Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Nature ; 629(8013): 927-936, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38588697

RESUMEN

Broad-spectrum RAS inhibition has the potential to benefit roughly a quarter of human patients with cancer whose tumours are driven by RAS mutations1,2. RMC-7977 is a highly selective inhibitor of the active GTP-bound forms of KRAS, HRAS and NRAS, with affinity for both mutant and wild-type variants3. More than 90% of cases of human pancreatic ductal adenocarcinoma (PDAC) are driven by activating mutations in KRAS4. Here we assessed the therapeutic potential of RMC-7977 in a comprehensive range of PDAC models. We observed broad and pronounced anti-tumour activity across models following direct RAS inhibition at exposures that were well-tolerated in vivo. Pharmacological analyses revealed divergent responses to RMC-7977 in tumour versus normal tissues. Treated tumours exhibited waves of apoptosis along with sustained proliferative arrest, whereas normal tissues underwent only transient decreases in proliferation, with no evidence of apoptosis. In the autochthonous KPC mouse model, RMC-7977 treatment resulted in a profound extension of survival followed by on-treatment relapse. Analysis of relapsed tumours identified Myc copy number gain as a prevalent candidate resistance mechanism, which could be overcome by combinatorial TEAD inhibition in vitro. Together, these data establish a strong preclinical rationale for the use of broad-spectrum RAS-GTP inhibition in the setting of PDAC and identify a promising candidate combination therapeutic regimen to overcome monotherapy resistance.


Asunto(s)
Antineoplásicos , Carcinoma Ductal Pancreático , Guanosina Trifosfato , Neoplasias Pancreáticas , Proteínas Proto-Oncogénicas p21(ras) , Animales , Femenino , Humanos , Ratones , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Variaciones en el Número de Copia de ADN , Resistencia a Antineoplásicos/efectos de los fármacos , Genes myc , Guanosina Trifosfato/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/genética , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/antagonistas & inhibidores , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto , Mutación
2.
J Med Chem ; 66(1): 149-169, 2023 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-36533617

RESUMEN

Hyperactivation of mTOR kinase by mutations in the PI3K/mTOR pathway or by crosstalk with other mutant cancer drivers, such as RAS, is a feature of many tumors. Multiple allosteric inhibitors of mTORC1 and orthosteric dual inhibitors of mTORC1 and mTORC2 have been developed as anticancer drugs, but their clinical utility has been limited. To address these limitations, we have developed a novel class of "bi-steric inhibitors" that interact with both the orthosteric and the allosteric binding sites in order to deepen the inhibition of mTORC1 while also preserving selectivity for mTORC1 over mTORC2. In this report, we describe the discovery and preclinical profile of the development candidate RMC-5552 and the in vivo preclinical tool compound RMC-6272. We also present evidence that selective inhibition of mTORC1 in combination with covalent inhibition of KRASG12C shows increased antitumor activity in a preclinical model of KRASG12C mutant NSCLC that exhibits resistance to KRASG12C inhibitor monotherapy.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proliferación Celular , Serina-Treonina Quinasas TOR , Diana Mecanicista del Complejo 2 de la Rapamicina , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Inhibidores de Proteínas Quinasas/química , Línea Celular Tumoral
3.
Science ; 381(6659): 794-799, 2023 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-37590355

RESUMEN

The discovery of small-molecule inhibitors requires suitable binding pockets on protein surfaces. Proteins that lack this feature are considered undruggable and require innovative strategies for therapeutic targeting. KRAS is the most frequently activated oncogene in cancer, and the active state of mutant KRAS is such a recalcitrant target. We designed a natural product-inspired small molecule that remodels the surface of cyclophilin A (CYPA) to create a neomorphic interface with high affinity and selectivity for the active state of KRASG12C (in which glycine-12 is mutated to cysteine). The resulting CYPA:drug:KRASG12C tricomplex inactivated oncogenic signaling and led to tumor regressions in multiple human cancer models. This inhibitory strategy can be used to target additional KRAS mutants and other undruggable cancer drivers. Tricomplex inhibitors that selectively target active KRASG12C or multiple RAS mutants are in clinical trials now (NCT05462717 and NCT05379985).


Asunto(s)
Productos Biológicos , Ciclofilina A , Inmunofilinas , Chaperonas Moleculares , Neoplasias , Proteínas Proto-Oncogénicas p21(ras) , Humanos , Productos Biológicos/química , Productos Biológicos/farmacología , Productos Biológicos/uso terapéutico , Cisteína/química , Cisteína/genética , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/antagonistas & inhibidores , Proteínas Proto-Oncogénicas p21(ras)/química , Proteínas Proto-Oncogénicas p21(ras)/genética , Transducción de Señal , Ciclofilina A/química , Ciclofilina A/metabolismo , Inmunofilinas/química , Inmunofilinas/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/genética
4.
bioRxiv ; 2023 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-38105998

RESUMEN

Broad-spectrum RAS inhibition holds the potential to benefit roughly a quarter of human cancer patients whose tumors are driven by RAS mutations. However, the impact of inhibiting RAS functions in normal tissues is not known. RMC-7977 is a highly selective inhibitor of the active (GTP-bound) forms of KRAS, HRAS, and NRAS, with affinity for both mutant and wild type (WT) variants. As >90% of human pancreatic ductal adenocarcinoma (PDAC) cases are driven by activating mutations in KRAS, we assessed the therapeutic potential of RMC-7977 in a comprehensive range of PDAC models, including human and murine cell lines, human patient-derived organoids, human PDAC explants, subcutaneous and orthotopic cell-line or patient derived xenografts, syngeneic allografts, and genetically engineered mouse models. We observed broad and pronounced anti-tumor activity across these models following direct RAS inhibition at doses and concentrations that were well-tolerated in vivo. Pharmacological analyses revealed divergent responses to RMC-7977 in tumor versus normal tissues. Treated tumors exhibited waves of apoptosis along with sustained proliferative arrest whereas normal tissues underwent only transient decreases in proliferation, with no evidence of apoptosis. Together, these data establish a strong preclinical rationale for the use of broad-spectrum RAS inhibition in the setting of PDAC.

5.
J Clin Invest ; 129(10): 4492-4505, 2019 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-31361600

RESUMEN

Metastatic castration-resistant prostate cancer (mCRPC) is a heterogeneous disease with diverse drivers of disease progression and mechanisms of therapeutic resistance. We conducted deep phenotypic characterization of CRPC metastases and patient-derived xenograft (PDX) lines using whole genome RNA sequencing, gene set enrichment analysis and immunohistochemistry. Our analyses revealed five mCRPC phenotypes based on the expression of well-characterized androgen receptor (AR) or neuroendocrine (NE) genes: (i) AR-high tumors (ARPC), (ii) AR-low tumors (ARLPC), (iii) amphicrine tumors composed of cells co-expressing AR and NE genes (AMPC), (iv) double-negative tumors (i.e. AR-/NE-; DNPC) and (v) tumors with small cell or NE gene expression without AR activity (SCNPC). RE1-silencing transcription factor (REST) activity, which suppresses NE gene expression, was lost in AMPC and SCNPC PDX models. However, knockdown of REST in cell lines revealed that attenuated REST activity drives the AMPC phenotype but is not sufficient for SCNPC conversion. We also identified a subtype of DNPC tumors with squamous differentiation and generated an encompassing 26-gene transcriptional signature that distinguished the five mCRPC phenotypes. Together, our data highlight the central role of AR and REST in classifying treatment-resistant mCRPC phenotypes. These molecular classifications could potentially guide future therapeutic studies and clinical trial design.


Asunto(s)
Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias , Neoplasias de la Próstata Resistentes a la Castración , Transcripción Genética , Humanos , Masculino , Metástasis de la Neoplasia , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Células PC-3 , Neoplasias de la Próstata Resistentes a la Castración/clasificación , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología
6.
Sci Transl Med ; 11(503)2019 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-31366581

RESUMEN

The androgen receptor (AR) is a driver of cellular differentiation and prostate cancer development. An extensive body of work has linked these normal and aberrant cellular processes to mRNA transcription; however, the extent to which AR regulates posttranscriptional gene regulation remains unknown. Here, we demonstrate that AR uses the translation machinery to shape the cellular proteome. We show that AR is a negative regulator of protein synthesis and identify an unexpected relationship between AR and the process of translation initiation in vivo. This is mediated through direct transcriptional control of the translation inhibitor 4EBP1. We demonstrate that lowering AR abundance increases the assembly of the eIF4F translation initiation complex, which drives enhanced tumor cell proliferation. Furthermore, we uncover a network of pro-proliferation mRNAs characterized by a guanine-rich cis-regulatory element that is particularly sensitive to eIF4F hyperactivity. Using both genetic and pharmacologic methods, we demonstrate that dissociation of the eIF4F complex reverses the proliferation program, resulting in decreased tumor growth and improved survival in preclinical models. Our findings reveal a druggable nexus that functionally links the processes of mRNA transcription and translation initiation in an emerging class of lethal AR-deficient prostate cancer.


Asunto(s)
Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Regulón/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular/genética , Proliferación Celular/fisiología , Humanos , Técnicas In Vitro , Intrones/genética , Masculino , Ratones , Neoplasias de la Próstata/genética , Receptores Androgénicos/genética , Regulón/genética
7.
Eur J Phys Rehabil Med ; 52(6): 774-781, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27575012

RESUMEN

BACKGROUND: Hemiplegic shoulder pain (HSP) impedes functional motor recovery of the affected limbs and negatively affects quality of life and daily activities. Kinesiology taping (KT) may provide improvement in hemiplegic shoulder pain and upper extremity function after an acute stroke. AIM: To assess the impact of KT on HSP, upper extremity functional outcomes, and the prevention of shoulder soft tissue injury in subacute stroke patients with hemiplegic shoulders during rehabilitation. DESIGN: Randomized, double-blind controlled trial. SETTING: Rehabilitation unit at a single medical center. POPULATION: Forty-four subacute stroke patients with hemiplegia. METHODS: Forty-four subacute stroke patients with hemiplegia participated in this study and were randomly allocated to the control group (sham KT) or experimental group (therapeutic KT). In the experimental group, a 3-week therapeutic KT with conventional inpatient rehabilitation was applied for 5 days per week. In the control group, the patients received a 3-week sham KT with conventional inpatient rehabilitation for 5 days per week. Shoulder subluxation, spasticity, hemiplegic shoulder pain, the Fugl-Meyer Assessment for Upper Extremity (FMA-UE), modified Barthel Index (MI), Stroke-Specific Quality of Life (SSQOL) scale, and shoulder sonography were measured before and after treatment. RESULTS: Pain-free flexion was significantly increased in hemiplegic shoulders after therapeutic KT. From 16 (70%) to 20 (87%) patients in the control and from 12 (57%) to 12 (57%) in the experimental groups had HSP after intervention, and a significant difference in the occurrence of HSP was found between these groups after treatment (P<0.05). Significant improvements (P<0.05) were noted in the FMA-UE, modified BI, and SSQOL scales after treatment in both groups. No significant differences between the groups were seen on shoulder sonography (P>0.05). CONCLUSIONS: Therapeutic KT may limit the development of HSP and improve shoulder flexion in subacute stroke patients with flaccid shoulders during inpatient rehabilitation. For subacute stroke patients with hemiplegia, therapeutic KT may not provide improvements in the upper extremity function, daily activity, and quality of life over sham KT during conventional inpatient rehabilitation. CLINICAL REHABILITATION IMPACT: Kinesiology taping may provide positive effects on shoulder flexion and decrease the occurrence of HSP in subacute stroke patients with hemiplegic shoulders during conventional inpatient rehabilitation.


Asunto(s)
Cinta Atlética , Hemiplejía/rehabilitación , Manejo del Dolor/métodos , Dolor de Hombro/rehabilitación , Rehabilitación de Accidente Cerebrovascular/métodos , Método Doble Ciego , Femenino , Hemiplejía/diagnóstico por imagen , Hemiplejía/fisiopatología , Humanos , Masculino , Persona de Mediana Edad , Espasticidad Muscular/fisiopatología , Espasticidad Muscular/rehabilitación , Dimensión del Dolor , Calidad de Vida , Dolor de Hombro/diagnóstico por imagen , Dolor de Hombro/fisiopatología , Resultado del Tratamiento , Ultrasonografía
8.
J Neurosci ; 23(23): 8360-9, 2003 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-12967998

RESUMEN

Opioid-receptor activation in cell lines results in phosphorylation of p42/44 mitogen-activated protein kinase (MAPK), which contributes to agonist-induced desensitization of adenylate cyclase signaling. In this study, morphine-induced MAPK modulation was examined in the mouse brain using antibodies against phosphorylated MAPK. Thirty minutes after systemic morphine, MAPK modulation was observed in brain areas associated with analgesia and reward. Activation of MAPK was increased in the anterior cingulate (Acc), somato-sensory and association cortices, and locus ceruleus (LC). In contrast, MAPK activation was decreased in the nucleus accumbens and central amygdala (CeA). Double-label confocal microscopy revealed that morphine-induced MAPK modulation occurred predominantly in cells not expressing mu-opioid receptors, with the exception of the LC. Furthermore, the NMDA receptor antagonist 3,3-(2-carboxypiperazine-4-yl)-propyl-1-phosphonate blocked morphine-induced MAPK modulation in several cortical areas including the Acc. We then examined morphine-induced MAPK modulation during expression of either analgesic tolerance or locomotor sensitization, which were differentiated by two repeated morphine regimens. Analgesic tolerance was accompanied by tolerance to morphine-induced MAPK modulation in all of the brain areas examined except the CeA. Locomotor sensitization resulted in sensitization to morphine-induced MAPK activation in the posterior basolateral amygdala. Additionally, a pronounced instatement of morphine-induced MAPK activation was observed in CA3 hippocampal processes. This instatement was observed during expression of tolerance; however, it was not significant during sensitization. In summary, these results provide distinct, region-specific mechanisms for morphine-induced MAPK modulation in the mouse brain and give insight into the brain circuitry involved in acute and adaptive opioid behaviors.


Asunto(s)
Analgésicos Opioides/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Morfina/farmacología , Animales , Esquema de Medicación , Tolerancia a Medicamentos/fisiología , Activación Enzimática/efectos de los fármacos , Antagonistas de Aminoácidos Excitadores/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Actividad Motora/efectos de los fármacos , Dimensión del Dolor/efectos de los fármacos , Fosforilación , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores Opioides mu/metabolismo , Recompensa
9.
J Neurosci ; 23(32): 10331-7, 2003 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-14614092

RESUMEN

Buprenorphine is a mixed opioid receptor agonist-antagonist used clinically for maintenance therapy in opiate addicts and pain management. Dose-response curves for buprenorphine-induced antinociception display ceiling effects or are bell shaped, which have been attributed to the partial agonist activity of buprenorphine at opioid receptors. Recently, buprenorphine has been shown to activate opioid receptor-like (ORL-1) receptors, also known as OP4 receptors. Here we demonstrate that buprenorphine, but not morphine, activates mitogen-activated protein kinase and Akt via ORL-1 receptors. Because the ORL-1 receptor agonist orphanin FQ/nociceptin blocks opioid-induced antinociception, we tested the hypothesis that buprenorphine-induced antinociception might be compromised by concomitant activation of ORL-1 receptors. In support of this hypothesis, the antinociceptive effect of buprenorphine, but not morphine, was markedly enhanced in mice lacking ORL-1 receptors using the tail-flick assay. Additional support for a modulatory role for ORL-1 receptors in buprenorphine-induced antinociception was that coadministration of J-113397, an ORL-1 receptor antagonist, enhanced the antinociceptive efficacy of buprenorphine in wild-type mice but not in mice lacking ORL-1 receptors. The ORL-1 antagonist also eliminated the bell-shaped dose-response curve for buprenorphine-induced antinociception in wild-type mice. Although buprenorphine has been shown to interact with multiple opioid receptors, mice lacking micro-opioid receptors failed to exhibit antinociception after buprenorphine administration. Our results indicate that the antinociceptive effect of buprenorphine in mice is micro-opioid receptor-mediated yet severely compromised by concomitant activation of ORL-1 receptors.


Asunto(s)
Buprenorfina/farmacología , Dimensión del Dolor/efectos de los fármacos , Proteínas Serina-Treonina Quinasas , Receptores Opioides mu/metabolismo , Receptores Opioides/metabolismo , Analgésicos Opioides/farmacología , Animales , Bencimidazoles/farmacología , Células Cultivadas , Cricetinae , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Morfina/farmacología , Antagonistas de Narcóticos/farmacología , Péptidos Opioides/farmacología , Piperidinas/farmacología , Proteínas Proto-Oncogénicas/efectos de los fármacos , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Receptores Opioides/deficiencia , Receptores Opioides/efectos de los fármacos , Receptores Opioides mu/deficiencia , Receptores Opioides mu/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Receptor de Nociceptina , Nociceptina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA