Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Genes Dev ; 37(7-8): 303-320, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-37024284

RESUMEN

MYC's key role in oncogenesis and tumor progression has long been established for most human cancers. In melanoma, its deregulated activity by amplification of 8q24 chromosome or by upstream signaling coming from activating mutations in the RAS/RAF/MAPK pathway-the most predominantly mutated pathway in this disease-turns MYC into not only a driver but also a facilitator of melanoma progression, with documented effects leading to an aggressive clinical course and resistance to targeted therapy. Here, by making use of Omomyc, the most characterized MYC inhibitor to date that has just successfully completed a phase I clinical trial, we show for the first time that MYC inhibition in melanoma induces remarkable transcriptional modulation, resulting in severely compromised tumor growth and a clear abrogation of metastatic capacity independently of the driver mutation. By reducing MYC's transcriptional footprint in melanoma, Omomyc elicits gene expression profiles remarkably similar to those of patients with good prognosis, underlining the therapeutic potential that such an approach could eventually have in the clinic in this dismal disease.


Asunto(s)
Melanoma , Humanos , Pronóstico , Melanoma/genética , Transducción de Señal , Carcinogénesis , Transformación Celular Neoplásica , Proteínas Proto-Oncogénicas c-myc/metabolismo
2.
Genes Dev ; 27(5): 504-13, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23475959

RESUMEN

The principal reason for failure of targeted cancer therapies is the emergence of resistant clones that regenerate the tumor. Therapeutic efficacy therefore depends on not only how effectively a drug inhibits its target, but also the innate or adaptive functional redundancy of that target and its attendant pathway. In this regard, the Myc transcription factors are intriguing therapeutic targets because they serve the unique and irreplaceable role of coordinating expression of the many diverse genes that, together, are required for somatic cell proliferation. Furthermore, Myc expression is deregulated in most-perhaps all-cancers, underscoring its irreplaceable role in proliferation. We previously showed in a preclinical mouse model of non-small-cell lung cancer that systemic Myc inhibition using the dominant-negative Myc mutant Omomyc exerts a dramatic therapeutic impact, triggering rapid regression of tumors with only mild and fully reversible side effects. Using protracted episodic expression of Omomyc, we now demonstrate that metronomic Myc inhibition not only contains Ras-driven lung tumors indefinitely, but also leads to their progressive eradication. Hence, Myc does indeed serve a unique and nondegenerate role in lung tumor maintenance that cannot be complemented by any adaptive mechanism, even in the most aggressive p53-deficient tumors. These data endorse Myc as a compelling cancer drug target.


Asunto(s)
Neoplasias Pulmonares/metabolismo , Fragmentos de Péptidos/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Animales , Animales Modificados Genéticamente , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Ratones , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/farmacología , Fragmentos de Péptidos/uso terapéutico , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/farmacología , Proteínas Proto-Oncogénicas c-myc/uso terapéutico , Análisis de Supervivencia , Proteína p53 Supresora de Tumor/metabolismo
3.
Nat Med ; 30(3): 762-771, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38321218

RESUMEN

Among the 'most wanted' targets in cancer therapy is the oncogene MYC, which coordinates key transcriptional programs in tumor development and maintenance. It has, however, long been considered undruggable. OMO-103 is a MYC inhibitor consisting of a 91-amino acid miniprotein. Here we present results from a phase 1 study of OMO-103 in advanced solid tumors, established to examine safety and tolerability as primary outcomes and pharmacokinetics, recommended phase 2 dose and preliminary signs of activity as secondary ones. A classical 3 + 3 design was used for dose escalation of weekly intravenous, single-agent OMO-103 administration in 21-day cycles, encompassing six dose levels (DLs). A total of 22 patients were enrolled, with treatment maintained until disease progression. The most common adverse events were grade 1 infusion-related reactions, occurring in ten patients. One dose-limiting toxicity occurred at DL5. Pharmacokinetics showed nonlinearity, with tissue saturation signs at DL5 and a terminal half-life in serum of 40 h. Of the 19 patients evaluable for response, 12 reached the predefined 9-week time point for assessment of drug antitumor activity, eight of those showing stable disease by computed tomography. One patient defined as stable disease by response evaluation criteria in solid tumors showed a 49% reduction in total tumor volume at best response. Transcriptomic analysis supported target engagement in tumor biopsies. In addition, we identified soluble factors that are potential pharmacodynamic and predictive response markers. Based on all these data, the recommended phase 2 dose was determined as DL5 (6.48 mg kg-1).ClinicalTrials.gov identifier: NCT04808362 .


Asunto(s)
Neoplasias , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/patología
4.
Cancers (Basel) ; 15(3)2023 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-36765784

RESUMEN

MYC is an oncoprotein causally involved in the majority of human cancers and a most wanted target for cancer treatment. Omomyc is the best-characterized MYC dominant negative to date. In the last years, it has been developed into a therapeutic miniprotein for solid tumor treatment and recently reached clinical stage. However, since the in vivo stability of therapeutic proteins, especially within the tumor vicinity, can be affected by proteolytic degradation, the perception of Omomyc as a valid therapeutic agent has been often questioned. In this study, we used a mass spectrometry approach to evaluate the stability of Omomyc in tumor biopsies from murine xenografts following its intravenous administration. Our data strongly support that the integrity of the functional domains of Omomyc (DNA binding and dimerization region) remains preserved in the tumor tissue for at least 72 hours following administration and that the protein shows superior pharmacokinetics in the tumor compartment compared with blood serum.

5.
Cancer Res Commun ; 2(2): 110-130, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-36860495

RESUMEN

MYC's role in promoting tumorigenesis is beyond doubt, but its function in the metastatic process is still controversial. Omomyc is a MYC dominant negative that has shown potent antitumor activity in multiple cancer cell lines and mouse models, regardless of their tissue of origin or driver mutations, by impacting on several of the hallmarks of cancer. However, its therapeutic efficacy against metastasis has not been elucidated yet. Here we demonstrate for the first time that MYC inhibition by transgenic Omomyc is efficacious against all breast cancer molecular subtypes, including triple-negative breast cancer, where it displays potent antimetastatic properties both in vitro and in vivo. Importantly, pharmacologic treatment with the recombinantly produced Omomyc miniprotein, recently entering a clinical trial in solid tumors, recapitulates several key features of expression of the Omomyc transgene, confirming its clinical applicability to metastatic breast cancer, including advanced triple-negative breast cancer, a disease in urgent need of better therapeutic options. Significance: While MYC role in metastasis has been long controversial, this manuscript demonstrates that MYC inhibition by either transgenic expression or pharmacologic use of the recombinantly produced Omomyc miniprotein exerts antitumor and antimetastatic activity in breast cancer models in vitro and in vivo, suggesting its clinical applicability.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Animales , Humanos , Ratones , Línea Celular , Unión Proteica , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-myc
6.
Life Sci Alliance ; 4(5)2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33653688

RESUMEN

The huge cadre of genes regulated by Myc has obstructed the identification of critical effectors that are essential for Myc-driven tumorigenesis. Here, we describe how only the lack of the receptor Fzd9, previously identified as a Myc transcriptional target, impairs sustained tumor expansion and ß-cell dedifferentiation in a mouse model of Myc-driven insulinoma, allows pancreatic islets to maintain their physiological structure and affects Myc-related global gene expression. Importantly, Wnt signaling inhibition in Fzd9-competent mice largely recapitulates the suppression of proliferation caused by Fzd9 deficiency upon Myc activation. Together, our results indicate that the Wnt signaling receptor Fzd9 is essential for Myc-induced tumorigenesis in pancreatic islets.


Asunto(s)
Adenoma de Células de los Islotes Pancreáticos/fisiopatología , Carcinogénesis/metabolismo , Receptores Frizzled/metabolismo , Adenoma de Células de los Islotes Pancreáticos/metabolismo , Animales , Movimiento Celular , Proliferación Celular , Femenino , Receptores Frizzled/genética , Receptores Frizzled/fisiología , Genes myc/genética , Genes myc/fisiología , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Vía de Señalización Wnt/genética , Vía de Señalización Wnt/fisiología , beta Catenina/metabolismo
7.
Cells ; 9(4)2020 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-32260326

RESUMEN

First designed and published in 1998 as a laboratory tool to study Myc perturbation, Omomyc has come a long way in the past 22 years. This dominant negative has contributed to our understanding of Myc biology when expressed, first, in normal and cancer cells, and later in genetically-engineered mice, and has shown remarkable anti-cancer properties in a wide range of tumor types. The recently described therapeutic effect of purified Omomyc mini-protein-following the surprising discovery of its cell-penetrating capacity-constitutes a paradigm shift. Now, much more than a proof of concept, the most characterized Myc inhibitor to date is advancing in its drug development pipeline, pushing Myc inhibition into the clinic.


Asunto(s)
Neoplasias/terapia , Fragmentos de Péptidos/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Secuencia de Aminoácidos , Animales , Investigación Biomédica , Epigénesis Genética , Humanos , Neoplasias/genética , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/química , Proteínas Proto-Oncogénicas c-myc/administración & dosificación , Proteínas Proto-Oncogénicas c-myc/química , Activación Transcripcional/genética
8.
Expert Opin Ther Targets ; 24(2): 101-114, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32003251

RESUMEN

Introduction: Lung cancer is the leading cause of cancer-related mortality globally. Despite recent advances with personalized therapies and immunotherapy, the prognosis remains dire and recurrence is frequent. Myc is an oncogene deregulated in human cancers, including lung cancer, where it supports tumorigenic processes and progression. Elevated Myc levels have also been associated with resistance to therapy.Areas covered: This article summarizes the genomic and transcriptomic studies that compile evidence for (i) MYC, MYCN, and MYCL amplification and overexpression in lung cancer patients, and (ii) their prognostic significance. We collected the most recent literature regarding the development of Myc inhibitors where the emphasis is on those inhibitors tested in lung cancer experimental models and their potential for future clinical application.Expert opinion: The targeting of Myc in lung cancer is potentially an unprecedented opportunity for inhibiting a key player in tumor progression and maintenance and therapeutic resistance. Myc inhibitory strategies are on the path to their clinical application but further work is necessary for the assessment of their use in combination with standard treatment approaches. Given the role of Myc in immune suppression, a significant opportunity may exist in the combination of Myc inhibitors with immunotherapies.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-myc/antagonistas & inhibidores , Animales , Desarrollo de Medicamentos , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunoterapia/métodos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Proteína Proto-Oncogénica N-Myc/genética , Proteínas Proto-Oncogénicas c-myc/genética
9.
Sci Transl Med ; 11(484)2019 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-30894502

RESUMEN

Inhibiting MYC has long been considered unfeasible, although its key role in human cancers makes it a desirable target for therapeutic intervention. One reason for its perceived undruggability was the fear of catastrophic side effects in normal tissues. However, we previously designed a dominant-negative form of MYC called Omomyc and used its conditional transgenic expression to inhibit MYC function both in vitro and in vivo. MYC inhibition by Omomyc exerted a potent therapeutic impact in various mouse models of cancer, causing only mild, well-tolerated, and reversible side effects. Nevertheless, Omomyc has been so far considered only a proof of principle. In contrast with that preconceived notion, here, we show that the purified Omomyc mini-protein itself spontaneously penetrates into cancer cells and effectively interferes with MYC transcriptional activity therein. Efficacy of the Omomyc mini-protein in various experimental models of non-small cell lung cancer harboring different oncogenic mutation profiles establishes its therapeutic potential after both direct tissue delivery and systemic administration, providing evidence that the Omomyc mini-protein is an effective MYC inhibitor worthy of clinical development.


Asunto(s)
Péptidos de Penetración Celular/farmacología , Fragmentos de Péptidos/farmacología , Proteínas Proto-Oncogénicas c-myc/antagonistas & inhibidores , Adenocarcinoma del Pulmón/tratamiento farmacológico , Adenocarcinoma del Pulmón/patología , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Péptidos de Penetración Celular/farmacocinética , Péptidos de Penetración Celular/uso terapéutico , ADN/metabolismo , Modelos Animales de Enfermedad , Elementos E-Box/genética , Femenino , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Ratones Endogámicos C57BL , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/farmacocinética , Fragmentos de Péptidos/uso terapéutico , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , Proteínas Proto-Oncogénicas c-myc/administración & dosificación , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Proto-Oncogénicas c-myc/farmacocinética , Proteínas Proto-Oncogénicas c-myc/farmacología , Proteínas Proto-Oncogénicas c-myc/uso terapéutico
10.
Oncotarget ; 9(27): 18734-18746, 2018 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-29721157

RESUMEN

Effectively treating KRAS-driven tumors remains an unsolved challenge. The inhibition of downstream signaling effectors is a way of overcoming the issue of direct targeting of mutant KRAS, which has shown limited efficacy so far. Bromodomain and Extra-Terminal (BET) protein inhibition has displayed anti-tumor activity in a wide range of cancers, including KRAS-driven malignancies. Here, we preclinically evaluate the effect of BET inhibition making use of a new BET inhibitor, BAY 1238097, against Pancreatic Ductal Adenocarcinoma (PDAC) and Non-Small Cell Lung Cancer (NSCLC) models harboring RAS mutations both in vivo and in vitro. Our results demonstrate that BET inhibition displays significant therapeutic impact in genetic mouse models of KRAS-driven PDAC and NSCLC, reducing both tumor area and tumor grade. The same approach also causes a significant reduction in cell number of a panel of RAS-mutated human cancer cell lines (8 PDAC and 6 NSCLC). In this context, we demonstrate that while BET inhibition by BAY 1238097 decreases MYC expression in some cell lines, at least in PDAC cells its anti-tumorigenic effect is independent of MYC regulation. Together, these studies reinforce the use of BET inhibition and prompt the optimization of more efficient and less toxic BET inhibitors for the treatment of KRAS-driven malignancies, which are in urgent therapeutic need.

11.
Cancer Res ; 75(8): 1675-81, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25878147

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is characterized by a dense stromal fibroinflammatory reaction that is a major obstacle to effective therapy. The desmoplastic stroma comprises many inflammatory cells, in particular mast cells as key components of the PDAC microenvironment, and such infiltration correlates with poor patient outcome. Indeed, it has been hypothesized that stromal ablation is critical to improve clinical response in patients with PDAC. Ibrutinib is a clinically approved Bruton's tyrosine kinase inhibitor that inhibits mast cells and tumor progression in a mouse model of ß-cell tumorigenesis. Here, we show that ibrutinib is highly effective at limiting the growth of PDAC in both transgenic mouse and patient-derived xenograft models of the disease. In these various experimental settings, ibrutinib effectively diminished fibrosis, extended survival, and improved the response to clinical standard-of-care therapy. Our results offer a preclinical rationale to immediately evaluate the clinical efficacy of ibrutinib in patients with PDAC.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/uso terapéutico , Neoplasias Pancreáticas/tratamiento farmacológico , Pirazoles/uso terapéutico , Pirimidinas/uso terapéutico , Adenina/análogos & derivados , Adenocarcinoma/patología , Animales , Antineoplásicos/farmacología , Femenino , Fibrosis/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Neoplasias Pancreáticas/patología , Piperidinas , Pirazoles/farmacología , Pirimidinas/farmacología , Células Tumorales Cultivadas , Microambiente Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Nat Commun ; 5: 4632, 2014 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-25130259

RESUMEN

Gliomas are the most common primary tumours affecting the adult central nervous system and respond poorly to standard therapy. Myc is causally implicated in most human tumours and the majority of glioblastomas have elevated Myc levels. Using the Myc dominant negative Omomyc, we previously showed that Myc inhibition is a promising strategy for cancer therapy. Here, we preclinically validate Myc inhibition as a therapeutic strategy in mouse and human glioma, using a mouse model of spontaneous multifocal invasive astrocytoma and its derived neuroprogenitors, human glioblastoma cell lines, and patient-derived tumours both in vitro and in orthotopic xenografts. Across all these experimental models we find that Myc inhibition reduces proliferation, increases apoptosis and remarkably, elicits the formation of multinucleated cells that then arrest or die by mitotic catastrophe, revealing a new role for Myc in the proficient division of glioma cells.


Asunto(s)
Astrocitoma/patología , Neoplasias Encefálicas/patología , Glioblastoma/patología , Glioma/patología , Mitosis/fisiología , Proteínas Proto-Oncogénicas c-myc/antagonistas & inhibidores , Animales , Apoptosis/fisiología , Astrocitoma/fisiopatología , Astrocitoma/terapia , Neoplasias Encefálicas/fisiopatología , Neoplasias Encefálicas/terapia , Línea Celular Tumoral , Proliferación Celular/fisiología , Modelos Animales de Enfermedad , Glioblastoma/fisiopatología , Glioblastoma/terapia , Glioma/fisiopatología , Glioma/terapia , Xenoinjertos , Humanos , Ratones , Ratones Transgénicos , Proteína Tirosina Fosfatasa no Receptora Tipo 1/fisiología , Proteínas Proto-Oncogénicas c-myc/fisiología , Enzimas Activadoras de Ubiquitina/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA