mice. HDACi treatments activated multiple PKCs and downstream substrates along with upregulated proapoptotic p21. For targeting tumorigenesis and metastasis with immediate clinical impact, we showed that new modalities of HDACi combined drugs with PKC inhibitory agent, curcumin or tamoxifen, not only suppressed HDACi-activated tumor progressive proteins and cell migration in vitro but also inhibited tumor growth and metastasis in vivo. CONCLUSION: Treatments of different structural classes of HDACi simultaneously induced cell death and promoted cell migration and metastasis in multiple cancer cell types. Suppression of HDACi-induced PKCs leads to development of low toxic and long-term therapeutic strategies to potentially treat cancer as a chronic disease.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Movimiento Celular/efectos de los fármacos , Inhibidores de Histona Desacetilasas/administración & dosificación , Neoplasias/tratamiento farmacológico , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Terapia Combinada , Curcumina/administración & dosificación , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ácidos Hidroxámicos/administración & dosificación , Inhibidores de la Metaloproteinasa de la Matriz/administración & dosificación , Metaloproteinasas de la Matriz/metabolismo , Ratones , Metástasis de la Neoplasia , Neoplasias/metabolismo , Neoplasias/patología , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Tamoxifeno/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA