Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Nature ; 560(7716): 55-60, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30022166

RESUMO

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.


Assuntos
Sistema Nervoso Central/patologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Animais , Anticorpos Neutralizantes/imunologia , Membrana Basal/metabolismo , Barreira Hematoencefálica/metabolismo , Medula Óssea , Movimento Celular , Sistema Nervoso Central/irrigação sanguínea , Sistema Nervoso Central/metabolismo , Líquido Cefalorraquidiano/metabolismo , Circulação Cerebrovascular , Classe I de Fosfatidilinositol 3-Quinases/antagonistas & inibidores , Progressão da Doença , Feminino , Xenoenxertos/imunologia , Xenoenxertos/patologia , Integrina alfa6/imunologia , Integrina alfa6/metabolismo , Laminina/metabolismo , Masculino , Camundongos , Camundongos SCID , Transplante de Neoplasias , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Receptores de Laminina/antagonistas & inibidores , Receptores de Laminina/imunologia , Receptores de Laminina/metabolismo , Crânio , Espaço Subaracnóideo
2.
Blood ; 121(24): 4821-31, 2013 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-23589674

RESUMO

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.


Assuntos
Crise Blástica/metabolismo , Medula Óssea/metabolismo , Osteoblastos/metabolismo , Osteopontina/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Microambiente Tumoral , Adulto , Animais , Anticorpos Neutralizantes/farmacologia , Antimetabólitos Antineoplásicos/farmacologia , Crise Blástica/tratamento farmacológico , Crise Blástica/genética , Crise Blástica/patologia , Medula Óssea/patologia , Adesão Celular/efeitos dos fármacos , Adesão Celular/genética , Criança , Pré-Escolar , Citarabina/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Humanos , Antígeno Ki-67/genética , Antígeno Ki-67/metabolismo , Masculino , Camundongos , Camundongos SCID , Transplante de Neoplasias , Neoplasia Residual/genética , Neoplasia Residual/metabolismo , Neoplasia Residual/patologia , Osteoblastos/patologia , Osteopontina/antagonistas & inibidores , Osteopontina/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Transdução de Sinais , Transplante Heterólogo
3.
Science ; 384(6702): eadh5548, 2024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-38900896

RESUMO

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.


Assuntos
Neoplasias Ósseas , Neoplasias da Mama , Integrina alfa6 , Neoplasias Meníngeas , Meninges , Vias Neurais , Animais , Feminino , Humanos , Camundongos , Membrana Basal/metabolismo , Neoplasias Ósseas/secundário , Neoplasias Ósseas/metabolismo , Neoplasias da Mama/patologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Movimento Celular , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Integrina alfa6/metabolismo , Laminina/metabolismo , Macrófagos/metabolismo , Neoplasias Meníngeas/metabolismo , Neoplasias Meníngeas/secundário , Meninges/patologia , Invasividade Neoplásica , Moléculas de Adesão de Célula Nervosa/metabolismo , Moléculas de Adesão de Célula Nervosa/genética , Transdução de Sinais , Vias Neurais/metabolismo , Camundongos SCID , Camundongos Knockout
4.
Semin Cancer Biol ; 20(2): 107-15, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20510363

RESUMO

Somatic stem cells play a well-defined and important role in tissue renewal. Their malignant counterparts, cancer stem cells, are thought to be responsible for tumor initiation and possibly chemotherapy resistance, although controversy remains regarding both the origin and characterization of these cells. Both somatic and cancer stem cells appear to occupy specialized microenvironments in many organs. These niches are important for both maintenance of quiescence and control of cellular survival and proliferation. Targeting cancer stem cells and their microenvironments may provide new therapies to eradicate tumors. The efficacy of several drugs in current use is mediated at least in part via effects on the microenvironment, and new drugs that target the niche are currently in clinical trials.


Assuntos
Diferenciação Celular , Neoplasias/patologia , Neoplasias/terapia , Células-Tronco Neoplásicas/patologia , Células-Tronco/fisiologia , Animais , Transformação Celular Neoplásica , Humanos
5.
Nature ; 435(7044): 969-73, 2005 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-15959517

RESUMO

The organization of cellular niches is known to have a key role in regulating normal stem cell differentiation and regeneration, but relatively little is known about the architecture of microenvironments that support malignant metastasis. Using dynamic in vivo confocal imaging, here we show that murine bone marrow contains unique anatomic regions defined by specialized endothelium. This vasculature expresses the adhesion molecule E-selectin and the chemoattractant stromal-cell-derived factor 1 (SDF-1) in discrete, discontinuous areas that influence the homing of a variety of tumour cell lines. Disruption of the interactions between SDF-1 and its receptor CXCR4 inhibits the homing of Nalm-6 cells (an acute lymphoblastic leukaemia cell line) to these vessels. Further studies revealed that circulating leukaemic cells can engraft around these vessels, suggesting that this molecularly distinct vasculature demarcates a microenvironment for early metastatic tumour spread in bone marrow. Finally, purified haematopoietic stem/progenitor cells and lymphocytes also localize to the same microdomains, indicating that this vasculature might also function in benign states to demarcate specific portals for the entry of cells into the marrow space. Specialized vascular structures therefore appear to delineate a microenvironment with unique physiology that can be exploited by circulating malignant cells.


Assuntos
Células da Medula Óssea/citologia , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Microscopia Confocal/métodos , Neoplasias/patologia , Animais , Células da Medula Óssea/metabolismo , Adesão Celular , Linhagem Celular Tumoral , Movimento Celular , Citocinas/metabolismo , Selectina E/metabolismo , Leucemia/metabolismo , Leucemia/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Receptores CXCR4/metabolismo , Crânio/citologia
6.
Nat Rev Cancer ; 21(7): 461-475, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33953370

RESUMO

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.


Assuntos
Leucemia/patologia , Metástase Neoplásica , Animais , Movimento Celular , Reprogramação Celular , Humanos , Leucemia/tratamento farmacológico , Invasividade Neoplásica
7.
Mol Cancer Ther ; 20(3): 455-466, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33402399

RESUMO

Paget's "seed and soil" hypothesis of metastatic spread has acted as a foundation of the field for over a century, with continued evolution as mechanisms of the process have been elucidated. The central nervous system (CNS) presents a unique soil through this lens, relatively isolated from peripheral circulation and immune surveillance with distinct cellular and structural composition. Research in primary and metastatic brain tumors has demonstrated that this tumor microenvironment (TME) plays an essential role in the growth of CNS tumors. In each case, the cancerous cells develop complex and bidirectional relationships that reorganize the local TME and reprogram the CNS cells, including endothelial cells, pericytes, astrocytes, microglia, infiltrating monocytes, and lymphocytes. These interactions create a structurally and immunologically permissive TME with malignant processes promoting positive feedback loops and systemic consequences. Strategies to interrupt interactions with the native CNS components, on "salting the soil," to create an inhospitable environment are promising in the preclinical setting. This review aims to examine the general and specific pathways thus far investigated in brain metastases and related work in glioma to identify targetable mechanisms that may have general application across the spectrum of intracranial tumors.


Assuntos
Neoplasias Encefálicas/patologia , Humanos , Metástase Neoplásica , Microambiente Tumoral
8.
Leuk Lymphoma ; 62(11): 2690-2702, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34355654

RESUMO

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.


Assuntos
Neoplasias do Sistema Nervoso Central , Leucemia-Linfoma Linfoblástico de Células Precursoras , Animais , Neoplasias do Sistema Nervoso Central/tratamento farmacológico , Humanos , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Recidiva , Transdução de Sinais , Microambiente Tumoral
9.
Adv Sci (Weinh) ; 7(5): 1900860, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32154065

RESUMO

The roles of mitochondrial dysfunction in carcinogenesis remain largely unknown. The effects of PTEN-induced putative kinase 1 (PINK1)-dependent mitophagy on the pathogenesis of multiple myeloma (MM) are determined. The levels of the PINK1-dependent mitophagy markers PINK1 and parkin RBR E3 ubiquitin protein ligase (PARK2) in CD138+ plasma cells are reduced in patients with MM and correlate with clinical outcomes in myeloma patients. Moreover, the induction of PINK1-dependent mitophagy with carbonylcyanide-m-chlorophenylhydrazone (CCCP) or salinomycin, or overexpression of PINK1 leads to inhibition of transwell migration, suppression of myeloma cell homing to calvarium, and decreased osteolytic bone lesions. Furthermore, genetic deletion of pink1 accelerates myeloma development in a spontaneous X-box binding protein-1 spliced isoform (XBP-1s) transgenic myeloma mouse model and in VK*MYC transplantable myeloma recipient mice. Additionally, treatment with salinomycin shows significant antimyeloma activities in vivo in murine myeloma xenograft models. Finally, the effects of PINK1-dependent mitophagy on myeloma pathogenesis are driven by the activation of the Mps one binder kinase activator (MOB1B)-mediated Hippo pathway and the subsequent downregulation of Yes-associated protein (YAP)/transcriptional co-activator with PDZ-binding motif (TAZ) expression. These data provide direct evidence that PINK1-dependent mitophagy plays a critical role in the pathogenesis of MM and is a potential therapeutic target.

11.
Mol Cell Oncol ; 4(4): e1214771, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28868340

RESUMO

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.

12.
JCI Insight ; 2(24)2017 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-29263312

RESUMO

Primary myelofibrosis is a myeloproliferative neoplasm associated with significant morbidity and mortality, for which effective therapies are lacking. ß-Arrestins are multifunctional adaptor proteins involved in developmental signaling pathways. One isoform, ß-arrestin2 (ßarr2), has been implicated in initiation and progression of chronic myeloid leukemia, another myeloproliferative neoplasm closely related to primary myelofibrosis. Accordingly, we investigated the relationship between ßarr2 and primary myelofibrosis. In a murine model of MPLW515L-mutant primary myelofibrosis, mice transplanted with donor ßarr2-knockout (ßarr2-/-) hematopoietic stem cells infected with MPL-mutant retrovirus did not develop myelofibrosis, whereas controls uniformly succumbed to disease. Although transplanted ßarr2-/- cells homed properly to marrow, they did not repopulate long-term due to increased apoptosis and decreased self-renewal of ßarr2-/- cells. In order to assess the effect of acute loss of ßarr2 in established primary myelofibrosis in vivo, we utilized a tamoxifen-induced Cre-conditional ßarr2-knockout mouse. Mice that received Cre (+) donor cells and developed myelofibrosis had significantly improved survival compared with controls. These data indicate that lack of antiapoptotic ßarr2 mediates marrow failure of murine hematopoietic stem cells overexpressing MPLW515L. They also indicate that ßarr2 is necessary for progression of primary myelofibrosis, suggesting that it may serve as a novel therapeutic target in this disease.


Assuntos
Mielofibrose Primária/metabolismo , beta-Arrestina 2/fisiologia , Animais , Apoptose/fisiologia , Medula Óssea/patologia , Proliferação de Células/fisiologia , Modelos Animais de Doenças , Progressão da Doença , Sobrevivência de Enxerto/fisiologia , Transplante de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/metabolismo , Masculino , Camundongos Knockout , Mielofibrose Primária/induzido quimicamente , Mielofibrose Primária/patologia , Tamoxifeno , beta-Arrestina 2/deficiência , beta-Arrestina 2/genética
13.
Sci Transl Med ; 8(340): 340ra73, 2016 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-27225183

RESUMO

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.


Assuntos
Neoplasias da Mama/complicações , Neoplasias da Mama/metabolismo , Micrometástase de Neoplasia/prevenção & controle , Animais , Benzilaminas , Medula Óssea/metabolismo , Medula Óssea/patologia , Linhagem Celular Tumoral , Quimiocina CXCL12/antagonistas & inibidores , Quimiocina CXCL12/metabolismo , Ciclamos , Selectina E/antagonistas & inibidores , Selectina E/metabolismo , Feminino , Citometria de Fluxo , Compostos Heterocíclicos/farmacologia , Humanos , Imuno-Histoquímica , Células MCF-7 , Camundongos , Camundongos SCID , Microscopia Confocal , Micrometástase de Neoplasia/patologia , Micrometástase de Neoplasia/fisiopatologia , Recidiva Local de Neoplasia/metabolismo , Recidiva Local de Neoplasia/patologia , Recidiva Local de Neoplasia/prevenção & controle , Ligação Proteica , Receptores CXCR4/antagonistas & inibidores , Receptores CXCR4/metabolismo , Células Tumorais Cultivadas
14.
Cell Stem Cell ; 15(3): 261-262, 2014 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-25192459

RESUMO

In recent years, it has become increasingly evident that hematological malignancies can alter their microenvironment, but the therapeutic implications of these changes and potential targets have not been well characterized. Recent findings now describe how sympathetic neuropathy can drive malignant transformation of the hematopoietic stem cell niche in hematopoietic malignancies.


Assuntos
Doenças do Sistema Nervoso Autônomo/patologia , Transformação Celular Neoplásica/patologia , Nicho de Células-Tronco , Animais , Células-Tronco Hematopoéticas/patologia , Humanos , Camundongos
15.
Leuk Lymphoma ; 54(10): 2274-80, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23418895

RESUMO

Our group has previously demonstrated that expression of the cytokine stem cell factor (SCF) by leukemic blasts is a frequent finding in pre-B acute lymphoblastic leukemia (ALL). Whether SCF expression is a feature of other B cell malignancies and whether cross-talk from the local microenvironment modulates malignant cell SCF production are, however, unknown. Here we show using immunohistochemistry that SCF is expressed by a wide variety of indolent and aggressive B cell malignancies involving the bone marrow (BM) or lymph nodes (LNs). In diseases such as follicular lymphoma (FL), however, where lymphoma cells uniquely associate with the BM endosteal niche, BM lymphoma does not express SCF, while LN involvement is SCF positive. In contrast, cases of FL with high-grade transformation in the BM are SCF positive. These data suggest that lymphoma cell interaction with the endosteal niche inhibits SCF production, and that FL cells become independent of this microenvironment effect following transformation.


Assuntos
Leucemia de Células B/metabolismo , Leucemia de Células B/patologia , Linfoma de Células B/metabolismo , Linfoma de Células B/patologia , Fator de Células-Tronco/metabolismo , Nicho de Células-Tronco , Adulto , Biópsia , Medula Óssea/patologia , Humanos , Imuno-Histoquímica , Linfonodos/patologia
16.
Leuk Lymphoma ; 53(3): 445-50, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21913806

RESUMO

Patients with high-risk AML, defined as those with advanced age, relapsed/refractory disease, unfavorable molecular and cytogenetic abnormalities, therapy-related myeloid neoplasm (t-MN) and multiple medical co-morbidities tend to respond poorly to standard cytarabine and daunorubicin induction therapy and have a poor prognosis. We performed a retrospective analysis of an alternative induction regimen using high dose cytarabine (HiDAC) and mitoxantrone (MITO) administered to 78 high-risk patients with AML at The University of Chicago from 2001 to 2008. The primary endpoints of the study were complete remission (CR) rate and death within 30 days of initiation of treatment. The median age was 63 years (range:23-85); 27% of these patients had a Charlson co-morbidity index (CCI) > 2. Forty-three (56%) patients had unfavorable cytogenetics, 28 (37%) had intermediate-risk cytogenetics and 5 (7%) had favorable cytogenetics. The CR rate was 45% and the CRi rate 10%; 7 patients (9%) died during induction. Notably, t-MN and relapsed/refractory patients had CR and induction death rates equivalent to de novo AML patients within this series. In this high risk AML population, HiDAC/MITO induction demonstrated an overall response rate of 55% with a low induction death rate of 9% and allowed 32 (41%) patients to proceed to allogeneic stem cell transplant.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Leucemia Mieloide/tratamento farmacológico , Doença Aguda , Adulto , Idoso , Idoso de 80 Anos ou mais , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Aberrações Cromossômicas , Terapia Combinada , Citarabina/administração & dosagem , Citarabina/efeitos adversos , Diarreia/induzido quimicamente , Relação Dose-Resposta a Droga , Feminino , Doenças Hematológicas/induzido quimicamente , Humanos , Infecções/etiologia , Estimativa de Kaplan-Meier , Leucemia Mieloide/genética , Leucemia Mieloide/cirurgia , Masculino , Pessoa de Meia-Idade , Mitoxantrona/administração & dosagem , Mitoxantrona/efeitos adversos , Proteínas de Neoplasias/genética , Segunda Neoplasia Primária/tratamento farmacológico , Segunda Neoplasia Primária/genética , Segunda Neoplasia Primária/cirurgia , Prognóstico , Recidiva , Indução de Remissão , Estudos Retrospectivos , Risco , Transplante de Células-Tronco , Transplante Homólogo , Adulto Jovem , Tirosina Quinase 3 Semelhante a fms/genética
17.
Transl Res ; 151(1): 1-9, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18061122

RESUMO

Specific tissue microenvironments, or niches, are critical for homing and maintenance of both stem cells and tumor cells in vivo. Little is known, however, about the molecular interactions between individual cells within these microenvironments. Recent studies that describe a newly identified hematopoietic stem and tumor cell vascular niche in the bone marrow (BM) suggest a critical role for vascular endothelial cell signaling and raise the possibility that bidirectional interactions of these cells with the vasculature regulate the niche dynamically. The mechanisms that govern hematopoietic stem cell (HSC)/tumor cell cross-talk with endothelial cells provide a promising new direction for future studies. Here we review recent advances that open new avenues of study in this field.


Assuntos
Células da Medula Óssea/fisiologia , Células Endoteliais/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Neoplasias/patologia , Neovascularização Fisiológica/fisiologia , Animais , Medula Óssea/irrigação sanguínea , Medula Óssea/fisiologia , Células da Medula Óssea/citologia , Comunicação Celular/fisiologia , Células Endoteliais/citologia , Células-Tronco Hematopoéticas/citologia , Humanos , Neoplasias/irrigação sanguínea
18.
Science ; 322(5909): 1861-5, 2008 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-19095944

RESUMO

The host tissue microenvironment influences malignant cell proliferation and metastasis, but little is known about how tumor-induced changes in the microenvironment affect benign cellular ecosystems. Applying dynamic in vivo imaging to a mouse model, we show that leukemic cell growth disrupts normal hematopoietic progenitor cell (HPC) bone marrow niches and creates abnormal microenvironments that sequester transplanted human CD34+ (HPC-enriched) cells. CD34+ cells in leukemic mice declined in number over time and failed to mobilize into the peripheral circulation in response to cytokine stimulation. Neutralization of stem cell factor (SCF) secreted by leukemic cells inhibited CD34+ cell migration into malignant niches, normalized CD34+ cell numbers, and restored CD34+ cell mobilization in leukemic mice. These data suggest that the tumor microenvironment causes HPC dysfunction by usurping normal HPC niches and that therapeutic inhibition of HPC interaction with tumor niches may help maintain normal progenitor cell function in the setting of malignancy.


Assuntos
Medula Óssea/patologia , Células-Tronco Hematopoéticas/fisiologia , Leucemia Mieloide Aguda/patologia , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patologia , Nicho de Células-Tronco/patologia , Animais , Antígenos CD34/análise , Benzilaminas , Contagem de Células , Linhagem Celular Tumoral , Movimento Celular , Quimiocina CXCL12/metabolismo , Ciclamos , Fator Estimulador de Colônias de Granulócitos/farmacologia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Compostos Heterocíclicos/farmacologia , Humanos , Leucemia Mieloide Aguda/metabolismo , Camundongos , Camundongos SCID , Transplante de Neoplasias , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/fisiopatologia , Fator de Células-Tronco/genética , Fator de Células-Tronco/metabolismo , Nicho de Células-Tronco/fisiopatologia , Transplante Heterólogo , Células Tumorais Cultivadas
19.
Cell Stem Cell ; 7(6): 645-6, 2010 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-21112557
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA