Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 513
Filtrar
1.
JAMA Netw Open ; 6(7): e2324977, 2023 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-37505498

RESUMEN

Importance: The development of oncology drugs is expensive and beset by a high attrition rate. Analysis of the costs and causes of translational failure may help to reduce attrition and permit the more appropriate use of resources to reduce mortality from cancer. Objective: To analyze the causes of failure and expenses incurred in clinical trials of novel oncology drugs, with the example of insulin-like growth factor-1 receptor (IGF-1R) inhibitors, none of which was approved for use in oncology practice. Design, Setting, and Participants: In this cross-sectional study, inhibitors of the IGF-1R and their clinical trials for use in oncology practice between January 1, 2000, and July 31, 2021, were identified by searching PubMed and ClinicalTrials.gov. A proprietary commercial database was interrogated to provide expenses incurred in these trials. If data were not available, estimates were made of expenses using mean values from the proprietary database. A search revealed studies of the effects of IGF-1R inhibitors in preclinical in vivo assays, permitting calculation of the percentage of tumor growth inhibition. Archival data on the clinical trials of IGF-1R inhibitors and proprietary estimates of their expenses were examined, together with an analysis of preclinical data on IGF-1R inhibitors obtained from the published literature. Main Outcomes and Measures: Expenses associated with research and development of IGF-1R inhibitors. Results: Sixteen inhibitors of IGF-1R studied in 183 clinical trials were found. None of the trials, in a wide range of tumor types, showed efficacy permitting drug approval. More than 12 000 patients entered trials of IGF-1R inhibitors in oncology indications in 2003 to 2021. These trials incurred aggregate research and development expenses estimated at between $1.6 billion and $2.3 billion. Analysis of the results of preclinical in vivo assays of IGF-1R inhibitors that supported subsequent clinical investigations showed mixed activity and protocols that poorly reflected the treatment of advanced metastatic tumors in humans. Conclusions and Relevance: Failed drug development in oncology incurs substantial expense. At an industry level, an estimated $50 billion to $60 billion is spent annually on failed oncology trials. Improved target validation and more appropriate preclinical models are required to reduce attrition, with more attention to decision-making before launching clinical trials. A more appropriate use of resources may better reduce cancer mortality.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina , Neoplasias , Humanos , Estudios Transversales , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico
2.
Genes (Basel) ; 13(1)2022 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-35052471

RESUMEN

OBJECTIVE: Women with polycystic ovary syndrome (PCOS) are at increased risk ofendometrial carcinoma (EC). Previous studies indicated that the combined therapy of Diane-35 and metformin significantly suppresses disease progression in PCOS patients with early EC; however, the mechanisms remain unclear. METHODS: An established murine model of PCOS with early EC, clinical specimens, and human EC cells was used in this study. The levels of protein and mRNA were measured with Western blotting and RT-PCR, respectively. Cell proliferation was determined with MTT, colony formation, and flow cytometry. Proteins were analyzed with immunofluorescence and immunohistochemistry. RESULTS: Diane-35 and metformin significantly inhibited proliferative activity and promoted apoptosis in EC cells. Additionally, cell autophagy was induced by the combined therapy. Quantitive PCR revealed that Diane-35 and metformin decreased androgen receptor (AR) expression but elevated GLUT4 expression. AR was found to repress GLUT4 expression by binding to the promoter of GLUT4. Moreover, the combined treatment mediated the onset of cellular autophagy by regulating the mTORC pathway via the suppression of IGF-1 and inhibited the development of EC by the activation of the PI3K/mTORC pathway. CONCLUSION: The results and previous clinical evidence support the use of Diane-35 and metformin combination therapy for patients with PCOS and early EC.


Asunto(s)
Apoptosis , Autofagia , Acetato de Ciproterona/farmacología , Neoplasias Endometriales/tratamiento farmacológico , Etinilestradiol/farmacología , Metformina/farmacología , Síndrome del Ovario Poliquístico/complicaciones , Antagonistas de Andrógenos/farmacología , Animales , Combinación de Medicamentos , Quimioterapia Combinada , Neoplasias Endometriales/etiología , Neoplasias Endometriales/metabolismo , Neoplasias Endometriales/patología , Femenino , Transportador de Glucosa de Tipo 4/genética , Transportador de Glucosa de Tipo 4/metabolismo , Hipoglucemiantes/farmacología , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Ratones , Receptores Androgénicos/química
3.
Mol Med Rep ; 24(2)2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34184083

RESUMEN

M2­like tumour­associated macrophages (TAMs) have been demonstrated to promote the growth of anaplastic thyroid carcinoma (ATC). However, the underlying mechanism of M2­like TAMs in ATC remains unclear. Thus, in the present study, the role and mechanism of M2­like TAMs in ATC were investigated. M2­like TAMs were induced by treatment with PMA, plus IL­4 and IL­13, and identified by flow cytometry. Transwell and sphere formation assays were applied to assess the invasion and stemness of ATC cells. The expression levels of insulin­like growth factor (IGF)­1 and IGF­2 were examined by ELISA and reverse transcription­quantitative PCR. Proteins related to the epithelial­mesenchymal transition (EMT), stemness and the PI3K/AKT/mTOR pathway were examined via western blotting. Immunohistochemistry (IHC) was used to detect the expression of the M2­like TAM markers CD68 and CD206 in ATC tissues and thyroid adenoma tissues. It was found that treatment with PMA plus IL­4 and IL­13 successfully induced M2­like TAMs. Following co­culture with M2­like TAMs, the invasive ability and stemness of ATC cells were significantly increased. The expression levels of the EMT­related markers N­cadherin and Vimentin, the stemness­related markers Oct4, Sox2 and CD133, and the insulin receptor (IR)­A/IGF1 receptor (IGF1R) were markedly upregulated, whereas E­cadherin expression was significantly decreased. In addition, the production of IGF­1 and IGF­2 was significantly increased. Of note, exogenous IGF­1/IGF­2 promoted the invasion and stemness of C643 cells, whereas blocking IGF­1 and IGF­2 inhibited metastasis and stemness by repressing IR­A/IGF­1R­mediated PI3K/AKT/mTOR signalling in the co­culture system. IHC results showed that the expression of CD68 and CD206 was obviously increased in ATC tissues. To conclude, M2­like TAMs accelerated the metastasis and increased the stemness of ATC cells, and the underlying mechanism may be related to the section of IGF by M2­like TAMs, which activates the IR­A/IGF1R­mediated PI3K/AKT/mTOR signalling pathway.


Asunto(s)
Células Madre Neoplásicas , Transducción de Señal , Somatomedinas/metabolismo , Carcinoma Anaplásico de Tiroides/metabolismo , Neoplasias de la Tiroides/metabolismo , Macrófagos Asociados a Tumores/metabolismo , Adulto , Anciano , Anticuerpos Neutralizantes/farmacología , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Línea Celular , Cromonas/farmacología , Femenino , Humanos , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Factor II del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Factor II del Crecimiento Similar a la Insulina/genética , Factor II del Crecimiento Similar a la Insulina/metabolismo , Factor II del Crecimiento Similar a la Insulina/farmacología , Masculino , Glicoproteínas de Membrana/metabolismo , Persona de Mediana Edad , Morfolinas/farmacología , Invasividad Neoplásica/inmunología , Metástasis de la Neoplasia/inmunología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/inmunología , Células Madre Neoplásicas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor de Insulina/antagonistas & inhibidores , Receptor de Insulina/metabolismo , Receptores Inmunológicos/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Somatomedinas/genética , Serina-Treonina Quinasas TOR/metabolismo , Carcinoma Anaplásico de Tiroides/inmunología , Neoplasias de la Tiroides/inmunología , Neoplasias de la Tiroides/patología , Adulto Joven
4.
Am J Physiol Cell Physiol ; 321(1): C82-C93, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34038245

RESUMEN

The resolution of inflammation is closely linked with tissue repair. Recent studies have revealed that macrophages suppress inflammatory reactions by producing lipid mediators, called specialized proresolving mediators (SPMs); however, the biological significance of SPMs in tissue repair remains to be fully elucidated in the heart. In this study, we focused on maresin-1 (MaR1) and examined the reparative effects of MaR1 in cardiomyocytes. The treatment with MaR1 increased cell size in cultured neonatal rat cardiomyocytes. Since the expression of fetal cardiac genes was unchanged by MaR1, physiological hypertrophy was induced by MaR1. SR3335, an inhibitor of retinoic acid-related orphan receptor α (RORα), mitigated MaR1-induced cardiomyocyte hypertrophy, consistent with the recent report that RORα is one of MaR1 receptors. Importantly, in response to MaR1, cardiomyocytes produced IGF-1 via RORα. Moreover, MaR1 activated phosphoinositide 3-kinase (PI3K)/Akt signaling pathway and wortmannin, a PI3K inhibitor, or triciribine, an Akt inhibitor, abrogated MaR1-induced cardiomyocyte hypertrophy. Finally, the blockade of IGF-1 receptor by NVP-AEW541 inhibited MaR-1-induced cardiomyocyte hypertrophy as well as the activation of PI3K/Akt pathway. These data indicate that MaR1 induces cardiomyocyte hypertrophy through RORα/IGF-1/PI3K/Akt pathway. Considering that MaR1 is a potent resolving factor, MaR1 could be a key mediator that orchestrates the resolution of inflammation with myocardial repair.


Asunto(s)
Cardiomegalia/genética , Cardiotónicos/farmacología , Ácidos Docosahexaenoicos/efectos adversos , Factor I del Crecimiento Similar a la Insulina/genética , Infarto del Miocardio/genética , Miocitos Cardíacos/efectos de los fármacos , Comunicación Paracrina/genética , Animales , Cardiomegalia/inducido químicamente , Cardiomegalia/patología , Cardiomegalia/prevención & control , Modelos Animales de Enfermedad , Ácidos Docosahexaenoicos/antagonistas & inhibidores , Regulación de la Expresión Génica , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/inducido químicamente , Infarto del Miocardio/patología , Infarto del Miocardio/prevención & control , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/antagonistas & inhibidores , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Comunicación Paracrina/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Cultivo Primario de Células , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirimidinas/farmacología , Pirroles/farmacología , Ratas , Ribonucleósidos/farmacología , Transducción de Señal , Sulfonamidas/farmacología , Tiofenos/farmacología , Wortmanina/farmacología
5.
J Mol Neurosci ; 71(11): 2299-2309, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33484421

RESUMEN

Pyroptosis is a programmed cell death process which is accompanied by inflammation. The aims of this in vitro and in vivo study were to reveal whether miR-129 contributed to neuronal pyroptosis and cognitive impairment and to further explore its mechanism involved. PC-12 cells were treated with LPS, miR-129 antagomir, AXL1717 (IGF-1 receptor blocker), or SB216763 (GSK3ß blocker). After that, expression of miR-129 was measured using qRT-PCR. Relationship between miR-129 and IGF-1 was revealed using luciferase reporter assay. Protein expression of IGF-1, p-Ser9-GSK3ß, NLRP3, and Caspase-1 was determined using western blotting. Pyroptosis rate was measured using flow cytometry. Wistar rats were fed with high-fat diet to induce neural inflammation and were further treated with miR-129 antagomir through intracerebroventricular injection. Then, cognitive impairment was assessed by water maze test. Expression of the proteins mentioned above was measured again in midbrain and hippocampus of the rats. In the PC-12 cells, LPS-induced neuronal pyroptosis can be alleviated by miR-129 antagomir. IGF-1 was a specific target for miR-129. Up-regulation and down-regulation of IGF-1/GSK3ß signaling pathway separately alleviated and deteriorated neuronal pyroptosis in the cells. In the rats, high-fat diet caused cognitive impairment following with neuronal pyroptosis and down-regulation of IGF-1/GSK3ß signaling pathway in midbrain and hippocampus tissues. Also, miR-129 antagomir improved these abnormalities in the rats. Inhibition of miR-129 improved neuronal pyroptosis and cognitive impairment through IGF-1/GSK3ß signaling pathway.


Asunto(s)
Disfunción Cognitiva/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , MicroARNs/genética , Neuronas/metabolismo , Piroptosis , Animales , Disfunción Cognitiva/etiología , Disfunción Cognitiva/metabolismo , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Lipopolisacáridos/toxicidad , Masculino , MicroARNs/metabolismo , Células PC12 , Ratas , Ratas Wistar , Transducción de Señal
6.
Mol Med Rep ; 22(5): 3804-3812, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33000209

RESUMEN

Glioma is a malignant brain cancer that exhibits high invasive ability and poor prognosis. MicroRNA (miR)­181d has been reported to be involved in the development of glioma. Therefore, the aim of the present study was to investigate whether miR­181d affected cellular progression by influencing the insulin like growth factor (IGF1)/PI3K/AKT axis. Western blot analysis was performed to analyze the expression levels of specific proteins, and a Cell Counting Kit­8 assay was used to assess the proliferative ability of cells. Cell cycle progression and cellular apoptosis were both measured using flow cytometry. The results indicated that miR­181d promoted cellular proliferation and cell cycle progression, while suppressing cellular apoptosis via the IGF1/PI3K/AKT axis. It was demonstrated that the IGF1 and PI3K/AKT inhibitors reversed these observed functions of miR­181d. Furthermore, miR­181d enhanced the growth of glioma xenografts in vivo, promoted cell cycle progression and suppressed cellular apoptosis within glioma xenograft tissues. Therefore, this newly identified miR­181d/IGF1/PI3K/AKT axis may provide novel insights into the pathogenesis of glioma.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Proliferación Celular/genética , Glioma/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , MicroARNs/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/genética , Animales , Apoptosis/genética , Neoplasias Encefálicas/patología , Ciclo Celular/genética , Línea Celular Tumoral , Cromonas/farmacología , Glioma/patología , Humanos , Imidazoles/farmacología , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Morfolinas/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Pirazinas/farmacología , Transfección , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
7.
J Biol Chem ; 295(45): 15292-15306, 2020 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-32868454

RESUMEN

Muscle atrophy is regulated by the balance between protein degradation and synthesis. FOXO1, a transcription factor, helps to determine this balance by activating pro-atrophic gene transcription when present in muscle fiber nuclei. Foxo1 nuclear efflux is promoted by AKT-mediated Foxo1 phosphorylation, eliminating FOXO1's atrophy-promoting effect. AKT activation can be promoted by insulin-like growth factor 1 (IGF1) or insulin via a pathway including IGF1 or insulin, phosphatidylinositol 3-kinase, and AKT. We used confocal fluorescence time-lapse imaging of FOXO1-GFP in adult isolated living muscle fibers maintained in culture to explore the effects of IGF1 and insulin on FOXO1-GFP nuclear efflux with and without pharmacological inhibitors. We observed that although AKT inhibitor blocks the IGF1- or insulin-induced effect on FOXO1 nuclear efflux, phosphatidylinositol 3-kinase inhibitors, which we show to be effective in these fibers, do not. We also found that inhibition of the protein kinase ACK1 or ATM contributes to the suppression of FOXO1 nuclear efflux after IGF1. These results indicate a novel pathway that has been unexplored in the IGF1- or insulin-induced regulation of FOXO1 and present information useful both for therapeutic interventions for muscle atrophy and for further investigative areas into insulin insensitivity and type 2 diabetes.


Asunto(s)
Núcleo Celular/metabolismo , Proteína Forkhead Box O1/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Insulina/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Animales , Núcleo Celular/efectos de los fármacos , Femenino , Proteína Forkhead Box O1/antagonistas & inhibidores , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Ratones , Fibras Musculares Esqueléticas/efectos de los fármacos , Imagen Óptica , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos
8.
Growth Horm IGF Res ; 55: 101341, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32890915

RESUMEN

OBJECTIVE: Insulin-like growth factor 1 (IGFI) is one of several growth factors which is induced by growth hormone (GH), which activates the Janus kinase 2 (JAK2)-signal transducer and activator of transcription 5 (STAT5) pathway, and plays crucial roles in normal human growth, metabolism, and systemic energy homeostasis. However, little is known about the negative regulation of IGF-I production under different physiological or pathological conditions. Herein, we explore whether activation of endoplasmic reticulum (ER) stress regulates IGF-I production and normal body growth. MATERIALS AND METHODS: C57BL/6 J mice were challenged with tunicamycin (Tm) to induce ER stress activation. 24 h after stimulation, hepatic mRNA expression was analyzed by RNA-Seq and validated by qPCR. Enzyme-linked immunosorbent assay (ELISA) was performed 24 h after Tm stimulation. Body growth was determined 16 days after Tm stimulation. Animals were then sacrificed and liver tissues were collected for further analysis. RESULTS: Mice challenged with Tm displayed a retardation of growth. Molecularly, we found that ER stress inhibited phosphorylation of STAT5. IGF-I transcription and circulating IGF-I were also dramatically decreased under ER stress activation. Moreover, our results demonstrate that IGF-I administration ameliorates Tm-induced growth retardation. CONCLUSIONS: ER stress induces growth retardation. ER stress inhibits hepatic GH-JAK2 signaling activation and its downstream target gene expression. These results warrant further research to explore the crosstalk between ER stress and growth hormone signaling in improving body growth.


Asunto(s)
Estrés del Retículo Endoplásmico , Trastornos del Crecimiento/patología , Hormona del Crecimiento/antagonistas & inhibidores , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Animales , Trastornos del Crecimiento/etiología , Trastornos del Crecimiento/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL
9.
J Endocrinol ; 246(3): 277-288, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32698133

RESUMEN

Timely adjustment of osmoregulation upon acute salinity stress is essential for the survival of euryhaline fish. This rapid response is thought to be tightly controlled by hormones; however, there are still questions unanswered. In this work, we tested the hypothesis that the endocrine hormone, insulin-like growth factor 1 (Igf1), a slow-acting hormone, is involved in the activation of salt secretion mechanisms in euryhaline medaka (Oryzias melastigma) during acclimation to acute salinity stress. In response to a 30-ppt seawater (SW) challenge, Na+/Cl- secretion was enhanced within 0.5 h, with concomitant organization of ionocyte multicellular complexes and without changes in expression of major transporters. Igf1 receptor inhibitors significantly impair the Na+/Cl- secretion and ionocyte multicellular complex responses without affecting transporter expression. Thus, Igf1 may activate salt secretion as part of the teleost response to acute salinity stress by exerting effects on transporter function and enhancing the formation of ionocyte multicellular complexes. These findings provide new insights into hormonal control of body fluid ionic/osmotic homeostasis during vertebrate evolution.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina/metabolismo , Cloruro de Sodio/farmacología , Animales , Proteínas de Peces/metabolismo , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Oryzias , Salinidad , Estrés Salino , Transducción de Señal/efectos de los fármacos
10.
Adv Cancer Res ; 147: 229-257, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32593402

RESUMEN

The insulin and insulin-like growth factor (IGF) family of proteins are part of a complex network that regulates cell proliferation and survival. While this system is undoubtedly important in prenatal development and postnatal cell growth, members of this family have been implicated in several different cancer types. Increased circulating insulin and IGF ligands have been linked to increased risk of cancer incidence. This observation has led to targeting the IGF system as a therapeutic strategy in a number of cancers. This chapter aims to describe the well-characterized biology of the IGF1R system, outline the rationale for targeting this system in cancer, summarize the clinical data as it stands, and discuss where we can go from here.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Receptor IGF Tipo 1/antagonistas & inhibidores , Animales , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Terapia Molecular Dirigida , Neoplasias/patología , Receptor IGF Tipo 1/metabolismo , Transducción de Señal
11.
Br J Cancer ; 122(9): 1324-1332, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32161368

RESUMEN

BACKGROUND: Xentuzumab, an insulin-like growth factor (IGF)-1/IGF-2-neutralising antibody, binds IGF-1 and IGF-2, inhibiting their growth-promoting signalling. Two first-in-human trials assessed the maximum-tolerated/relevant biological dose (MTD/RBD), safety, pharmacokinetics, pharmacodynamics, and activity of xentuzumab in advanced/metastatic solid cancers. METHODS: These phase 1, open-label trials comprised dose-finding (part I; 3 + 3 design) and expansion cohorts (part II; selected tumours; RBD [weekly dosing]). Primary endpoints were MTD/RBD. RESULTS: Study 1280.1 involved 61 patients (part I: xentuzumab 10-1800 mg weekly, n = 48; part II: 1000 mg weekly, n = 13); study 1280.2, 64 patients (part I: 10-3600 mg three-weekly, n = 33; part II: 1000 mg weekly, n = 31). One dose-limiting toxicity occurred; the MTD was not reached for either schedule. Adverse events were generally grade 1/2, mostly gastrointestinal. Xentuzumab showed dose-proportional pharmacokinetics. Total plasma IGF-1 increased dose dependently, plateauing at ~1000 mg/week; at ≥450 mg/week, IGF bioactivity was almost undetectable. Two partial responses occurred (poorly differentiated nasopharyngeal carcinoma and peripheral primitive neuroectodermal tumour). Integration of biomarker and response data by Bayesian Logistic Regression Modeling (BLRM) confirmed the RBD. CONCLUSIONS: Xentuzumab was well tolerated; MTD was not reached. RBD was 1000 mg weekly, confirmed by BLRM. Xentuzumab showed preliminary anti-tumour activity. CLINICAL TRIAL REGISTRATION: NCT01403974; NCT01317420.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Factor II del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Adulto , Anciano , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Neutralizantes/administración & dosificación , Anticuerpos Neutralizantes/inmunología , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Factor I del Crecimiento Similar a la Insulina/inmunología , Factor II del Crecimiento Similar a la Insulina/inmunología , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Neoplasias/genética , Neoplasias/patología , Adulto Joven
12.
Drugs ; 80(5): 509-512, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32157641

RESUMEN

Teprotumumab (teprotumumab-trbw; TEPEZZA™ - Horizon Therapeutics) is a monoclonal antibody insulin-like growth factor-I receptor (IGF-IR) antagonist developed for the treatment of thyroid eye disease (Graves ophthalmopathy/orbitopathy, thyroid-associated ophthalmopathy). Based on positive results from two multinational clinical trials teprotumumab was recently approved for this indication in the US. This article summarizes the milestones in the development of teprotumumab leading to this first approval for thyroid eye disease.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Aprobación de Drogas , Oftalmopatía de Graves/tratamiento farmacológico , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Anticuerpos Monoclonales Humanizados/química , Humanos
13.
Mol Cancer Ther ; 19(4): 1059-1069, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32054790

RESUMEN

Androgen deprivation therapy and second-generation androgen receptor signaling inhibitors such as enzalutamide are standard treatments for advanced/metastatic prostate cancer. Unfortunately, most men develop resistance and relapse; signaling via insulin-like growth factor (IGF) has been implicated in castration-resistant prostate cancer. We evaluated the antitumor activity of xentuzumab (IGF ligand-neutralizing antibody), alone and in combination with enzalutamide, in prostate cancer cell lines (VCaP, DuCaP, MDA PCa 2b, LNCaP, and PC-3) using established in vitro assays, and in vivo, using LuCaP 96CR, a prostate cancer patient-derived xenograft (PDX) model. Xentuzumab + enzalutamide reduced the viability of phosphatase and tensin homolog (PTEN)-expressing VCaP, DuCaP, and MDA PCa 2b cells more than either single agent, and increased antiproliferative activity and apoptosis induction in VCaP. Xentuzumab or xentuzumab + enzalutamide inhibited IGF type 1 receptor and AKT serine/threonine kinase (AKT) phosphorylation in VCaP, DuCaP, and MDA PCa 2b cells; xentuzumab had no effect on AKT phosphorylation and proliferation in PTEN-null LNCaP or PC-3 cells. Knockdown of PTEN led to loss of antiproliferative activity of xentuzumab and reduced activity of xentuzumab + enzalutamide in VCaP cells. Xentuzumab + enzalutamide inhibited the growth of castration-resistant LuCaP 96CR PDX with acquired resistance to enzalutamide, and improved survival in vivo The data suggest that xentuzumab + enzalutamide combination therapy may overcome castration resistance and could be effective in patients who are resistant to enzalutamide alone. PTEN status as a biomarker of responsiveness to combination therapy needs further investigation.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Neutralizantes/farmacología , Factor II del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Feniltiohidantoína/análogos & derivados , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Animales , Apoptosis , Benzamidas , Ciclo Celular , Proliferación Celular , Quimioterapia Combinada , Humanos , Masculino , Ratones , Ratones SCID , Nitrilos , Feniltiohidantoína/farmacología , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Laryngoscope ; 130(6): 1470-1478, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31433065

RESUMEN

OBJECTIVES: The insulin-like growth factor-1 receptor (IGF1R) has been implicated in therapeutic resistance in head and neck squamous cell carcinoma (HNSCC), and small molecule tyrosine kinase inhibitors (TKIs) of IGF1R activity may have anticancer activity. Therefore, the relationship between survival and IGF1R expression was assessed for oral cavity (OC) cancer, and the antitumor effects of two IGF1R-TKIs, OSI-906 and BMS-754807, were evaluated in HNSCC cell lines in vitro. METHODS: Clinical outcome data and tissue microarray immunohistochemistry were used to generate IGF1R expression-specific survival curves. Immunoblot, alamarBlue proliferation assay, trypan blue exclusion viability test, clonogenic assay, flow cytometry, and reverse phase protein array (RPPA) were used to evaluate in vitro responses to IGF1R-TKIs. RESULTS: For patients with stage III/IV OCSCC, higher IGF1R expression was associated with poorer overall 5-year survival (P = 0.029). Both BMS-754807 and OSI-906 caused dose-dependent inhibition of IGF1R and Akt phosphorylation and inhibited proliferation; BMS-754807 was more potent than OSI-906. Both drugs reduced HNSCC cell viability; only OSI-906 was able to eliminate all viable cells at 10 µM. The two drugs similarly inhibited clonogenic cell survival. At 1 µM, only BMS-754807 caused a fourfold increase in the basal apoptotic rate. RPPA demonstrated broad effects of both drugs on canonical IGF1R signaling pathways and also inhibition of human epidermal growth factor receptor-3 (HER3), Src, paxillin, and ezrin phosphorylation. CONCLUSION: OSI-906 and BMS-754807 inhibit IGF1R activity in HNSCC cell lines with reduction in prosurvival and proliferative signaling and with concomitant antiproliferative and proapoptotic effects. Such antagonists may have utility as adjuvants to existing therapies for HNSCC. LEVEL OF EVIDENCE: NA Laryngoscope, 130:1470-1478, 2020.


Asunto(s)
Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Imidazoles/uso terapéutico , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Pirazinas/uso terapéutico , Pirazoles/uso terapéutico , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Triazinas/uso terapéutico , Neoplasias de Cabeza y Cuello/patología , Humanos , Imidazoles/farmacología , Factor I del Crecimiento Similar a la Insulina/biosíntesis , Neoplasias de la Boca/tratamiento farmacológico , Estadificación de Neoplasias , Pirazinas/farmacología , Pirazoles/farmacología , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Resultado del Tratamiento , Triazinas/farmacología , Células Tumorales Cultivadas
15.
J Neuroendocrinol ; 31(12): e12810, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31715027

RESUMEN

The onset of puberty is the result of an increase in secretion of hypothalamic gonadotrophin-releasing hormone (GnRH). This action is a result of not only the development of stimulatory inputs to its release, but also the gradual decrease in inhibitory inputs that restrain release of the peptide prior to pubertal onset. Dynorphin (DYN) is one of the inhibitory inputs produced in the medial basal hypothalamus (MBH); however, little is known about what substance(s) control its prepubertal synthesis and release. Because neurokinin B (NKB) increases in the hypothalamus as puberty approaches, we considered it a candidate for such a role. An initial study investigated the acute effects of an NKB agonist, senktide, on the secretion of DYN from MBH tissues incubated in vitro. In other experiments, central injections of senktide were administered to animals for 4 days then MBHs were collected for assessment of DYN synthesis or for the in vitro secretion of both DYN and GnRH. Because insulin-like growth factor (IGF)-1 has been shown to play an important role at puberty, additional animals received central injections of this peptide for 4 days to assess NKB and DYN synthesis or the in vitro secretion of NKB. The results obtained show that senktide administration up-regulates the NKB receptor protein, at the same time as suppressing the DYN and its receptor. Senktide consistently suppressed DYN and elevated GnRH secretion in the same tissue incubates from both the acute and chronic studies. IGF-1 administration caused an increase in NKB protein, at the same time as decreasing DYN protein. Furthermore, the central administration of IGF-1 caused an increase in NKB release, an action blocked by the IGF-1 receptor blocker, JB-1. These results indicate that the IGF-1/NKB pathway contributes to suppressing the DYN inhibitory tone on prepubertal GnRH secretion and thus facilitates the puberty-related increase in the release of GnRH to accelerate the onset of puberty.


Asunto(s)
Dinorfinas/metabolismo , Hipotálamo Medio/metabolismo , Animales , Femenino , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo Medio/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/administración & dosificación , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Factor I del Crecimiento Similar a la Insulina/farmacología , Microinyecciones , Neuroquinina B/metabolismo , Fragmentos de Péptidos/farmacología , Péptidos/farmacología , Ratas , Receptor IGF Tipo 1/antagonistas & inhibidores , Receptores de Neuroquinina-3/biosíntesis , Receptores Opioides/biosíntesis , Maduración Sexual , Sustancia P/análogos & derivados , Sustancia P/farmacología , Regulación hacia Arriba
16.
Drug Des Devel Ther ; 13: 3161-3170, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31564832

RESUMEN

PURPOSE: To investigate the role of IGF-1 signaling pathway in the treatment of uterine leiomyomas with mifepristone. PATIENTS AND METHODS: From October 2015 to December 2018, 50 patients with uterine leiomyoma were included in this study. Overexpression or siRNA of IGF-1 in primary human uterine leiomyoma cells were treated with or without mifepristone. MTT was used to evaluate cell viability in assays of cell proliferation and cytotoxicity. IGF-1 expression in the cells was measured with real-time RT-PCR and Western blotting and manipulated with lentivirus ectopic overexpression or siRNA silencing. RESULTS: Inhibition of cell viability by mifepristone was found dependent on drug concentration and treatment time. IGF-1 and phosphorylation-ERK1/2 expression were decreased, while phosphorylation-AKT expression was increased after mifepristone treatment. IGF-1 significantly promoted cell growth, while IGF-1 knockdown and mifepristone showed synergistic inhibition effects on cell growth. The overexpression of IGF-1 reversed the inhibition of cell growth and ERK1/2 phosphorylation but showed no effect on AKT phosphorylation. CONCLUSION: Our study for the first time demonstrated that IGF-1 signaling via ERK1/2 appears to be an important target of mifepristone in the treatment of uterine leiomyomas, which may provide a new approach to avoid leiomyoma re-growth after cessation of mifepristone.


Asunto(s)
Antineoplásicos/farmacología , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Leiomioma/tratamiento farmacológico , Mifepristona/farmacología , Transducción de Señal/efectos de los fármacos , Neoplasias Uterinas/tratamiento farmacológico , Adulto , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Leiomioma/metabolismo , Leiomioma/patología , Células Tumorales Cultivadas , Neoplasias Uterinas/metabolismo , Neoplasias Uterinas/patología
17.
Anticancer Drugs ; 30(7): e0744, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31305291

RESUMEN

Sevoflurane is a new type of inhalation anesthetic used widely in the clinic. It has the characteristics of rapid induction, rapid recovery, and less irritative to the airway. Studies have shown that sevoflurane can affect the invasion and migration of a variety of malignant tumors. However, its effects on human glioma cells and related mechanisms are not clear. Cultured U251 and U87 cells were pretreated with sevoflurane. The effect of sevoflurane on proliferation was evaluated by MTT, and cell migration assay, cell apoptosis, and invasion ability were evaluated by wound-healing assay, cell apoptosis, and Transwell assays. Insulin-like growth factor-1 (IGF-1) and PI3K/AKT signaling pathway gene expression in sevoflurane-treated cell lines was measured by western blotting analysis, respectively. 5% sevoflurane significantly inhibited proliferation ability in both U251 and U87 cells. Sevoflurane inhibited glioma cells invasion and migration, and promoted apoptosis. Sevoflurane inhibited IGF-1 and promoted the expression of apoptosis-related proteins in glioma cells. In addition, sevoflurane inhibited the PI3K/AKT signaling pathway in glioma cells. This study clarifies that sevoflurane inhibits proliferation, invasion, and migration, and promotes apoptosis in glioma cells. These effects are regulated by IGF-1, an upstream gene of the PI3K/AKT signaling pathway. These findings may be significant for the selection of anesthetic agents in glioma surgery to improve the prognosis of patients.


Asunto(s)
Apoptosis , Proliferación Celular , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/patología , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sevoflurano/farmacología , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Movimiento Celular , Glioma/genética , Glioma/metabolismo , Humanos , Invasividad Neoplásica , Fosfatidilinositol 3-Quinasas/genética , Inhibidores de Agregación Plaquetaria/farmacología , Proteínas Proto-Oncogénicas c-akt/genética , Células Tumorales Cultivadas
18.
Am J Physiol Endocrinol Metab ; 317(4): E686-E698, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31361542

RESUMEN

This study investigated the role of insulin-like growth factor-1/insulin-like growth factor-1 receptor (IGF-1/IGF-1R) in the genesis and progression of diabetic kidney disease (DKD) in a streptozotocin (STZ)-induced mouse diabetes model. We showed elevated IGF-1 expression in the DKD kidneys after 16 wk of diabetic onset. Intraperitoneal administration of IGF-1R inhibitor (glycogen synthase kinase-3ß, GSK4529) from week 8 to week 16 postdiabetes induction ameliorated urinary albumin excretion and kidney histological changes due to diabetes, including amelioration of glomerulomegaly, inflammatory infiltration, and tubulointerstitial fibrosis. The GSK4529 treatment also attenuated alterations in renal tubular expression of E-cad and matrix protein fibronectin. Moreover, renal fibrosis in DKD (without treatment) was associated with Snail1 overexpression that was effectively prevented by IGF-1R inhibition. Further experiments in cultured renal epithelial cells (NRK) showed that IGF-1 silencing reproduced in vivo effects of IGF-1R inhibition with markedly attenuated Snail1 expression and near normalization of the Ecad1 and fibronectin expression pattern. Further Snail1 silencing prevented high-glucose-induced changes without affecting IGF-1 expression, consistent with Snail1 acting downstream to IGF-1. The antifibrotic effects were also shown with benazepril or insulin treatment but to a much lesser degree. In summary, in STZ-induced diabetic mice, activation of IGF-1 in diabetic kidneys induces fibrogenesis through Snail1 upregulation. The diabetes-related histological and functional changes, as well as fibrogenesis, can be attenuated by IGF-1/IGF-1R inhibition.


Asunto(s)
Nefropatías Diabéticas/genética , Nefropatías Diabéticas/prevención & control , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Receptor IGF Tipo 1/antagonistas & inhibidores , Factores de Transcripción de la Familia Snail/biosíntesis , Animales , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Nefropatías Diabéticas/metabolismo , Fibrosis/patología , Fibrosis/prevención & control , Glucosa/farmacología , Imidazoles/farmacología , Inflamación/patología , Riñón/patología , Glomérulos Renales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Nefritis Intersticial/patología , Piridinas/farmacología , Factores de Transcripción de la Familia Snail/efectos de los fármacos , Factores de Transcripción de la Familia Snail/genética
19.
Eur Rev Med Pharmacol Sci ; 23(8): 3151-3158, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31081065

RESUMEN

OBJECTIVE: This study aimed to investigate the inhibitory effect of Odanacatib on orthodontic recurrence in rats. MATERIALS AND METHODS: Forty rats were selected to establish a planting anchorage molar movement model, and 50 g of force was used for the mesial movement of the right maxillary first molar. Forty rats were randomly divided into the observation group (n=20) and control group (n=20). Odanacatib (60 µl, 1.25 µM) was locally injected into the mucoperiosteum around the right maxillary first molar of rats in the experimental group, and an equal amount of normal saline was injected into rats in the control group. A Vernier caliper was used for measuring the recurrence movement distance and recurrence rate of rats, Micro-CT for scanning the bone mineral density (BMD) and bone volume fraction (BVF) of the alveolar bone, TRAP special staining for observing changes in osteoclasts and quantitative Real Time-Polymerase Chain Reaction (qRT-PCR) for detecting the mRNA expressions of cathepsin K (CatK) and insulin-like growth factor 1 (IGF-1) in periodontal tissues. RESULTS: After 3 weeks of modeling, the movement distance of the first molar of rats in the two groups was 1.16±0.19 mm. The molar movement distance and recurrence rate of rats were significantly higher in the control group than those in the observation group (p<0.05). The BMD and BVF of the alveolar bone of rats were markedly lower in the control group than those in the observation group (p<0.05). There was no statistically significant difference in the number of osteoclasts between the observation group (26.15±3.92) and the control group (27.01±2.74) (t=0.882, p=0.383). The CatK mRNA expression of rats was remarkably lower in the observation group than that in the control group (p<0.05). The IGF-1 mRNA expression of rats was significantly higher in the observation group than that in the control group (p<0.05). CONCLUSIONS: By promoting the IGF-1 mRNA expression and increasing the BMD and BVF of the alveolar bone, Odanacatib inhibits orthodontic recurrence and has no effect on osteoclast activity.


Asunto(s)
Compuestos de Bifenilo/uso terapéutico , Catepsina K/antagonistas & inhibidores , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Osteoclastos/efectos de los fármacos , Migración del Diente/tratamiento farmacológico , Técnicas de Movimiento Dental/métodos , Proceso Alveolar/diagnóstico por imagen , Proceso Alveolar/efectos de los fármacos , Animales , Compuestos de Bifenilo/administración & dosificación , Catepsina K/genética , Modelos Animales de Enfermedad , Factor I del Crecimiento Similar a la Insulina/genética , Movimiento Mesial de los Dientes/tratamiento farmacológico , Osteoclastos/metabolismo , Periodoncio/efectos de los fármacos , Periodoncio/metabolismo , ARN Mensajero/genética , Ratas Endogámicas , Recurrencia , Migración del Diente/metabolismo , Microtomografía por Rayos X
20.
Cancer Lett ; 458: 29-38, 2019 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-31129148

RESUMEN

This article has been retracted at the request of the Editor-in-Chief due to concerns regarding the legitimacy of images and data presented in the paper. Though a corrigendum (Can. Lett. Vol. 469, 2020, pages 524-535) was previously published to address some of these concerns, this corrigendum has also been found to contain errors and therefore cannot stand. Specific concerns are listed below. The Editor and Publisher received a letter from the University of Portsmouth alerting us to an investigation into alleged research misconduct. The University concluded their investigation with external experts and determined that misconduct did take place in relation to the research involved in this paper. Upon our separate investigation, it has been determined that the paper headline relies on showing that there was considerable reduction of IGF1R, IL6R and EGFR post treatment in all cell lines. During review, it was determined that this cannot be concluded from the presented data. For example, in SEBTA-003 the EGFR levels go up and there is no difference in IGFR1. It is apparent from Fig 4d that in the SEBTA-003 cell line the EGFR level does not go down, which is stated in the Results section on page 32, it is rather going up. The data for IGFR1 are inconclusive and there are concerns regarding the blot. The general implications would be that the effects of the drug IP1867B does not seem to be the same for all tested cell lines, and this should have been discussed in detail by the authors. Additionally, in subsequent experiments (Fig. 4g and h) the SEBTA-003 cell line (no reduction of EGFR, rather increased expression) and the other 3 cell lines (reduction of EGFR) show similar responses. This is particularly evident in Fig. 4g: Two cell lines are compared, SEBTA-003 (increased EGFR expression) and UP-029 (decreased EGFR expression), both behave similarly after exposure to drugs. The corrigendum (https://doi.org/10.1016/j.canlet.2019.10.002) issue is with respect to the Supplemental Figure 6i EGFR, particularly panel IP1867B. The Corrigendum states that the left part is a cut out of the very right part. If so, the bands for IP1867B should show the same staining pattern - but they do not. Also, in the Corrigendum, there are incorrect mentions between day 14 in the Figure and day 19 in the Figure legend. All authors were informed of the retraction in advance. Drs. Pritchard and Duckworth agreed to the retraction. The corresponding author, Dr Hill, did not agree to the retraction. No response had been received from Drs. Mihajluk, Simms, Reay, Madureira, Howarth, Murray, Nasser and Pilkinton at the time of the retraction being published.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Aspirina/administración & dosificación , Neoplasias Encefálicas/tratamiento farmacológico , Glioma/tratamiento farmacológico , Receptor IGF Tipo 1/antagonistas & inhibidores , Animales , Aspirina/farmacología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Hipoxia de la Célula/fisiología , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/biosíntesis , Receptores ErbB/genética , Excipientes/administración & dosificación , Femenino , Glioma/genética , Glioma/metabolismo , Glioma/patología , Humanos , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Factor I del Crecimiento Similar a la Insulina/biosíntesis , Factor I del Crecimiento Similar a la Insulina/genética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Clasificación del Tumor , Receptor IGF Tipo 1/biosíntesis , Receptor IGF Tipo 1/genética , Temozolomida/administración & dosificación , Temozolomida/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA