Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Science ; 384(6702): eadh5548, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38900896

RESUMEN

The molecular mechanisms that regulate breast cancer cell (BCC) metastasis and proliferation within the leptomeninges (LM) are poorly understood, which limits the development of effective therapies. In this work, we show that BCCs in mice can invade the LM by abluminal migration along blood vessels that connect vertebral or calvarial bone marrow and meninges, bypassing the blood-brain barrier. This process is dependent on BCC engagement with vascular basement membrane laminin through expression of the neuronal pathfinding molecule integrin α6. Once in the LM, BCCs colocalize with perivascular meningeal macrophages and induce their expression of the prosurvival neurotrophin glial-derived neurotrophic factor (GDNF). Intrathecal GDNF blockade, macrophage-specific GDNF ablation, or deletion of the GDNF receptor neural cell adhesion molecule (NCAM) from BCCs inhibits breast cancer growth within the LM. These data suggest integrin α6 and the GDNF signaling axis as new therapeutic targets against breast cancer LM metastasis.


Asunto(s)
Neoplasias Óseas , Neoplasias de la Mama , Integrina alfa6 , Neoplasias Meníngeas , Meninges , Vías Nerviosas , Animales , Femenino , Humanos , Ratones , Membrana Basal/metabolismo , Neoplasias Óseas/secundario , Neoplasias Óseas/metabolismo , Neoplasias de la Mama/patología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/genética , Línea Celular Tumoral , Movimiento Celular , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Integrina alfa6/metabolismo , Laminina/metabolismo , Macrófagos/metabolismo , Neoplasias Meníngeas/metabolismo , Neoplasias Meníngeas/secundario , Meninges/patología , Invasividad Neoplásica , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Moléculas de Adhesión de Célula Nerviosa/genética , Transducción de Señal , Vías Nerviosas/metabolismo , Ratones SCID , Ratones Noqueados
2.
Leuk Lymphoma ; 62(11): 2690-2702, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34355654

RESUMEN

The majority of adult patients with acute lymphoblastic leukemia (ALL) suffer relapse, and in patients with central nervous system (CNS) metastasis, prognosis is particularly poor. We recently demonstrated a novel route of ALL CNS metastasis dependent on PI3Kδ regulation of the laminin receptor integrin α6. B-ALL cells did not, however, rely on PI3Kδ signaling for growth. Here we show that broad targeting of PI3K isoforms can induce growth arrest in B-ALL, reducing systemic disease burden in mice treated with a single agent pan-PI3Ki, copanlisib. Moreover, we show that cellular stress activates PI3K/Akt-dependent survival pathways in B-ALL, exposing their vulnerability to PI3Kδ and pan-PI3Ki. The addition of a brief course of copanlisib to chemotherapy delivered the combined benefits of increased survival, decreased systemic disease, and reduced CNS metastasis. These data suggest the promising, multifaceted potential of pan-PI3Ki for B-ALL CNS prophylaxis, systemic disease control, and chemosensitization.


Asunto(s)
Neoplasias del Sistema Nervioso Central , Leucemia-Linfoma Linfoblástico de Células Precursoras , Animales , Neoplasias del Sistema Nervioso Central/tratamiento farmacológico , Humanos , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Recurrencia , Transducción de Señal , Microambiente Tumoral
3.
Nat Rev Cancer ; 21(7): 461-475, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33953370

RESUMEN

In contrast to solid cancers, which often require genetic modifications and complex cellular reprogramming for effective metastatic dissemination, leukaemic cells uniquely possess the innate ability for migration and invasion. Dedifferentiated, malignant leukocytes retain the benign leukocytes' capacity for cell motility and survival in the circulation, while acquiring the potential for rapid and uncontrolled cell division. For these reasons, leukaemias, although not traditionally considered as metastatic diseases, are in fact models of highly efficient metastatic spread. Accordingly, they are often aggressive and challenging diseases to treat. In this Perspective, we discuss the key molecular processes that facilitate metastasis in a variety of leukaemic subtypes, the clinical significance of leukaemic invasion into specific tissues and the current pipeline of treatments targeting leukaemia metastasis.


Asunto(s)
Leucemia/patología , Metástasis de la Neoplasia , Animales , Movimiento Celular , Reprogramación Celular , Humanos , Leucemia/tratamiento farmacológico , Invasividad Neoplásica
4.
Nature ; 560(7716): 55-60, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30022166

RESUMEN

Acute lymphoblastic leukaemia (ALL) has a marked propensity to metastasize to the central nervous system (CNS). In contrast to brain metastases from solid tumours, metastases of ALL seldom involve the parenchyma but are isolated to the leptomeninges, which is an infrequent site for carcinomatous invasion. Although metastasis to the CNS occurs across all subtypes of ALL, a unifying mechanism for invasion has not yet been determined. Here we show that ALL cells in the circulation are unable to breach the blood-brain barrier in mice; instead, they migrate into the CNS along vessels that pass directly between vertebral or calvarial bone marrow and the subarachnoid space. The basement membrane of these bridging vessels is enriched in laminin, which is known to coordinate pathfinding of neuronal progenitor cells in the CNS. The laminin receptor α6 integrin is expressed in most cases of ALL. We found that α6 integrin-laminin interactions mediated the migration of ALL cells towards the cerebrospinal fluid in vitro. Mice with ALL xenografts were treated with either a PI3Kδ inhibitor, which decreased α6 integrin expression on ALL cells, or specific α6 integrin-neutralizing antibodies and showed significant reductions in ALL transit along bridging vessels, blast counts in the cerebrospinal fluid and CNS disease symptoms despite minimally decreased bone marrow disease burden. Our data suggest that α6 integrin expression, which is common in ALL, allows cells to use neural migratory pathways to invade the CNS.


Asunto(s)
Sistema Nervioso Central/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Animales , Anticuerpos Neutralizantes/inmunología , Membrana Basal/metabolismo , Barrera Hematoencefálica/metabolismo , Médula Ósea , Movimiento Celular , Sistema Nervioso Central/irrigación sanguínea , Sistema Nervioso Central/metabolismo , Líquido Cefalorraquídeo/metabolismo , Circulación Cerebrovascular , Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Progresión de la Enfermedad , Femenino , Xenoinjertos/inmunología , Xenoinjertos/patología , Integrina alfa6/inmunología , Integrina alfa6/metabolismo , Laminina/metabolismo , Masculino , Ratones , Ratones SCID , Trasplante de Neoplasias , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Receptores de Laminina/antagonistas & inhibidores , Receptores de Laminina/inmunología , Receptores de Laminina/metabolismo , Cráneo , Espacio Subaracnoideo
5.
Mol Cell Oncol ; 4(4): e1214771, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28868340

RESUMEN

E-selectin is a key mediator of breast cancer cell (BCC) metastatic entry into the bone and stromal-derived factor 1 (SDF-1) is a critical molecular anchor for BCCs within discrete pro-dormancy bone marrow (BM) niches. Small-molecule inhibitors blocked metastatic entry and mobilized established disease from BM, suggesting a new treatment strategy to prevent breast cancer relapse.

6.
Sci Transl Med ; 8(340): 340ra73, 2016 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-27225183

RESUMEN

Breast cancer metastatic relapse can occur years after therapy, indicating that disseminated breast cancer cells (BCCs) have a prolonged dormant phase before becoming proliferative. A major site of disease dissemination and relapse is bone, although the critical signals that allow circulating BCCs to identify bone microvasculature, enter tissue, and tether to the microenvironment are poorly understood. Using real-time in vivo microscopy of bone marrow (BM) in a breast cancer xenograft model, we show that dormant and proliferating BCCs occupy distinct areas, with dormant BCCs predominantly found in E-selectin- and stromal cell-derived factor 1 (SDF-1)-rich perisinusoidal vascular regions. We use highly specific inhibitors of E-selectin and C-X-C chemokine receptor type 4 (CXCR4) (SDF-1 receptor) to demonstrate that E-selectin and SDF-1 orchestrate opposing roles in BCC trafficking. Whereas E-selectin interactions are critical for allowing BCC entry into the BM, the SDF-1/CXCR4 interaction anchors BCCs to the microenvironment, and its inhibition induces mobilization of dormant micrometastases into circulation. Homing studies with primary BCCs also demonstrate that E-selectin regulates their entry into bone through the sinusoidal niche, and immunohistochemical staining of patient BMs shows dormant micrometastatic disease adjacent to SDF-1(+) vasculature. These findings shed light on how BCCs traffic within the host, and suggest that simultaneous blockade of CXCR4 and E-selectin in patients could molecularly excise dormant micrometastases from the protective BM environment, preventing their emergence as relapsed disease.


Asunto(s)
Neoplasias de la Mama/complicaciones , Neoplasias de la Mama/metabolismo , Micrometástasis de Neoplasia/prevención & control , Animales , Bencilaminas , Médula Ósea/metabolismo , Médula Ósea/patología , Línea Celular Tumoral , Quimiocina CXCL12/antagonistas & inhibidores , Quimiocina CXCL12/metabolismo , Ciclamas , Selectina E/antagonistas & inhibidores , Selectina E/metabolismo , Femenino , Citometría de Flujo , Compuestos Heterocíclicos/farmacología , Humanos , Inmunohistoquímica , Células MCF-7 , Ratones , Ratones SCID , Microscopía Confocal , Micrometástasis de Neoplasia/patología , Micrometástasis de Neoplasia/fisiopatología , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/patología , Recurrencia Local de Neoplasia/prevención & control , Unión Proteica , Receptores CXCR4/antagonistas & inhibidores , Receptores CXCR4/metabolismo , Células Tumorales Cultivadas
7.
Blood ; 121(24): 4821-31, 2013 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-23589674

RESUMEN

Malignant cells may evade death from cytotoxic agents if they are in a dormant state. The host microenvironment plays important roles in cancer progression, but how niches might control cancer cell dormancy is little understood. Here we show that osteopontin (OPN), an extracellular matrix molecule secreted by osteoblasts, can function to anchor leukemic blasts in anatomic locations supporting tumor dormancy. We demonstrate that acute lymphoblastic leukemia (ALL) cells specifically adhere to OPN in vitro and secrete OPN when localized to the endosteal niche in vivo. Using intravital microscopy to perform imaging studies of the calvarial bone marrow (BM) of xenografted mice, we show that OPN is highly expressed adjacent to dormant tumor cells within the marrow. Inhibition of the OPN-signaling axis significantly increases the leukemic cell Ki-67 proliferative index and leads to a twofold increase in tumor burden in treated mice. Moreover, using cell-cycle-dependent Ara-C chemotherapy to produce minimal residual disease (MRD) in leukemic mice, we show that OPN neutralization synergizes with Ara-C to reduce detectable BM MRD. Taken together, these data suggest that ALL interacts with extracellular OPN within the malignant BM, and that this interaction induces cell cycle exit in leukemic blasts, protecting them from cytotoxic chemotherapy.


Asunto(s)
Crisis Blástica/metabolismo , Médula Ósea/metabolismo , Osteoblastos/metabolismo , Osteopontina/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Microambiente Tumoral , Adulto , Animales , Anticuerpos Neutralizantes/farmacología , Antimetabolitos Antineoplásicos/farmacología , Crisis Blástica/tratamiento farmacológico , Crisis Blástica/genética , Crisis Blástica/patología , Médula Ósea/patología , Adhesión Celular/efectos de los fármacos , Adhesión Celular/genética , Niño , Preescolar , Citarabina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Femenino , Humanos , Antígeno Ki-67/genética , Antígeno Ki-67/metabolismo , Masculino , Ratones , Ratones SCID , Trasplante de Neoplasias , Neoplasia Residual/genética , Neoplasia Residual/metabolismo , Neoplasia Residual/patología , Osteoblastos/patología , Osteopontina/antagonistas & inhibidores , Osteopontina/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Transducción de Señal , Trasplante Heterólogo
8.
Bioorg Med Chem ; 19(20): 6055-68, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-21920765

RESUMEN

Heat shock protein 90 is an emerging target for oncology therapeutics. Inhibitors of this molecular chaperone, which is responsible for the maintenance of a number of oncogenic proteins, have shown promise in clinical trials and represent a new and exciting area in the treatment of cancer. Heat shock protein 90 inhibitors have huge structural diversity, and here we present the lead identification of novel inhibitors based on ß-lactam and imine templates. ß-Lactam 5 and imines 12 and 18 exhibit binding to heat shock protein 90-α with IC(50) values of 5.6 µM, 14.5 µM, and 22.1 µM, respectively. The binding affinity displayed by these compounds positions them as lead compounds for the design of future inhibitors of heat shock protein 90 based on the ß-lactam and imine templates.


Asunto(s)
Antineoplásicos/química , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Iminas/antagonistas & inhibidores , beta-Lactamas/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Femenino , Proteínas HSP90 de Choque Térmico/química , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Modelos Moleculares , Relación Estructura-Actividad , beta-Lactamas/síntesis química , beta-Lactamas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA