Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Mol Metab ; 80: 101880, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38218536

RESUMEN

OBJECTIVE: Glucagon-like peptide 1 (GLP-1) receptor agonists reduce food intake, producing remarkable weight loss in overweight and obese individuals. While much of this weight loss is fat mass, there is also a loss of lean mass, similar to other approaches that induce calorie deficit. Targeting signaling pathways that regulate skeletal muscle hypertrophy is a promising avenue to preserve lean mass and modulate body composition. Myostatin and Activin A are TGFß-like ligands that signal via the activin type II receptors (ActRII) to antagonize muscle growth. Pre-clinical and clinical studies demonstrate that ActRII blockade induces skeletal muscle hypertrophy and reduces fat mass. In this manuscript, we test the hypothesis that combined ActRII blockade and GLP-1 receptor agonism will preserve muscle mass, leading to improvements in skeletomuscular and metabolic function and enhanced fat loss. METHODS: In this study, we explore the therapeutic potential of bimagrumab, a monoclonal antibody against ActRII, to modify body composition alone and during weight loss induced by GLP-1 receptor agonist semaglutide in diet-induced obese mice. Mechanistically, we define the specific role of the anabolic kinase Akt in mediating the hypertrophic muscle effects of ActRII inhibition in vivo. RESULTS: Treatment of obese mice with bimagrumab induced a ∼10 % increase in lean mass while simultaneously decreasing fat mass. Daily treatment of obese mice with semaglutide potently decreased body weight; this included a significant decrease in both muscle and fat mass. Combination treatment with bimagrumab and semaglutide led to superior fat mass loss while simultaneously preserving lean mass despite reduced food intake. Treatment with both drugs was associated with improved metabolic outcomes, and increased lean mass was associated with improved exercise performance. Deletion of both Akt isoforms in skeletal muscle modestly reduced, but did not prevent, muscle hypertrophy driven by ActRII inhibition. CONCLUSIONS: Collectively, these data demonstrate that blockade of ActRII signaling improves body composition and metabolic parameters during calorie deficit driven by GLP-1 receptor agonism and demonstrate the existence of Akt-independent pathways supporting muscle hypertrophy in the absence of ActRII signaling.


Asunto(s)
Receptores de Activinas Tipo II , Anticuerpos Monoclonales Humanizados , Receptor del Péptido 1 Similar al Glucagón , Obesidad , Proteínas Proto-Oncogénicas c-akt , Pérdida de Peso , Animales , Ratones , Receptores de Activinas Tipo II/antagonistas & inhibidores , Receptores de Activinas Tipo II/metabolismo , Activinas/metabolismo , Anticuerpos Bloqueadores/metabolismo , Anticuerpos Bloqueadores/farmacología , Anticuerpos Bloqueadores/uso terapéutico , Receptor del Péptido 1 Similar al Glucagón/agonistas , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Hipertrofia/metabolismo , Ratones Obesos , Músculo Esquelético/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Anticuerpos Monoclonales Humanizados/administración & dosificación , Obesidad/tratamiento farmacológico
2.
Cancer Med ; 10(1): 286-296, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33179858

RESUMEN

Osteosarcoma is a cancer of pathological bone remodeling with high mortality and severe comorbidity. New therapies are urgently needed. Activin A, a member of the transforming growth factor ß (TGFß) superfamily, has been suggested to stimulate proliferation and invasion of osteosarcoma cells in vitro, thus representing a potential therapeutic target. In this study, inhibition of the activin receptor signaling pathway was explored as a therapy for osteosarcoma. In a murine intratibial osteosarcoma xenograft model, two types of inhibitors were tested: (a) a soluble activin type IIA decoy receptor (ActRIIA-mFc), or (b) a modified variant of follistatin (FSTΔHBS -hFc), either alone or in combination with a bisphosphonate. Both inhibitors reduced primary tumor development by nearly 50% compared to vehicle treatment. When ActRIIA-mFc was combined with bisphosphonate, the effect on tumor size became even more pronounced (78% reduction vs. vehicle). Moreover, FSTΔHBS -hFc increased body weight in the face of tumor progression (14% increase vs. vehicle), and ActRIIA-mFc reduced the number of lung metastases when combined with bisphosphonate. The present study demonstrates a novel approach to treating osteosarcoma and encourages further investigation of inhibition of the activin receptor signaling pathway as an intervention against the disease.


Asunto(s)
Receptores de Activinas Tipo II/antagonistas & inhibidores , Antineoplásicos/farmacología , Neoplasias Óseas/tratamiento farmacológico , Folistatina/farmacología , Fragmentos Fc de Inmunoglobulinas/farmacología , Inmunoglobulina G/farmacología , Osteosarcoma/tratamiento farmacológico , Tibia/efectos de los fármacos , Receptores de Activinas Tipo II/metabolismo , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Conservadores de la Densidad Ósea/farmacología , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Remodelación Ósea/efectos de los fármacos , Línea Celular Tumoral , Difosfonatos/farmacología , Humanos , Ratones SCID , Necrosis , Osteosarcoma/genética , Osteosarcoma/metabolismo , Osteosarcoma/patología , Proteínas Recombinantes de Fusión/farmacología , Transducción de Señal , Tibia/metabolismo , Tibia/patología , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
3.
FASEB J ; 34(8): 9911-9924, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32427381

RESUMEN

Signaling through activin receptors regulates skeletal muscle mass and activin receptor 2B (ACVR2B) ligands are also suggested to participate in myocardial infarction (MI) pathology in the heart. In this study, we determined the effect of systemic blockade of ACVR2B ligands on cardiac function in experimental MI, and defined its efficacy to revert muscle wasting in ischemic heart failure (HF). Mice were treated with soluble ACVR2B decoy receptor (ACVR2B-Fc) to study its effect on post-MI cardiac remodeling and on later HF. Cardiac function was determined with echocardiography, and myocardium analyzed with histological and biochemical methods for hypertrophy and fibrosis. Pharmacological blockade of ACVR2B ligands did not rescue the heart from ischemic injury or alleviate post-MI remodeling and ischemic HF. Collectively, ACVR2B-Fc did not affect cardiomyocyte hypertrophy, fibrosis, angiogenesis, nor factors associated with cardiac regeneration except modification of certain genes involved in metabolism or cell growth/survival. ACVR2B-Fc, however, was able to reduce skeletal muscle wasting in chronic ischemic HF, accompanied by reduced LC3II as a marker of autophagy and increased mTOR signaling and Cited4 expression as markers of physiological hypertrophy in quadriceps muscle. Our results ascertain pharmacological blockade of ACVR2B ligands as a possible therapy for skeletal muscle wasting in ischemic HF. Pharmacological blockade of ACVR2B ligands preserved myofiber size in ischemic HF, but did not compromise cardiac function nor exacerbate cardiac remodeling after ischemic injury.


Asunto(s)
Receptores de Activinas Tipo II/antagonistas & inhibidores , Modelos Animales de Enfermedad , Corazón/fisiología , Atrofia Muscular/prevención & control , Isquemia Miocárdica/complicaciones , Factores de Transcripción/metabolismo , Remodelación Ventricular/fisiología , Receptores de Activinas Tipo II/metabolismo , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Atrofia Muscular/etiología , Atrofia Muscular/metabolismo , Atrofia Muscular/patología , Transducción de Señal , Factores de Transcripción/genética
4.
CPT Pharmacometrics Syst Pharmacol ; 9(6): 342-352, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32419339

RESUMEN

Quantitative understanding about the dynamics of drug-target interactions in biological systems is essential, especially in rare disease programs with small patient populations. Follistatin, by antagonism of myostatin and activin, which are negative regulators of skeletal muscle and inflammatory response, is a promising therapeutic target for Duchenne Muscular Dystrophy. In this study, we constructed a quantitative systems pharmacology model for FS-EEE-Fc, a follistatin recombinant protein to investigate its efficacy from dual target binding, and, subsequently, to project its human efficacious dose. Based on model simulations, with an assumed efficacy threshold of 7-10% muscle volume increase, 3-5 mg/kg weekly dosing of FS-EEE-Fc is predicted to achieve meaningful clinical outcome. In conclusion, the study demonstrated an application of mechanism driven approach at early stage of a rare disease drug development to support lead compound optimization, enable human dose, pharmacokinetics, and efficacy predictions.


Asunto(s)
Folistatina/farmacocinética , Modelos Biológicos , Músculo Esquelético/efectos de los fármacos , Distrofia Muscular de Duchenne/tratamiento farmacológico , Fármacos Neuromusculares/farmacocinética , Biología de Sistemas , Receptores de Activinas Tipo II/antagonistas & inhibidores , Receptores de Activinas Tipo II/metabolismo , Animales , Cálculo de Dosificación de Drogas , Folistatina/administración & dosificación , Humanos , Ligandos , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/fisiopatología , Miostatina/antagonistas & inhibidores , Miostatina/metabolismo , Fármacos Neuromusculares/administración & dosificación , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/farmacocinética , Transducción de Señal
5.
Chem Pharm Bull (Tokyo) ; 68(3): 194-200, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32115526

RESUMEN

Fibrodysplasia ossificans progressiva (FOP) and diffuse intrinsic pontine glioma (DIPG) are diseases that typically manifest in childhood and are associated with severely reduced life expectancy. However, there are currently no effective therapies for these diseases, which remain incurable. Activin receptor-like kinase-2 (ALK2), encoded by the ACVR1 gene, is a bone morphogenetic protein (BMP) type-I receptor subtype that plays an important physiological role in the development of bones, muscles, brain, and other organs. Constitutively active mutants of ALK2 have been identified as causative of FOP and involved in the tumorigenesis of DIPG owing to abnormal activation of BMP signaling, and therefore have emerged as promising treatment targets. Here, we describe these two diseases, along with the link to ALK2 signal transduction, and highlight potential ALK2 inhibitors that are under development to offer new hope for patients with FOP and DIPG.


Asunto(s)
Receptores de Activinas Tipo II/antagonistas & inhibidores , Glioma Pontino Intrínseco Difuso/tratamiento farmacológico , Miositis Osificante/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Receptores de Activinas Tipo II/metabolismo , Glioma Pontino Intrínseco Difuso/metabolismo , Humanos , Miositis Osificante/metabolismo , Inhibidores de Proteínas Quinasas/química , Transducción de Señal/efectos de los fármacos
6.
Cancer Chemother Pharmacol ; 84(4): 909-917, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31444620

RESUMEN

PURPOSE: This study aimed to evaluate the maximum tolerated dose (MTD) and recommended phase II dose (RPTD), as well as the safety and tolerability of PF-03446962, a monoclonal antibody targeting activin receptor like kinase 1 (ALK-1), in combination with regorafenib in patients with refractory metastatic colorectal cancer. METHODS: The first stage of this study was a standard "3 + 3" open-label dose-escalation scheme. Cohorts of 3-6 subjects were started with 120 mg of regorafenib given PO daily for 3 weeks of a 4 week cycle, plus 4.5 mg/kg of PF-03446962 given IV every 2 weeks. Doses of both drugs were adjusted according to dose-limiting toxicities (DLT). Plasma was collected for multiplexed ELISA analysis of factors related to tumor growth and angiogenesis. RESULTS: Seventeen subjects were enrolled, of whom 11 were deemed evaluable. Seven subjects were enrolled at dose level 1, and four were enrolled at level - 1. Overall, three DLTs were observed during the dose-escalation phase: two in level 1 and one in level - 1. A planned dose-expansion cohort was not started due to early termination of the clinical trial. Common adverse events were infusion-related reaction, fatigue, palmar-plantar erythrodysesthesia syndrome, abdominal pain, dehydration, nausea, back pain, anorexia, and diarrhea. One subject achieved stable disease for 5.5 months, but discontinued treatment due to adverse events. CONCLUSIONS: The regimen of regorafenib and PF-03446962 was associated with unacceptable toxicity and did not demonstrate notable clinical activity in patients with refractory metastatic colorectal cancer.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Neoplasias Colorrectales , Monitoreo de Drogas/métodos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Compuestos de Fenilurea , Piridinas , Receptores de Activinas Tipo II/antagonistas & inhibidores , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Neoplasias Colorrectales/sangre , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Relación Dosis-Respuesta a Droga , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/diagnóstico , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/etiología , Terminación Anticipada de los Ensayos Clínicos , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/efectos adversos , Inhibidores Enzimáticos/farmacocinética , Femenino , Humanos , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Estadificación de Neoplasias , Compuestos de Fenilurea/administración & dosificación , Compuestos de Fenilurea/efectos adversos , Compuestos de Fenilurea/farmacocinética , Piridinas/administración & dosificación , Piridinas/efectos adversos , Piridinas/farmacocinética
7.
Am J Physiol Endocrinol Metab ; 316(5): E852-E865, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30860875

RESUMEN

Cancer-associated cachexia reduces survival, which has been attenuated by blocking the activin receptor type 2B (ACVR2B) ligands in mice. The purpose of this study was to unravel the underlying physiology and novel cachexia biomarkers by use of the colon-26 (C26) carcinoma model of cancer cachexia. Male BALB/c mice were subcutaneously inoculated with C26 cancer cells or vehicle control. Tumor-bearing mice were treated with vehicle (C26+PBS) or soluble ACVR2B either before (C26+sACVR/b) or before and after (C26+sACVR/c) tumor formation. Skeletal muscle and serum metabolomics analysis was conducted by gas chromatography-mass spectrometry. Cancer altered various biologically functional groups representing 1) amino acids, 2) energy sources, and 3) nucleotide-related intermediates. Muscle metabolomics revealed increased content of free phenylalanine in cancer that strongly correlated with the loss of body mass within the last 2 days of the experiment. This correlation was also detected in serum. Decreased ribosomal RNA content and phosphorylation of a marker of pyrimidine synthesis revealed changes in nucleotide metabolism in cancer. Overall, the effect of the experimental C26 cancer predominated over blocking ACVR2B ligands in both muscle and serum. However, the level of methyl phosphate, which was decreased in muscle in cancer, was restored by sACVR2B-Fc treatment. In conclusion, experimental cancer affected muscle and blood metabolomes mostly independently of blocking ACVR2B ligands. Of the affected metabolites, we have identified free phenylalanine as a promising biomarker of muscle atrophy or cachexia. Finally, the decreased capacity for pyrimidine nucleotide and protein synthesis in tumor-bearing mice opens up new avenues in cachexia research.


Asunto(s)
Receptores de Activinas Tipo II/antagonistas & inhibidores , Caquexia/metabolismo , Neoplasias del Colon/metabolismo , Metaboloma/fisiología , Músculo Esquelético/metabolismo , Aminoácidos/metabolismo , Animales , Caquexia/etiología , Línea Celular Tumoral , Neoplasias del Colon/complicaciones , Fragmentos Fc de Inmunoglobulinas/farmacología , Masculino , Redes y Vías Metabólicas , Metaboloma/efectos de los fármacos , Ratones , Músculo Esquelético/efectos de los fármacos , Organofosfatos/metabolismo , Fenilalanina/metabolismo , Biosíntesis de Proteínas/efectos de los fármacos , Biosíntesis de Proteínas/fisiología , Nucleótidos de Pirimidina/metabolismo , Proteínas Recombinantes
8.
Kidney Int ; 95(3): 506-517, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30598193

RESUMEN

The past two decades have witnessed tremendous progress in our understanding of the mechanisms underlying wasting and cachexia in chronic kidney disease (CKD) and in other chronic illnesses, such as cancer and heart failure. In all these conditions wasting is an effect of the activation of protein degradation in muscle, a response that increases the risk of morbidity and mortality. Major recent advances in our knowledge on how CKD and inflammation affect cellular signaling include the identification of the myostatin (MSTN)/activin system, and its related transcriptional program that promotes protein degradation. In addition, the identification of the role of MSTN/activin in the vascular wall shows premise that its inhibition can better control or prevent some effects of CKD on vessels, such as accelerated atherosclerosis and vascular calcifications. In this review, we summarize the expanding role of MSTN activation in promoting muscle atrophy and the recent clinical studies that investigated the efficacy of MSTN/activin pathway antagonism in sarcopenic patients. Moreover, we also review the utility of MSTN inhibition in the experimental models of CKD and its potential advantages in CKD patients. Lessons learned from clinical studies on MSTN antagonism in sarcopenic patients tell us that the anabolic intervention is likely better if we use a block of the two ActRII receptors. At the same time, however, it is becoming clear that MSTN-targeted therapies should not be seen as a substitute for physical activity and nutritional supplementation which are mandatory to successfully manage patients with wasting.


Asunto(s)
Activinas/metabolismo , Anabolizantes/farmacología , Miostatina/metabolismo , Insuficiencia Renal Crónica/complicaciones , Sarcopenia/patología , Receptores de Activinas Tipo II/antagonistas & inhibidores , Receptores de Activinas Tipo II/metabolismo , Anabolizantes/uso terapéutico , Animales , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Terapia por Ejercicio , Conducta Alimentaria/fisiología , Humanos , Músculo Esquelético/patología , Miostatina/antagonistas & inhibidores , Proteolisis/efectos de los fármacos , Insuficiencia Renal Crónica/rehabilitación , Sarcopenia/etiología , Sarcopenia/prevención & control , Transducción de Señal/efectos de los fármacos , Resultado del Tratamiento
9.
Muscle Nerve ; 58(6): 834-842, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30025155

RESUMEN

INTRODUCTION: Postnatal activin/myostatin type IIB receptor (ActRIIB) blockade increases skeletal muscle mass and strength but also increases muscle fatigability and impairs oxidative metabolism. The objective of this study was to determine in vivo whether this increased fatigability is due to energy supply limitation. METHODS: The impact of 8-week ActRIIB blockade with soluble receptor (sActRIIB-Fc) on muscle function and adenosine triphosphate (ATP) fluxes was investigated noninvasively by using multimodal magnetic resonance and indirect calorimetry measurements in wild-type mice. RESULTS: Activin/myostatin type IIB receptor blockade reduced (-41%) the muscle apparent mitochondrial capacity and increased (+11%) the basal body energy expenditure. During a fatiguing exercise, ActRIIB blockade decreased both oxidative ATP production rate (-32%) and fatigue resistance (-36%), but these changes affected neither the total ATP production rate nor the contractile ATP cost. DISCUSSION: These findings demonstrate that the increased fatigability after ActRIIB blockade is not due to limitation in energy supply and/or disturbance in contractile ATP cost. Muscle Nerve 58:834-842, 2018.


Asunto(s)
Receptores de Activinas Tipo II/metabolismo , Adenosina Trifosfato/metabolismo , Músculo Esquelético/fisiología , Receptores de Activinas Tipo II/antagonistas & inhibidores , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Calorimetría , Creatina/metabolismo , Metabolismo Energético , Femenino , Espectroscopía de Resonancia Magnética , Ratones , Ratones Endogámicos C57BL , Contracción Muscular/efectos de los fármacos , Contracción Muscular/fisiología , Músculo Esquelético/efectos de los fármacos , Proteínas Recombinantes de Fusión/farmacología , Estadísticas no Paramétricas
10.
Kidney Int ; 93(1): 147-158, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28843411

RESUMEN

We examined activin receptor type IIA (ActRIIA) activation in chronic kidney disease (CKD) by signal analysis and inhibition in mice with Alport syndrome using the ActRIIA ligand trap RAP-011 initiated in 75-day-old Alport mice. At 200 days of age, there was severe CKD and associated Mineral and Bone Disorder (CKD-MBD), consisting of osteodystrophy, vascular calcification, cardiac hypertrophy, hyperphosphatemia, hyperparathyroidism, elevated FGF23, and reduced klotho. The CKD-induced bone resorption and osteoblast dysfunction was reversed, and bone formation was increased by RAP-011. ActRIIA inhibition prevented the formation of calcium apatite deposits in the aortic adventitia and tunica media and significantly decreased the mean aortic calcium concentration from 0.59 in untreated to 0.36 mg/g in treated Alport mice. Aortic ActRIIA stimulation in untreated mice increased p-Smad2 levels and the transcription of sm22α and αSMA. ActRIIA inhibition reversed aortic expression of the osteoblast transition markers Runx2 and osterix. Heart weight was significantly increased by 26% in untreated mice but remained normal during RAP-011 treatment. In 150-day-old mice, GFR was significantly reduced by 55%, but only by 30% in the RAP-011-treated group. In 200-day-old mice, the mean BUN was 100 mg/dl in untreated mice compared to 60 mg/dl in the treated group. In the kidneys of 200-day-old mice, ActRIIA and p-Smad2 were induced and MCP-1, fibronectin, and interstitial fibrosis were stimulated; all were attenuated by RAP-011 treatment. Hence, the activation of ActRIIA signaling during early CKD contributes to the CKD-MBD components of osteodystrophy and cardiovascular disease and to renal fibrosis. Thus, the inhibition of ActRIIA signaling is efficacious in improving and delaying CKD-MBD in this model of Alport syndrome.


Asunto(s)
Receptores de Activinas Tipo II/metabolismo , Resorción Ósea/metabolismo , Cardiomegalia/metabolismo , Trastorno Mineral y Óseo Asociado a la Enfermedad Renal Crónica/metabolismo , Nefritis Hereditaria/metabolismo , Insuficiencia Renal Crónica/metabolismo , Calcificación Vascular/metabolismo , Actinas/metabolismo , Receptores de Activinas Tipo II/antagonistas & inhibidores , Receptores de Activinas Tipo II/genética , Animales , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patología , Vasos Sanguíneos/fisiopatología , Remodelación Ósea , Resorción Ósea/genética , Resorción Ósea/fisiopatología , Resorción Ósea/prevención & control , Huesos/metabolismo , Huesos/patología , Huesos/fisiopatología , Cardiomegalia/genética , Cardiomegalia/fisiopatología , Cardiomegalia/prevención & control , Trastorno Mineral y Óseo Asociado a la Enfermedad Renal Crónica/genética , Trastorno Mineral y Óseo Asociado a la Enfermedad Renal Crónica/fisiopatología , Trastorno Mineral y Óseo Asociado a la Enfermedad Renal Crónica/prevención & control , Colágeno Tipo IV/deficiencia , Colágeno Tipo IV/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Modelos Animales de Enfermedad , Factor-23 de Crecimiento de Fibroblastos , Fibrosis , Tasa de Filtración Glomerular , Riñón/metabolismo , Riñón/patología , Riñón/fisiopatología , Ratones Noqueados , Proteínas de Microfilamentos/metabolismo , Proteínas Musculares/metabolismo , Miocardio/metabolismo , Miocardio/patología , Nefritis Hereditaria/tratamiento farmacológico , Nefritis Hereditaria/genética , Nefritis Hereditaria/fisiopatología , Fosforilación , Proteínas Recombinantes de Fusión/farmacología , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/fisiopatología , Insuficiencia Renal Crónica/prevención & control , Transducción de Señal , Proteína Smad2/metabolismo , Factor de Transcripción Sp7/metabolismo , Calcificación Vascular/genética , Calcificación Vascular/fisiopatología , Calcificación Vascular/prevención & control , Remodelación Vascular
11.
J Cachexia Sarcopenia Muscle ; 9(2): 417-432, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29230965

RESUMEN

BACKGROUND: Toxicity of chemotherapy on skeletal muscles and the heart may significantly contribute to cancer cachexia, mortality, and decreased quality of life. Doxorubicin (DOX) is an effective cytostatic agent, which unfortunately has toxic effects on many healthy tissues. Blocking of activin receptor type IIB (ACVR2B) ligands is an often used strategy to prevent skeletal muscle loss, but its effects on the heart are relatively unknown. METHODS: The effects of DOX treatment with or without pre-treatment with soluble ACVR2B-Fc (sACVR2B-Fc) were investigated. The mice were randomly assigned into one of the three groups: (1) vehicle (PBS)-treated controls, (2) DOX-treated mice (DOX), and (3) DOX-treated mice administered with sACVR2B-Fc during the experiment (DOX + sACVR2B-Fc). DOX was administered with a cumulative dose of 24 mg/kg during 2 weeks to investigate cachexia outcome in the heart and skeletal muscle. To understand similarities and differences between skeletal and cardiac muscles in their responses to chemotherapy, the tissues were collected 20 h after a single DOX (15 mg/kg) injection and analysed with genome-wide transcriptomics and mRNA and protein analyses. The combination group was pre-treated with sACVR2B-Fc 48 h before DOX administration. Major findings were also studied in mice receiving only sACVR2B-Fc. RESULTS: The DOX treatment induced similar (~10%) wasting in skeletal muscle and the heart. However, transcriptional changes in response to DOX were much greater in skeletal muscle. Pathway analysis and unbiased transcription factor analysis showed that p53-p21-REDD1 is the main common pathway activated by DOX in both skeletal and cardiac muscles. These changes were attenuated by blocking ACVR2B ligands especially in skeletal muscle. Tceal7 (3-fold to 5-fold increase), transferrin receptor (1.5-fold increase), and Ccl21 (0.6-fold to 0.9-fold decrease) were identified as novel genes responsive to blocking ACVR2B ligands. Overall, at the transcriptome level, ACVR2B ligand blocking had only minor influence in the heart while it had marked effects in skeletal muscle. The same was also true for the effects on tissue wasting. This may be explained in part by about 18-fold higher gene expression of myostatin in skeletal muscle compared with the heart. CONCLUSIONS: Cardiac and skeletal muscles display similar atrophy after DOX treatment, but the mechanisms for this may differ between the tissues. The present results suggest that p53-p21-REDD1 signalling is the main common DOX-activated pathway in these tissues and that blocking activin receptor ligands attenuates this response, especially in skeletal muscle supporting the overall stronger effects of this treatment in skeletal muscles.


Asunto(s)
Receptores de Activinas Tipo II/antagonistas & inhibidores , Caquexia/prevención & control , Corazón/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Animales , Antibióticos Antineoplásicos/efectos adversos , Caquexia/inducido químicamente , Doxorrubicina/efectos adversos , Humanos , Ligandos , Masculino , Ratones , Calidad de Vida
12.
Proc Natl Acad Sci U S A ; 114(47): 12448-12453, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29109273

RESUMEN

The TGF-ß family ligands myostatin, GDF11, and activins are negative regulators of skeletal muscle mass, which have been reported to primarily signal via the ActRIIB receptor on skeletal muscle and thereby induce muscle wasting described as cachexia. Use of a soluble ActRIIB-Fc "trap," to block myostatin pathway signaling in normal or cachectic mice leads to hypertrophy or prevention of muscle loss, perhaps suggesting that the ActRIIB receptor is primarily responsible for muscle growth regulation. Genetic evidence demonstrates however that both ActRIIB- and ActRIIA-deficient mice display a hypertrophic phenotype. Here, we describe the mode of action of bimagrumab (BYM338), as a human dual-specific anti-ActRIIA/ActRIIB antibody, at the molecular and cellular levels. As shown by X-ray analysis, bimagrumab binds to both ActRIIA and ActRIIB ligand binding domains in a competitive manner at the critical myostatin/activin binding site, hence preventing signal transduction through either ActRII. Myostatin and the activins are capable of binding to both ActRIIA and ActRIIB, with different affinities. However, blockade of either single receptor through the use of specific anti-ActRIIA or anti-ActRIIB antibodies achieves only a partial signaling blockade upon myostatin or activin A stimulation, and this leads to only a small increase in muscle mass. Complete neutralization and maximal anabolic response are achieved only by simultaneous blockade of both receptors. These findings demonstrate the importance of ActRIIA in addition to ActRIIB in mediating myostatin and activin signaling and highlight the need for blocking both receptors to achieve a strong functional benefit.


Asunto(s)
Receptores de Activinas Tipo II/antagonistas & inhibidores , Anticuerpos Bloqueadores/farmacología , Anticuerpos Monoclonales/farmacología , Hipertrofia/inducido químicamente , Músculo Esquelético/efectos de los fármacos , Receptores de Activinas Tipo II/metabolismo , Activinas/metabolismo , Animales , Anticuerpos Bloqueadores/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Proteínas Morfogenéticas Óseas/metabolismo , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Factores de Diferenciación de Crecimiento/metabolismo , Células HEK293 , Humanos , Hipertrofia/patología , Masculino , Ratones , Ratones SCID , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Miostatina/metabolismo , Ratas , Ratas Wistar , Proteínas Recombinantes/metabolismo , Transducción de Señal/efectos de los fármacos , Síndrome Debilitante/tratamiento farmacológico , Síndrome Debilitante/patología
13.
J Biol Chem ; 292(33): 13714-13726, 2017 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-28646109

RESUMEN

Bone morphogenetic proteins 9 and 10 (BMP9/BMP10) are circulating cytokines with important roles in endothelial homeostasis. The aim of this study was to investigate the roles of BMP9 and BMP10 in mediating monocyte-endothelial interactions using an in vitro flow adhesion assay. Herein, we report that whereas BMP9/BMP10 alone had no effect on monocyte recruitment, at higher concentrations both cytokines synergized with tumor necrosis factor-α (TNFα) to increase recruitment to the vascular endothelium. The BMP9/BMP10-mediated increase in monocyte recruitment in the presence of TNFα was associated with up-regulated expression levels of E-selectin, vascular cell adhesion molecule (VCAM-1), and intercellular adhesion molecule 1 (ICAM-1) on endothelial cells. Using siRNAs to type I and II BMP receptors and the signaling intermediaries (Smads), we demonstrated a key role for ALK2 in the BMP9/BMP10-induced surface expression of E-selectin, and both ALK1 and ALK2 in the up-regulation of VCAM-1 and ICAM-1. The type II receptors, BMPR-II and ACTR-IIA were both required for this response, as was Smad1/5. The up-regulation of cell surface adhesion molecules by BMP9/10 in the presence of TNFα was inhibited by LDN193189, which inhibits ALK2 but not ALK1. Furthermore, LDN193189 inhibited monocyte recruitment induced by TNFα and BMP9/10. BMP9/10 increased basal IκBα protein expression, but did not alter p65/RelA levels. Our findings suggest that higher concentrations of BMP9/BMP10 synergize with TNFα to induce the up-regulation of endothelial selectins and adhesion molecules, ultimately resulting in increased monocyte recruitment to the vascular endothelium. This process is mediated mainly via the ALK2 type I receptor, BMPR-II/ACTR-IIA type II receptors, and downstream Smad1/5 signaling.


Asunto(s)
Receptores de Activinas Tipo I/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Endotelio Vascular/metabolismo , Factores de Diferenciación de Crecimiento/metabolismo , Monocitos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Receptores de Activinas Tipo I/antagonistas & inhibidores , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo II/antagonistas & inhibidores , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Aorta , Adhesión Celular/efectos de los fármacos , Células Cultivadas , Selectina E/química , Selectina E/genética , Selectina E/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/inmunología , Factor 2 de Diferenciación de Crecimiento , Humanos , Molécula 1 de Adhesión Intercelular/química , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Cinética , Monocitos/citología , Monocitos/efectos de los fármacos , Monocitos/inmunología , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Pirazoles/farmacología , Pirimidinas/farmacología , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/agonistas , Regulación hacia Arriba/efectos de los fármacos , Molécula 1 de Adhesión Celular Vascular/química , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
14.
Oncotarget ; 8(15): 25115-25130, 2017 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-28445989

RESUMEN

TMPRSS2:ERG (T/E) gene fusions are present in approximately 50% of all prostate cancer (PCa) cases. The expression of fusion mRNAs from distinct T/E variants is associated with clinicopathological parameters, while the underlying molecular processes remain unclear. We characterized the molecular mechanisms and functional implications caused by doxycycline (Dox)-inducible overexpression of the frequent T/E III and VI fusion variants in LNCaP cells. Induction of T/E expression resulted in increased cellular migratory and invasive potential, and reduced proliferation and accumulation in G1 phase. T/E overexpressing cells showed epithelial-to-mesenchymal transition (EMT), as demonstrated by upregulation of TGF-ß and WNT pathway genes, mesenchymal markers, and increased phosphorylation of the p38 MAPK. Augmented secretion of TGF-ß1 and -ß2, and T/E-mediated regulation of ALK1, a member of the TGF-ß receptor family, was detected. ALK1 inhibition in T/E overexpressing cells blocked p38 phosphorylation and reduced the expression of the TGF-ß target genes VIM, MMP1, CDH2, and SNAI2. We found a T/E variant VI-specific induction of miR-503 associated with reduced expression of SMAD7 and CDH1. Overexpression of miR-503 led to increased levels of VIM and MMP1. Our findings indicate that TGF-ß signaling is a major determinant of EMT in T/E overexpressing LNCaP cells. We provide evidence that T/E VI-specific transcriptional modulation by miR-503 accounts for differences in the activation of EMT pathway genes, promoting the aggressive phenotype of tumors expressing T/E variant VI. We suggest that ALK1-mediated TGF-ß signaling is a novel oncogenic mechanism in T/E positive PCa.


Asunto(s)
Transición Epitelial-Mesenquimal/genética , Proteínas de Fusión Oncogénica/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Serina Endopeptidasas/genética , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Receptores de Activinas Tipo II/antagonistas & inhibidores , Receptores de Activinas Tipo II/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Variación Genética , Humanos , Masculino , Modelos Biológicos , Neoplasias de la Próstata/patología , Transducción de Señal/efectos de los fármacos , Proteína smad7/antagonistas & inhibidores , Proteína smad7/metabolismo , Transcripción Genética , Regulador Transcripcional ERG/genética , beta Catenina/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
15.
J Med Chem ; 60(4): 1495-1508, 2017 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-28103025

RESUMEN

We synthesized 1 (San78-130), a reversible version of L-783277, as a selective and potent ALK1 inhibitor. Our study showed that 1 possesses great kinase selectivity against a panel of 342 kinases and more potent activity against ALK1 than L-783277. Among the six ALK isotypes (ALK1-6), ALK1 is most significantly inhibited by compound 1. Compound 1 suppresses the BMP9-induced Smad1/5 pathway by mainly inhibiting ALK1 in C2C12 cells. Our molecular dynamics simulations suggest that H-bonding interaction between the C-4' hydroxyl group of 1 and Arg334 of ALK1 substantially contributes to the ALK1 inhibition. To the best of our knowledge, 1 is the first selective ALK1 inhibitor. Furthermore, compound 1 promoted angiogenesis in both endothelial tube formation and microfluidic chip based 3D angiogenesis assays, suggesting that 1 could be a lead compound for therapeutic angiogenesis agents. Our study may provide an insight into designing selective and potent inhibitors against ALK1.


Asunto(s)
Receptores de Activinas Tipo II/antagonistas & inhibidores , Lactonas/química , Lactonas/farmacología , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Resorcinoles/química , Resorcinoles/farmacología , Receptores de Activinas Tipo II/química , Receptores de Activinas Tipo II/metabolismo , Secuencia de Aminoácidos , Inductores de la Angiogénesis/química , Inductores de la Angiogénesis/farmacología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Alineación de Secuencia , Transducción de Señal/efectos de los fármacos , Proteínas Smad/metabolismo
16.
Clin Cancer Res ; 23(14): 3557-3565, 2017 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-28031424

RESUMEN

Purpose: Activin receptor-like kinase 1 (ALK1) is a novel target in angiogenesis. Concurrent targeting of ALK1 and VEGF signaling results in augmented inhibition of tumor growth in renal cell carcinoma (RCC) xenograft models. Dalantercept is an ALK1-receptor fusion protein that acts as a ligand trap for bone morphogenetic proteins 9 and 10. The DART Study evaluated the safety, tolerability, pharmacokinetics, pharmacodynamics, and antitumor activity of dalantercept plus axitinib in patients with advanced RCC and determined the optimal dose for further testing.Experimental Design: Patients received dalantercept 0.6, 0.9, or 1.2 mg/kg subcutaneously every 3 weeks plus axitinib 5 mg by mouth twice daily until disease progression or intolerance.Results: Twenty-nine patients were enrolled in the dose escalation (n = 15) and expansion (n = 14) cohorts. There were no dose-limiting toxicities or grade 4/5 treatment-related adverse events. In addition to common VEGFR tyrosine kinase inhibitor effects, such as fatigue and diarrhea, commonly seen treatment-related adverse events were peripheral edema, epistaxis, pericardial effusion, and telangiectasia. The objective response rate by RECIST v1.1 was 25% with responses seen at all dose levels. The overall median progression-free survival was 8.3 months.Conclusions: The combination of dalantercept plus axitinib is well tolerated and associated with clinical activity. On the basis of safety and efficacy results, the 0.9 mg/kg dose level was chosen for further study in a randomized phase II trial of dalantercept plus axitinib versus placebo plus axitinib. Clin Cancer Res; 23(14); 3557-65. ©2016 AACR.


Asunto(s)
Receptores de Activinas Tipo II/administración & dosificación , Receptores de Activinas Tipo II/genética , Carcinoma de Células Renales/tratamiento farmacológico , Imidazoles/administración & dosificación , Fragmentos Fc de Inmunoglobulinas/administración & dosificación , Indazoles/administración & dosificación , Proteínas Recombinantes de Fusión/administración & dosificación , Factor A de Crecimiento Endotelial Vascular/genética , Receptores de Activinas Tipo II/efectos adversos , Receptores de Activinas Tipo II/antagonistas & inhibidores , Anciano , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Axitinib , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Supervivencia sin Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Imidazoles/efectos adversos , Fragmentos Fc de Inmunoglobulinas/efectos adversos , Indazoles/efectos adversos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/efectos adversos , Proteínas Recombinantes de Fusión/efectos adversos , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores
17.
J Cell Physiol ; 232(7): 1774-1793, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27859236

RESUMEN

The efficacy of two ActRIIB ligand-trapping agents (RAP-031 and RAP-435) in treating muscular dystrophy was examined by determining their morphological effects on the severely dystrophic triangularis sterni (TS) muscle of the mdx mouse, a model for Duchenne muscular dystrophy. These agents trap all endogenous ligands to the ActRIIB receptor and thereby block myostatin signaling in a highly selective manner. Short-term (1 month) and long-term (3 months) in vivo treatment of 1-month-old mdx mice increased myonuclei and fiber cross section (FCS) density but did not alter individual fiber size. Vehicle-treated mdx mice exhibited age-dependent increases in myonuclei and FCS density, and age-dependent reductions in centronucleation that were each enhanced by treatment with RAP-435. Distributions of FCS area (FCSA) in the mdx TS were 90% identical to those from untreated age-matched nondystrophic mice and were unaltered by the substantial fiber hyperplasia observed with age and RAP-435 treatment. These results were inconsistent with injury-induced fiber regeneration which produces altered FCSA distributions characterized by a distinct class of smaller regenerated fibers. Nondystrophic mice exhibited a constant postnatal density of fiber cross sections and myonuclei, and RAP-435 treatment of nondystrophic mice increased TS mean FCSA but had no effects on myonuclei or FCS density. These results demonstrating a continual postnatal proliferation and fusion of satellite cells and a response to myostatin blockade characteristic of developing prenatal muscle suggest that the lack of dystrophin directly results in unrestrained postnatal satellite cell activation that is not necessarily dependent upon prior fiber degeneration. J. Cell. Physiol. 232: 1774-1793, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Receptores de Activinas Tipo II/antagonistas & inhibidores , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/patología , Receptores de Activinas Tipo II/metabolismo , Animales , Animales Recién Nacidos , Peso Corporal , Núcleo Celular/metabolismo , Hiperplasia , Ligandos , Ratones Endogámicos mdx , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Músculo Esquelético/patología , Distrofia Muscular Animal/metabolismo
18.
PLoS One ; 11(11): e0166803, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27870893

RESUMEN

Spinal muscular atrophy (SMA) is a devastating neurodegenerative disorder that causes progressive muscle atrophy and weakness. Using adeno-associated virus-mediated gene transfer, we evaluated the potential to improve skeletal muscle weakness via systemic, postnatal inhibition of either myostatin or all signaling via the activin receptor type IIB (ActRIIB). After demonstrating elevated p-SMAD3 content and differential content of ActRIIB ligands, 4-week-old male C/C SMA model mice were treated intraperitoneally with 1x1012 genome copies of pseudotype 2/8 virus encoding a soluble form of the ActRIIB extracellular domain (sActRIIB) or protease-resistant myostatin propeptide (dnMstn) driven by a liver specific promoter. At 12 weeks of age, muscle mass and function were improved in treated C/C mice by both treatments, compared to controls. The fast fiber type muscles had a greater response to treatment than did slow muscles, and the greatest therapeutic effects were found with sActRIIB treatment. Myostatin/activin inhibition, however, did not rescue C/C mice from the reduction in motor unit numbers of the tibialis anterior muscle. Collectively, this study indicates that myostatin/activin inhibition represents a potential therapeutic strategy to increase muscle mass and strength, but not neuromuscular junction defects, in less severe forms of SMA.


Asunto(s)
Receptores de Activinas Tipo II/genética , Músculo Esquelético/patología , Atrofia Muscular Espinal/terapia , Miostatina/antagonistas & inhibidores , Péptidos/genética , Receptores de Activinas Tipo II/antagonistas & inhibidores , Receptores de Activinas Tipo II/química , Animales , Dependovirus/genética , Modelos Animales de Enfermedad , Terapia Genética , Vectores Genéticos/administración & dosificación , Humanos , Masculino , Ratones , Contracción Muscular , Músculo Esquelético/fisiopatología , Atrofia Muscular Espinal/patología , Atrofia Muscular Espinal/fisiopatología , Miostatina/genética , Tamaño de los Órganos , Péptidos/farmacología , Fenotipo
19.
Curr Hematol Malig Rep ; 11(6): 416-424, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27595736

RESUMEN

Distinct subtypes of lower risk myelodysplastic syndromes display ring sideroblasts in the bone marrow, i. e., erythroid progenitors characterized by excessive iron deposited in the mitochondria. This morphological feature is frequently associated with somatic mutations in components of the splicing machinery that constitutes the underlying molecular principle of the disease. Conventional treatment regimen with erythropoiesis-stimulating agents often fails to induce sustained erythroid improvement in these patients that harbor defects in late-stage erythroblasts downstream of erythropoietin action. In the present review, we will discuss activin receptor ligand traps as novel therapeutic strategies particularly for sideroblastic subgroups of myelodysplastic syndromes that were recently shown to alleviate anemia by specifically inhibiting aberrant TGF-ß signaling and thereby promoting erythroid differentiation.


Asunto(s)
Receptores de Activinas Tipo II/metabolismo , Síndromes Mielodisplásicos/patología , Receptores de Activinas Tipo II/antagonistas & inhibidores , Activinas/uso terapéutico , Humanos , Fragmentos Fc de Inmunoglobulinas/uso terapéutico , Sobrecarga de Hierro/complicaciones , Sobrecarga de Hierro/genética , Sobrecarga de Hierro/patología , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/genética , Fosfoproteínas/genética , Factores de Empalme de ARN/genética , Proteínas Recombinantes de Fusión/uso terapéutico , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/metabolismo
20.
Biochem Soc Trans ; 44(4): 1142-9, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27528762

RESUMEN

Angiogenesis is a hallmark of cancer and is now a validated therapeutic target in the clinical setting. Despite the initial success, anti-angiogenic compounds impinging on the vascular endothelial growth factor (VEGF) pathway display limited survival benefits in patients and resistance often develops due to activation of alternative pathways. Thus, finding and validating new targets is highly warranted. Activin receptor-like kinase (ALK)1 is a transforming growth factor beta (TGF-ß) type I receptor predominantly expressed in actively proliferating endothelial cells (ECs). ALK1 has been shown to play a pivotal role in regulating angiogenesis by binding to bone morphogenetic protein (BMP)9 and 10. Two main pharmacological inhibitors, an ALK1-Fc fusion protein (Dalantercept/ACE-041) and a fully human antibody against the extracellular domain of ALK1 (PF-03446962) are currently under clinical development. Herein, we briefly recapitulate the role of ALK1 in blood vessel formation and the current status of the preclinical and clinical studies on inhibition of ALK1 signalling as an anti-angiogenic strategy. Future directions in terms of new combination regimens will also be presented.


Asunto(s)
Receptores de Activinas Tipo II/antagonistas & inhibidores , Inhibidores de la Angiogénesis/uso terapéutico , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/prevención & control , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/metabolismo , Neoplasias/irrigación sanguínea , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/uso terapéutico , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...