Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Invest New Drugs ; 41(4): 596-605, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37415001

RESUMEN

Tepotinib is a highly selective, potent, mesenchymal-epithelial transition factor (MET) inhibitor, approved for the treatment of non-small cell lung cancer harboring MET exon 14 skipping alterations. The aims of this work were to investigate the potential for drug-drug interactions via cytochrome P450 (CYP) 3A4/5 or P-glycoprotein (P-gp) inhibition. In vitro studies were conducted in human liver microsomes, human hepatocyte cultures and Caco-2 cell monolayers to investigate whether tepotinib or its major metabolite (MSC2571109A) inhibited or induced CYP3A4/5 or inhibited P-gp. Two clinical studies were conducted to investigate the effect of multiple dose tepotinib (500 mg once daily orally) on the single dose pharmacokinetics of a sensitive CYP3A4 substrate (midazolam 7.5 mg orally) and a P-gp substrate (dabigatran etexilate 75 mg orally) in healthy participants. Tepotinib and MSC2571109A showed little evidence of direct or time-dependent CYP3A4/5 inhibition (IC50 > 15 µM) in vitro, although MSC2571109A did show mechanism-based CYP3A4/5 inhibition. Tepotinib did not induce CYP3A4/5 activity in vitro, although both tepotinib and MSC2571109A increased CYP3A4 mRNA. In clinical studies, tepotinib had no effect on the pharmacokinetics of midazolam or its metabolite 1'-hydroxymidazolam. Tepotinib increased dabigatran maximum concentration and area under the curve extrapolated to infinity by 38% and 51%, respectively. These changes were not considered to be clinically relevant. Tepotinib was considered safe and well tolerated in both studies. The potential of tepotinib to cause clinically relevant DDI with CYP3A4- or P-gp-dependent drugs at the clinical dose is considered low. Study 1 (midazolam): NCT03628339 (registered 14 August 2018). Study 2 (dabigatran): NCT03492437 (registered 10 April 2018).


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Citocromo P-450 CYP3A/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Midazolam/farmacocinética , Sistema Enzimático del Citocromo P-450/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Dabigatrán/farmacocinética , Células CACO-2 , Subfamilia B de Transportador de Casetes de Unión a ATP , Interacciones Farmacológicas
2.
Clin Transl Sci ; 16(12): 2628-2639, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37905356

RESUMEN

Peposertib is an orally administered inhibitor of DNA-dependent protein kinase. We evaluated the effect of food on its pharmacokinetics, and examined the pharmacokinetics of an oral suspension (OS) of disintegrated tablets, in a phase I, open-label, crossover three-period study (NCT04702698). Twelve healthy volunteers were randomized to one of six treatment sequences. They received a single dose of peposertib 100 mg as film-coated tablets under fasted or fed conditions ("tablet fasted" or "tablet fed") or as an OS under fasted conditions ("OS fasted"), with washout between treatments. Using healthy volunteers was possible because, despite its mechanism of action being suppression of DNA repair, peposertib has shown no genotoxic effect in animals. A mild food effect was observed with peposertib tablets. Fed-to-fasted ratios were: area under the curve from time 0 to time t (AUC0-t ), 123.81% (90% confidence interval [CI]: 108.04, 141.87%); AUC from zero to infinity (AUC0-∞ ), 110.28% (90% CI 100.71, 120.77%); and maximum concentration (Cmax ) 104.47% (90% CI: 79.15, 137.90%). Cmax was delayed under fed conditions (median time to maximum concentration [Tmax ] was 3.5 h [tablet fed] vs. 1 h [tablet fasted]). OS-to-tablet (fasted) ratios were: AUC0-t , 124.83% (90% CI: 111.50%, 139.76%); AUC0-∞ , 119.05% (90% CI: 104.47, 135.67%); and Cmax 173.29% (90% CI: 135.78, 221.16%). Median Tmax was 0.5 h (OS fasted) versus 1 h (tablet). All treatments were well-tolerated in healthy volunteers. Peposertib tablets can be taken with or without food; if combined with chemotherapy or radiotherapy, the delay in Cmax must be considered to optimize the chemo- or radiosensitizing effect. The peposertib OS form represents an alternative route of administration in patients with specific cancers causing dysphagia. However, the OS form should be part of future dose optimization strategies in relevant settings.


Asunto(s)
Ayuno , Humanos , Administración Oral , Disponibilidad Biológica , Estudios Cruzados , Voluntarios Sanos , Comprimidos
3.
Pharmacol Res Perspect ; 9(5): e00842, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34414672

RESUMEN

This study evaluated the safety, tolerability, pharmacokinetics (PK), and pharmacodynamics (PD) of single and multiple oral doses of enpatoran (formerly named M5049), a new toll-like receptor (TLR) 7 and 8 dual antagonist, and the effect of food on a single dose in healthy participants. In this single phase 1, randomized (3:1), double-blind, placebo-controlled study, 96 participants received single and multiple ascending oral doses of enpatoran. Participants in single-dose cohorts received one dose of enpatoran (1, 3, 9, 25, 50, 100, or 200 mg) or placebo using a sentinel dosing strategy. Multiple-dose cohorts received enpatoran (9, 25, or 200 mg once daily, or 25 or 50 mg twice daily) or placebo for 14 days. Safety, tolerability, PK, and PD (ex vivo-stimulated cytokine secretion) were assessed in both parts. The effect of food was assessed in an open-label, one-way crossover study in the 25 mg single-dose cohort. Single- and multiple-oral doses of enpatoran up to 200 mg were well tolerated and no significant dose-limiting adverse events or safety signals were observed under fasting or fed conditions. PK parameters were linear and dose-proportional across the dose range evaluated, with a slightly delayed absorption and lower peak concentration observed at 25 mg with food. Exposure-dependent inhibition of ex vivo-stimulated interleukin-6 secretion was observed, with maximum inhibition at 200 mg. Enpatoran was well tolerated at doses up to 200 mg. Further investigation of enpatoran is warranted as a potential treatment for diseases driven by TLR7/8 overactivation, such as systemic lupus erythematosus and COVID-19 pneumonia.


Asunto(s)
Factores Inmunológicos/farmacología , Receptor Toll-Like 7/antagonistas & inhibidores , Receptor Toll-Like 8/antagonistas & inhibidores , Administración Oral , Adulto , COVID-19/inmunología , Método Doble Ciego , Femenino , Humanos , Lupus Eritematoso Sistémico/tratamiento farmacológico , Lupus Eritematoso Sistémico/inmunología , Masculino , SARS-CoV-2 , Tratamiento Farmacológico de COVID-19
4.
Clin Cancer Res ; 15(1): 324-9, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19118061

RESUMEN

PURPOSE: Recently, gene amplification and overexpression of KIT as well as activating mutations in the KIT gene have been described to occur in certain subsets of melanoma. These findings suggest KIT as a potential target for therapy with imatinib mesylate in these melanomas. To date, data on the KIT status in uveal melanoma (UM) is limited. EXPERIMENTAL DESIGN: We analyzed the expression of the KIT protein (CD117, c-kit) and its ligand, stem cell factor (SCF), in primary and metastatic UM. RESULTS: By immunohistochemistry, SCF-positive tumor cells (>90%) were detectable in 43% of primary UM and in 58% of UM metastases. Strong expression of KIT (>90%) in tumor cells was present in 55% of primary UM and in 76% of UM metastases. This overexpression of both KIT and SCF suggests the clinical application of imatinib mesylate in metastatic UM. This notion was tested in a clinical study using Simon's two-stage design. Patients received imatinib (600 mg p.o. daily) until progress or unacceptable toxicities. The trial did not enter stage II as no objective response was observed in the first group. This observation prompted further molecular analysis, which revealed no mutations in the genomic sequence of KIT in exons 11, 13, 17, and 18. Moreover, the mitogen-activated protein kinase pathway was not activated in any of the tumors as measured by ERK phosphorylation. CONCLUSIONS: These results show the lack of clinical effectiveness of imatinib in UM, which was originally anticipated based on the high levels of KIT and SCF expression.


Asunto(s)
Melanoma/tratamiento farmacológico , Melanoma/genética , Piperazinas/uso terapéutico , Proteínas Proto-Oncogénicas c-kit/metabolismo , Pirimidinas/uso terapéutico , Neoplasias de la Úvea/tratamiento farmacológico , Neoplasias de la Úvea/genética , Anciano , Anciano de 80 o más Años , Benzamidas , Femenino , Humanos , Mesilato de Imatinib , Masculino , Melanoma/patología , Melanoma/secundario , Persona de Mediana Edad , Mutación , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Factor de Células Madre/metabolismo , Regulación hacia Arriba , Neoplasias de la Úvea/metabolismo
5.
Curr Biol ; 15(18): 1629-39, 2005 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-16169484

RESUMEN

BACKGROUND: Malignant melanoma is a highly aggressive neoplastic disease whose incidence is increasing rapidly. In recent years, the use of interferon alpha (IFNalpha) has become the most established adjuvant immunotherapy for melanoma of advanced stage. IFNalpha is a potent inhibitor of melanoma cell proliferation, and the signal transducer and activator of transcription STAT1 is crucial for its antiproliferative action. Although advanced melanomas clinically resistant to IFNalpha are frequently characterized by inefficient STAT1 signaling, the mechanisms underlying advanced-stage interferon resistance are poorly understood. RESULTS: Here, we demonstrate that IFNalpha activates STAT5 in melanoma cells and that in IFNalpha-resistant cells STAT5 is overexpressed. Significantly, the knockdown of STAT5 in interferon-resistant melanoma cells restored the growth-inhibitory response to IFNalpha. When STAT5 was overexpressed in IFNalpha-sensitive cells, it counteracted interferon-induced growth inhibition. The overexpressed STAT5 diminished IFNalpha-triggered STAT1 activation, most evidently through upregulation of the inhibitor of cytokine-signaling CIS. CONCLUSIONS: Our data demonstrate that overexpression and activation of STAT5 enable melanoma cells to overcome cytokine-mediated antiproliferative signaling. Thus, overexpression of STAT5 can counteract IFNalpha signaling in melanoma cells, and this finally can result in cytokine-resistant and progressively growing tumor cells. These findings have significant implications for the clinical failure of IFNalpha therapy of advanced melanoma because they demonstrate that IFNalpha induces the activation of STAT5 in melanoma cells, and in STAT5-overexpressing cells, this contributes to IFNalpha resistance.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos/fisiología , Inmunoterapia/efectos adversos , Interferón-alfa/farmacología , Melanoma/metabolismo , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT5/metabolismo , Western Blotting , Línea Celular Tumoral , Citocinas/antagonistas & inhibidores , Cartilla de ADN , Resistencia a Antineoplásicos/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inmunohistoquímica , Inmunoprecipitación , Interferón-alfa/metabolismo , Melanoma/terapia , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos
6.
BMC Cancer ; 6: 7, 2006 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-16405733

RESUMEN

BACKGROUND: Since tacrolimus ointment was approved by the U.S. Food and Drug Administration (FDA) as a promising treatment for atopic dermatitis, it has been approved in more than 30 additional countries, including numerous European Union member nations. Moreover, in the current clinical routine the use of this drug is no longer restricted to the approved indication, but has been extended to a wide variety of inflammatory skin diseases including some with the potential of malignant transformation. So far, the side-effects reported from the topical use of tacrolimus have been relatively minor (e.g. burning, pruritus, erythema). Recently, however, the FDA reviewed the safety of topical tacrolimus, which resulted in a warning that the use of calcineurin inhibitors may be associated with an increased risk of cancer. CASE PRESENTATION: Oral lichen planus (OLP) was diagnosed in a 56-year-old women in February 1999. After several ineffective local and systemic therapeutic measures an off-label treatment of this recalcitrant condition using Tacrolimus 0.1% ointment was initiated in May 2002. After a few weeks of treatment most of the lesions ameliorated, with the exception of the plaques on the sides of the tongue. Nevertheless, the patient became free of symptoms which, however, reoccurred once tacrolimus was weaned, as a consequence treatment was maintained. In April 2005, the plaques on the left side of the tongue appeared increasingly compact and a biopsy specimen confirmed the suspected diagnosis of an oral squamous cell carcinoma. CONCLUSION: The suspected causal relationship between topical use of tacrolimus and the development of a squamous cell carcinoma prompted us to test the notion that the carcinogenicity of tacrolimus may go beyond mere immune suppression. To this end, tacrolimus has been shown to have an impact on cancer signalling pathways such as the MAPK and the p53 pathway. In the given case, we were able to demonstrate that these pathways had also been altered subsequent to tacrolimus therapy.


Asunto(s)
Carcinoma de Células Escamosas/inducido químicamente , Inmunosupresores/efectos adversos , Liquen Plano Oral/tratamiento farmacológico , Tacrolimus/efectos adversos , Neoplasias de la Lengua/inducido químicamente , Administración Tópica , Inhibidores de la Calcineurina , Transformación Celular Neoplásica/patología , Activación Enzimática , Femenino , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Inmunosupresores/farmacología , Liquen Plano Oral/patología , Persona de Mediana Edad , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Tacrolimus/farmacología , Ubiquitina-Proteína Ligasas
7.
Arch Dermatol Res ; 297(1): 26-30, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15906050

RESUMEN

Survivin, a member of the inhibitors of apoptosis protein family, regulates both cellular proliferation and apoptotic cell death. While the human survivin gene is highly expressed in the developing fetus, in adults its expression is restricted to highly proliferating normal tissues and neoplastic tumors tissues. In the present study, we compared the expression of survivin in melanoma and benign melanocytic lesions such as junctional, compound, dermal, congenital, blue and spitz nevi. This analysis reveals a heterogeneous expression of survivin with respect to both the intensity, frequency and cellular localization. In junctional, compound and blue nevi, survivin was present in nuclear localization, whereas in spitz nevi survivin was detectable in the cytoplasm. In dermal and congenital nevi, survivin was present in both localizations with predominance of the nuclear compartment. Interestingly, this distribution was similar to that observed in primary melanoma; whereas in metastatic melanoma the predominance of the nuclear localization of survivin was lost. Our data demonstrate that although survivin is expressed in a large number of benign nevi, the balance between its cytoplasmic and nuclear expression was immensely heterogeneous between lesions with suspected different developmental origins.


Asunto(s)
Núcleo Celular/química , Citoplasma/química , Melanoma/química , Proteínas Asociadas a Microtúbulos/análisis , Proteínas de Neoplasias/análisis , Nevo/química , Neoplasias Cutáneas/química , Humanos , Inmunohistoquímica , Proteínas Inhibidoras de la Apoptosis , Survivin
8.
J Invest Dermatol ; 118(4): 600-5, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11918705

RESUMEN

Cellular immune responses to melanoma are tightly regulated and include specific T cell responses to self antigens such as Mart-1 and gp100. Thus, additional signals apart from those mediated by the T cell receptor are needed to ensure T cell activation. Recently, the stress inducible major histocompatibility complex molecules, MHC class I related chain, were identified as an activator of both natural killer and T cells via interaction with their receptor NKG2D. Herein, we report the expression of MIC in 31 of 40 primary cutaneous melanomas and in 13 of 20 metastatic lesions. Moreover, lymphocytes infiltrating the tumor were found to express NKG2D. Detailed analysis identified both CD3+ T cells as well as CD56+ natural killer cells contributing to this NKG2D+ tumor infiltrating lymphocyte population present.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/biosíntesis , Melanoma/inmunología , Neoplasias Cutáneas/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Células Clonales , Femenino , Antígenos de Histocompatibilidad Clase I/análisis , Humanos , Inmunohistoquímica , Células Asesinas Naturales/química , Células Asesinas Naturales/metabolismo , Masculino , Melanoma/metabolismo , Melanoma/secundario , Persona de Mediana Edad , Subfamilia K de Receptores Similares a Lectina de Células NK , Receptores Inmunológicos/análisis , Receptores Inmunológicos/biosíntesis , Receptores de Células Asesinas Naturales , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/secundario , Estrés Fisiológico/inmunología , Linfocitos T/química , Linfocitos T/citología , Linfocitos T/metabolismo
9.
J Invest Dermatol ; 118(4): 595-9, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11918704

RESUMEN

Selection and activation of T cells is tightly regulated by both antigen-specific receptors and co-receptors to ensure that responses to self antigens are largely avoided. By T cell receptor clonotypic mapping and staining with tetrameric HLA-peptide complexes, we demonstrate the presence of melanocyte differentiation antigen MART-1 specific T cells in the areas of destruction of both neoplastic and normal melanocytic cells in a case of a primary melanoma and its associated hypopigmentation. These self reactive T cells expressed CD94/NKG2 major histocompatibility complex class I specific C-type lectin-like receptors. This family of receptors includes both activating and inhibitory isoforms. Thus, we performed a detailed analysis that revealed the exclusive presence of inhibitory NKG2-A/B receptors in the vitiligo-like leukoderma, whereas both the inhibitory receptors and the activating NKG2-C/E isoforms were present within the tumor. Our data suggest the differential expression of killer inhibitory receptors as a possible mechanism to regulate T cell responses to self antigens.


Asunto(s)
Lectinas Tipo C , Melanoma/inmunología , Neoplasias Cutáneas/inmunología , Linfocitos T/inmunología , Vitíligo/inmunología , Antígenos CD/genética , Antígenos de Neoplasias , Autoinmunidad/inmunología , Células Clonales , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Tolerancia Inmunológica/inmunología , Inmunohistoquímica , Antígeno MART-1 , Glicoproteínas de Membrana/genética , Subfamília C de Receptores Similares a Lectina de Células NK , Subfamília D de Receptores Similares a Lectina de las Células NK , Proteínas de Neoplasias/análisis , Receptores de Antígenos de Linfocitos T alfa-beta/análisis , Receptores Inmunológicos/genética , Receptores de Células Asesinas Naturales , Linfocitos T/química , Linfocitos T/citología
10.
BMC Cancer ; 4: 62, 2004 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-15361259

RESUMEN

BACKGROUND: Mutations of the BRAF gene are the most common genetic alteration in melanoma. Moreover, BRAF mutations are already present in benign nevi. Being overexpressed and mutated, B-Raf is a potential target for the immune system and as this mutation seems to be an early event, a humoral immune response against this antigen might serve as a diagnostic tool for detection of high risk patients. METHODS: 372 sera of 148 stage IV melanoma patients and 119 sera of non-melanoma patients were screened for B-Raf, B-Raf V599E and C-Raf specific antibodies by an ELISA assay. Sera were screened for specific total Ig and for IgG. Serum titers were compared with a two tailed Mann-Whitney U test. Sera with titers of 1:300 or higher were termed positive and groups were compared with a two tailed Fisher's exact test. RESULTS: B-Raf specific antibodies recognizing both B-Raf and B-Raf V599E were detected in 8.9% of the sera of melanoma patients and in 2,5% of the control group. Raf specific IgG was detected in some patients at very low levels. B-Raf specific antibody responses did not correlate with clinical parameters but in some cases, B-Raf antibodies emerged during disease progression. CONCLUSION: These findings imply that B-Raf is immunogenic in melanoma patients and that it might serve as a potential target for immunotherapy. However, B-Raf specific antibodies emerge at rather late stages of melanoma progression and are present only with a low frequency indicating that spontaneous B-Raf specific antibodies are not an early marker for melanoma, but rather may serve as a therapeutic target.


Asunto(s)
Anticuerpos Antineoplásicos/sangre , Melanoma/inmunología , Proteínas Proto-Oncogénicas B-raf/inmunología , Neoplasias Cutáneas/inmunología , Anticuerpos Antineoplásicos/inmunología , Especificidad de Anticuerpos , Ensayo de Inmunoadsorción Enzimática , Humanos , Inmunoglobulina G/sangre , Neoplasias Cutáneas/sangre
12.
Cancer Immunol Immunother ; 55(5): 481-94, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16220326

RESUMEN

Immunotherapy has been widely investigated for its potential use in cancer therapy and it becomes more and more apparent that the selection of target antigens is essential for its efficacy. Indeed, limited clinical efficacy is partly due to immune evasion mechanisms of neoplastic cells, e.g. downregulation of expression or presentation of the respective antigens. Consequently, antigens contributing to tumor cell survival seem to be more suitable therapeutic targets. However, even such antigens may be subject to immune evasion due to impaired processing and cell surface expression. Since development and progression of tumors is not only dependent on cancer cells themselves but also on the active contribution of the stromal cells, e.g. by secreting growth supporting factors, enzymes degrading the extracellular matrix or angiogenic factors, the tumor stroma may also serve as a target for immune intervention. To this end several antigens have been identified which are induced or upregulated on the tumor stroma. Tumor stroma-associated antigens are characterized by an otherwise restricted expression pattern, particularly with respect to differentiated tissues, and they have been successfully targeted by passive and active immunotherapy in preclinical models. Moreover, some of these strategies have already been translated into clinical trials.


Asunto(s)
Antígenos de Neoplasias/inmunología , Inmunoterapia , Neoplasias/inmunología , Neoplasias/terapia , Células del Estroma/inmunología , Animales , Células Endoteliales/inmunología , Fibroblastos/inmunología , Humanos , Macrófagos/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA