Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Br J Cancer ; 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38806726

RESUMO

BACKGROUND: Intrinsic and extrinsic factors in the tumour microenvironment (TME) contribute to therapeutic resistance. Here we demonstrate that transforming growth factor (TGF)-ß1 produced in the TME increased drug resistance of neuroblastoma (NB) cells. METHODS: Human NB cell lines were tested in vitro for their sensitivity to Doxorubicin (DOX) and Etoposide (ETOP) in the presence of tumour-associated macrophages (TAM) and mesenchymal stromal cells/cancer-associated fibroblasts (MSC/CAF). These experiments were validated in xenotransplanted and primary tumour samples. RESULTS: Drug resistance was associated with an increased expression of efflux transporter and anti-apoptotic proteins. Upregulation was dependent on activation of nuclear factor (NF)-κB by TGF-ß-activated kinase (TAK1) and SMAD2. Resistance was reversed upon pharmacologic and genetic inhibitions of NF-κB, and TAK1/SMAD2. Interleukin-6, leukaemia inhibitory factor and oncostatin M were upregulated by this TGF-ß/TAK1/NF-κB/SMAD2 signalling pathway contributing to drug resistance via an autocrine loop activating STAT3. An analysis of xenotransplanted NB tumours revealed an increased presence of phospho (p)-NF-κB in tumours co-injected with MSC/CAF and TAM, and these tumours failed to respond to Etoposide but responded if treated with a TGF-ßR1/ALK5 inhibitor. Nuclear p-NF-κB was increased in patient-derived tumours rich in TME cells. CONCLUSIONS: The data provides a novel insight into a targetable mechanism of environment-mediated drug resistance.

2.
Int J Cancer ; 137(4): 797-809, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-25648303

RESUMO

The potential role of osteoblasts in bone and bone marrow (BM) metastases in neuroblastoma (NBL) remains unclear. In this study, we examined the effect of NBL cells on the osteoblastic differentiation of BM-derived mesenchymal stromal cells (BMMSC). We show that the presence of NBL cells enhanced the osteoblastic differentiation of BMMSC driven by bone morphogenetic protein (BMP)-4, in the absence of any effect on NBL cell proliferation. Expression profiles of BMMSC driven toward osteoblastic differentiation revealed an increase in vascular endothelial growth factor A (Vegfa) expression in the presence of NBL cells. We demonstrated that NBL cells increased BMMSC-derived VEGFA mRNA and protein and that this was enhanced by BMP-4. However, in similar conditions, neither the addition of an mVEGFA blocking antibody nor exogenous recombinant (r) mVEGFA affected osteoblastic differentiation. In contrast, siRNA- mediated knock-down of VEGFA in BMMSC prevented osteoblastic differentiation in BMP-4-treated cocultures, an effect that was not reversed in the presence of rmVEGFA. An analysis of murine bones injected with hNBL cells revealed an increase of mVEGFA producing cells near tumor cells concomitantly with an increase in Vegfa and Runx2 mRNA. This coincided with an increase in osteoclasts, in Rankl/Opg mRNA ratio and with the formation of osteolytic lesions. Thus NBL cells promote osteoblastogenesis in the BM by increasing VEGFA expression in BMMSC. Our study provides a new insight into the role of VEGFA in NBL metastases by pointing to the role of stroma-derived intracrine VEGFA in osteoblastogenesis.


Assuntos
Diferenciação Celular/genética , Ativação Linfocitária/genética , Células-Tronco Mesenquimais/metabolismo , Fator A de Crescimento do Endotélio Vascular/biossíntese , Animais , Proteína Morfogenética Óssea 4/administração & dosagem , Linhagem Celular , Proliferação de Células/genética , Subunidade alfa 1 de Fator de Ligação ao Core/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos , Neuroblastoma/metabolismo , Osteoblastos/metabolismo , Osteoprotegerina/biossíntese , RNA Mensageiro/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética
3.
Med Sci (Paris) ; 30(4): 445-51, 2014 Apr.
Artigo em Francês | MEDLINE | ID: mdl-24801042

RESUMO

Over the last decade, it has become clear that cancer is not just a disease of the genes, and that the tumor microenvironment (TME) plays an important role in cancer progression. Interactions between tumor cells and the TME, made of the extracellular matrix (ECM) and of non-transformed cells (designated here as stromal cells), promote cancer cell survival and drug resistance. Many of the mechanisms involved are known and are either contact-dependent or contact-independent. Contact between tumor cells and the ECM or stromal cells as well as the production of soluble factors and microvesicles all contribute. The bone marrow plays a special role in environment-mediated drug resistance as it is not only a sanctuary protecting tumor cells from cytotoxic drugs, but also a source of many stromal cells that colonize primary tumors and contribute to the pre-metastatic niche. As our understanding of the mechanisms by which the tumor microenvironment promotes therapeutic resistance progresses, clinical trials testing agents that disrupt the interaction between tumor cells and the stroma have been initiated. This new avenue of therapy is promising.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Neoplasias/tratamento farmacológico , Microambiente Tumoral , Matriz Extracelular/fisiologia , Humanos , Células Estromais/fisiologia
4.
Cancer Cell ; 7(5): 408-9, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15894261

RESUMO

In this issue of Cancer Cell, a paper by Lynch et al. demonstrates how the careful study of changes that occur at the interface between tumor cells and stromal cells led to the discovery of a new function for matrix metalloproteinase-7 (MMP-7) in the formation of osteolytic lesions in prostate cancer. The data suggest that MMP-7 inhibition could be a therapeutic target in prostate cancer.


Assuntos
Metaloproteinase 7 da Matriz/metabolismo , Neoplasias da Próstata/terapia , Células Estromais/metabolismo , Animais , Proteínas de Transporte/metabolismo , Expressão Gênica , Masculino , Metaloproteinase 7 da Matriz/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Modelos Biológicos , Osteoclastos/metabolismo , Osteoclastos/patologia , Osteólise/etiologia , Osteólise/metabolismo , Osteólise/patologia , Neoplasias da Próstata/complicações , Neoplasias da Próstata/metabolismo , Ligante RANK , Ratos , Receptor Ativador de Fator Nuclear kappa-B
5.
J Extracell Vesicles ; 12(5): e12326, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37194998

RESUMO

The capture of tumour-derived extracellular vesicles (TEVs) by cells in the tumour microenvironment (TME) contributes to metastasis and notably to the formation of the pre-metastatic niche (PMN). However, due to the challenges associated with modelling release of small EVs in vivo, the kinetics of PMN formation in response to endogenously released TEVs have not been examined. Here, we have studied the endogenous release of TEVs in mice orthotopically implanted with metastatic human melanoma (MEL) and neuroblastoma (NB) cells releasing GFP-tagged EVs (GFTEVs) and their capture by host cells to demonstrate the active contribution of TEVs to metastasis. Human GFTEVs captured by mouse macrophages in vitro resulted in transfer of GFP vesicles and the human exosomal miR-1246. Mice orthotopically implanted with MEL or NB cells showed the presence of TEVs in the blood between 5 and 28 days after implantation. Moreover, kinetic analysis of TEV capture by resident cells relative to the arrival and outgrowth of TEV-producing tumour cells in metastatic organs demonstrated that the capture of TEVs by lung and liver cells precedes the homing of metastatic tumour cells, consistent with the critical roles of TEVs in PMN formation. Importantly, TEV capture at future sites of metastasis was associated with the transfer of miR-1246 to lung macrophages, liver macrophages, and stellate cells. This is the first demonstration that the capture of endogenously released TEVs is organotropic as demonstrated by the presence of TEV-capturing cells only in metastatic organs and their absence in non-metastatic organs. The capture of TEVs in the PMN induced dynamic changes in inflammatory gene expression which evolved to a pro-tumorigenic reaction as the niche progressed to the metastatic state. Thus, our work describes a novel approach to TEV tracking in vivo that provides additional insights into their role in the earliest stages of metastatic progression.


Assuntos
Vesículas Extracelulares , Melanoma , MicroRNAs , Humanos , Animais , Camundongos , Vesículas Extracelulares/metabolismo , Cinética , MicroRNAs/metabolismo , Melanoma/metabolismo , Inflamação/metabolismo , Microambiente Tumoral
6.
Oncoimmunology ; 11(1): 2146860, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36479153

RESUMO

Tumor-associated macrophages (TAM) and cancer-associated fibroblasts (CAF) and their precursor mesenchymal stromal cells (MSC) are often detected together in tumors, but how they cooperate is not well understood. Here, we show that TAM and CAF are the most abundant nonmalignant cells and are present together in untreated human neuroblastoma (NB) tumors that are also poorly infiltrated with T and natural killer (NK) cells. We then show that MSC and CAF-MSC harvested from NB tumors protected human monocytes (MN) from spontaneous apoptosis in an interleukin (IL)-6 dependent mechanism. The interactions of MN and MSC with NB cells resulted in a significant induction or increase in the expression of several pro-tumorigenic cytokines/chemokines (TGF-ß1, MCP-1, IL-6, IL-8, and IL-4) but not of anti-tumorigenic cytokines (TNF-α, IL-12) by MN or MSC, while also inducing cytokine expression in quiescent NB cells. We then identified a TGF-ß1/IL-6 pathway where TGF-ß1 stimulated the expression of IL-6 in NB cells and MSC, promoting TAM survival. Evidence for the contribution of TAM and MSC to the activation of this pathway was then provided in xenotransplanted NB tumors and patients with primary tumors by demonstrating a direct correlation between the presence of CAF and p-SMAD2 and p-STAT3. The data highlight a new mechanism of interaction between TAM and CAF supporting their pro-tumorigenic function in cancer.


Assuntos
Fibroblastos , Interleucina-6 , Macrófagos , Neuroblastoma , Fator de Crescimento Transformador beta1 , Humanos , Neuroblastoma/imunologia , Fibroblastos/imunologia , Macrófagos/imunologia , Animais
7.
Cancer Metastasis Rev ; 29(2): 249-61, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20411303

RESUMO

Over the last decade, there has been a growing interest in the role of mesenchymal stem cells (MSC) in cancer progression. These cells have the potential to give rise to a variety of mesenchymal cells like osteoblasts, chondrocytes, adipocytes, fibroblasts, and muscle cells. In contrast to their hematopoetic counterparts, MSC are not as clearly defined, which makes the interpretation of their role in cancer progression more complex. However, the nature of the relationship between MSC and tumor cells appears dual. Primary and metastatic tumors attract MSC in their microenvironment where they become tumor-associated fibroblasts, affect tumor cell survival and angiogenesis, and have an immunomodulatory function, and vice versa in the bone marrow MSC attract tumor cells and contribute to a microenvironment that promotes osteolysis, tumor growth, survival, and drug resistance. Whether MSC are pro- or anti-tumorigenic is a subject of controversial reports that is in part explained by the complexity of their interaction with tumor cells and the large range of cytokines and growth factors they produce. The study of these interactions is a fertile ground of investigation that--as already demonstrated in the case of myeloma--should lead to novel therapeutic approaches in cancer. In this article, the biology and role of MSC in cancer is reviewed with a primary focus on bone marrow-derived MSC.


Assuntos
Células-Tronco Mesenquimais/fisiologia , Neoplasias/patologia , Animais , Células da Medula Óssea/fisiologia , Humanos , Invasividade Neoplásica/patologia , Metástase Neoplásica/patologia
8.
Pediatr Blood Cancer ; 57(2): 275-82, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21671363

RESUMO

BACKGROUND: Zoledronic acid, a bisphosphonate, delays progression of bone metastases in adult malignancies. Bone is a common metastatic site of advanced neuroblastoma. We previously reported efficacy of zoledronic acid in a murine model of neuroblastoma bone invasion prompting this Phase I trial of zoledronic acid with cyclophosphamide in children with neuroblastoma and bone metastases. The primary objective was to determine recommended dosing of zoledronic acid for future trials. PROCEDURE: Escalating doses of intravenous zoledronic acid were given every 28 days with oral metronomic cyclophosphamide (25 mg/m(2)/day). Toxicity, response, zoledronic acid pharmacokinetics, bone turnover markers, serum IL-6, and sIL-6R were evaluated. RESULTS: Twenty-one patients, median age 7.5 (range 0.8-25.6) years were treated with 2 mg/m(2) (n = 4), 3 mg/m(2) (n = 3), or 4 mg/m(2) (n = 14) zoledronic acid. Fourteen patients were evaluable for dose escalation. A median of one (range 1-18) courses was given. Two dose limiting toxicities (grade 3 hypophosphatemia) occurred at 4 mg/m(2) zoledronic acid. Other grades 3-4 toxicities included hypocalcemia (n = 2), elevated transaminases (n = 1), neutropenia (n = 2), anemia (n = 1), lymphopenia (n = 1), and hypokalemia (n = 1). Osteosclerosis contributed to fractures in one patient after 18 courses. Responses in evaluable patients included 1 partial response, 9 stable disease (median 4.5 courses, range 3-18), and 10 progressions. Zoledronic acid pharmacokinetics were similar to adults. Markers of osteoclast activity and serum IL-6 levels decreased with therapy. CONCLUSIONS: Zoledronic acid with metronomic cyclophosphamide is well tolerated with clinical and biologic responses in recurrent/refractory neuroblastoma. The recommended dose of zoledronic acid is 4 mg/m(2) every 28 days.


Assuntos
Neoplasias Ósseas/prevenção & controle , Neoplasias Ósseas/secundário , Difosfonatos/administração & dosagem , Imidazóis/administração & dosagem , Neuroblastoma/tratamento farmacológico , Adolescente , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Criança , Pré-Escolar , Ciclofosfamida/administração & dosagem , Ciclofosfamida/efeitos adversos , Difosfonatos/efeitos adversos , Difosfonatos/farmacocinética , Feminino , Humanos , Imidazóis/efeitos adversos , Imidazóis/farmacocinética , Lactente , Masculino , Dose Máxima Tolerável , Neuroblastoma/patologia , Análise de Sobrevida , Ácido Zoledrônico
9.
J Clin Invest ; 117(9): 2702-12, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17710228

RESUMO

Valpha24-invariant natural killer T (NKT) cells are potentially important for antitumor immunity. We and others have previously demonstrated positive associations between NKT cell presence in primary tumors and long-term survival in distinct human cancers. However, the mechanism by which aggressive tumors avoid infiltration with NKT and other T cells remains poorly understood. Here, we report that the v-myc myelocytomatosis viral related oncogene, neuroblastoma derived (MYCN), the hallmark of aggressive neuroblastoma, repressed expression of monocyte chemoattractant protein-1/CC chemokine ligand 2 (MCP-1/CCL2), a chemokine required for NKT cell chemoattraction. MYCN knockdown in MYCN-amplified neuroblastoma cell lines restored CCL2 production and NKT cell chemoattraction. Unlike other oncogenes, MYCN repressed chemokine expression in a STAT3-independent manner, requiring an E-box element in the CCL2 promoter to mediate transcriptional repression. MYCN overexpression in neuroblastoma xenografts in NOD/SCID mice severely inhibited their ability to attract human NKT cells, T cells, and monocytes. Patients with MYCN-amplified neuroblastoma metastatic to bone marrow had 4-fold fewer NKT cells in their bone marrow than did their nonamplified counterparts, indicating that the MYCN-mediated immune escape mechanism, which we believe to be novel, is operative in metastatic cancer and should be considered in tumor immunobiology and for the development of new therapeutic strategies.


Assuntos
Movimento Celular , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Proteínas Nucleares/metabolismo , Proteínas Oncogênicas/metabolismo , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/metabolismo , Animais , Medula Óssea/patologia , Células Cultivadas , Quimiocina CCL2/metabolismo , Feminino , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos SCID , Proteína Proto-Oncogênica N-Myc , Metástase Neoplásica/patologia , Neuroblastoma/genética , Proteínas Nucleares/genética , Proteínas Oncogênicas/genética , Fenótipo , Regiões Promotoras Genéticas/genética , Ligação Proteica , Interferência de RNA , Fator de Transcrição STAT3/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Cancers (Basel) ; 12(11)2020 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-33114328

RESUMO

The tumor microenvironment (TME) plays a critical role in tumor progression. Among its multiple components are cancer-associated fibroblasts (CAFs) that are the main suppliers of extracellular matrix molecules and important contributors to inflammation. As a source of growth factors, cytokines, chemokines and other regulatory molecules, they participate in cancer progression, metastasis, angiogenesis, immune cell reprogramming and therapeutic resistance. Nevertheless, their role is not fully understood, and is sometimes controversial due to their heterogeneity. CAFs are heterogeneous in their origin, phenotype, function and presence within tumors. As a result, strategies to target CAFs in cancer therapy have been hampered by the difficulties in better defining the various populations of CAFs and by the lack of clear recognition of their specific function in cancer progression. This review discusses how a greater understanding of the heterogeneous nature of CAFs could lead to better approaches aimed at their use or at their targeting in the treatment of cancer.

11.
Cancers (Basel) ; 12(10)2020 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-33050533

RESUMO

The contribution of the tumor microenvironment (TME) to cancer progression has been well recognized in recent decades. As cancer therapeutic strategies are increasingly precise and include immunotherapies, knowledge of the nature and function of the TME in a tumor becomes essential. Our understanding of the TME in neuroblastoma (NB), the second most common solid tumor in children, has significantly progressed from an initial focus on its Schwannian component to a better awareness of its complex nature, which includes not only immune but also non-immune cells such as cancer-associated fibroblasts (CAFs), the contribution of which to inflammation and interaction with tumor-associated macrophages (TAMs) is now recognized. Recent studies on the TME landscape of NB tumors also suggest significant differences between MYCN-amplified (MYCN-A) and non-amplified (MYCN-NA) tumors, in their content in stromal and inflammatory cells and their immunosuppressive activity. Extracellular vesicles (EVs) released by cells in the TME and microRNAs (miRs) present in their cargo could play important roles in the communication between NB cells and the TME. This review article discusses these new aspects of the TME in NB and the impact that information on the TME landscape in NB will have in the design of precise, biomarker-integrated clinical trials.

12.
Cancer Res ; 67(19): 9346-55, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17909043

RESUMO

Metastasis to the bone is seen in 56% of patients with neuroblastoma and contributes to morbidity and mortality. Using a murine model of bone invasion, we have reported previously that neuroblastoma cells invade the bone by activating osteoclasts. Here, we investigated the antitumoral and antiosteolytic activities of zoledronic acid, a bisphosphonate inhibitor of osteoclasts, in combination with cytotoxic chemotherapy in our model. We first show that zoledronic acid given at the same time (early prevention) or 2 weeks after tumor cell injection (late prevention) significantly prevented the formation of severe osteolytic lesions. It also prevented formation of these lesions when given 4 weeks after tumor cell injection (intervention) when combined with chemotherapy including cyclophosphamide and topotecan. The combination of zoledronic acid + cyclophosphamide/topotecan also significantly improved survival (P < 0.001). In mice treated with zoledronic acid, we observed a marked inhibition of osteoclasts inside the bone associated with a decrease in tumor cell proliferation and increase in tumor cell apoptosis. In vitro, zoledronic acid inhibited neuroblastoma cell proliferation and induced apoptosis, and these effects were significantly enhanced by the addition of 4-hydroxyperoxycyclophosphamide (4-HC). The proapoptotic effect of zoledronic acid and zoledronic acid in combination with 4-HC on tumor cells was associated with an increase in caspase-3 activity and a decrease in phosphorylated Bcl-2, Bcl-2, and Bcl-X(L) expression. Zoledronic acid inhibited the association of Ras with the plasma membrane and activation of c-Raf, Akt, and extracellular signal-regulated kinase 1/2. The data indicate that zoledronic acid, in addition to inhibiting osteoclasts, is active against tumor cells and suggest that zoledronic acid in combination with cytotoxic chemotherapy may be effective in children with neuroblastoma that has metastasized to the bone.


Assuntos
Conservadores da Densidade Óssea/farmacologia , Neoplasias Ósseas/prevenção & controle , Neoplasias Ósseas/secundário , Difosfonatos/farmacologia , Imidazóis/farmacologia , Neuroblastoma/tratamento farmacológico , Neuroblastoma/patologia , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias Ósseas/metabolismo , Caspase 3/metabolismo , Processos de Crescimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ciclofosfamida/administração & dosagem , Difosfonatos/administração & dosagem , Ativação Enzimática/efeitos dos fármacos , Humanos , Imidazóis/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Neuroblastoma/metabolismo , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Osteoclastos/patologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Topotecan/administração & dosagem , Ensaios Antitumorais Modelo de Xenoenxerto , Ácido Zoledrônico , Proteínas ras/metabolismo
13.
Clin Cancer Res ; 25(15): 4761-4774, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31068371

RESUMO

PURPOSE: We determined whether elimination of CD105+ cells in the tumor microenvironment (TME) with anti-CD105 antibodies enhanced anti-disialoganglioside (GD2) antibody dinutuximab therapy of neuroblastoma when combined with activated natural killer (aNK) cells. EXPERIMENTAL DESIGN: The effect of MSCs and monocytes on antibody-dependent cellular cytotoxicity (ADCC) mediated by dinutuximab with aNK cells against neuroblastoma cells was determined in vitro. ADCC with anti-CD105 mAb TRC105 and aNK cells against MSCs, monocytes, and endothelial cells, which express CD105, was evaluated. Anti-neuroblastoma activity in immunodeficient NSG mice of dinutuximab with aNK cells without or with anti-CD105 mAbs was determined using neuroblastoma cell lines and a patient-derived xenograft. RESULTS: ADCC mediated by dinutuximab with aNK cells against neuroblastoma cells in vitro was suppressed by addition of MSCs and monocytes, and dinutuximab with aNK cells was less effective against neuroblastomas formed with coinjected MSCs and monocytes in NSG mice than against those formed by tumor cells alone. Anti-CD105 antibody TRC105 with aNK cells mediated ADCC against MSCs, monocytes, and endothelial cells. Neuroblastomas formed in NSG mice by two neuroblastoma cell lines or a patient-derived xenograft coinjected with MSCs and monocytes were most effectively treated with dinutuximab and aNK cells when anti-human (TRC105) and anti-mouse (M1043) CD105 antibodies were added, which depleted human MSCs and murine endothelial cells and macrophages from the TME. CONCLUSIONS: Immunotherapy of neuroblastoma with anti-GD2 antibody dinutuximab and aNK cells is suppressed by CD105+ cells in the TME, but suppression is overcome by adding anti-CD105 antibodies to eliminate CD105+ cells.


Assuntos
Citotoxicidade Celular Dependente de Anticorpos/imunologia , Antineoplásicos/farmacologia , Endoglina/antagonistas & inibidores , Gangliosídeos/antagonistas & inibidores , Imunoterapia/métodos , Células Matadoras Naturais/imunologia , Neuroblastoma/tratamento farmacológico , Animais , Anticorpos Monoclonais/farmacologia , Linhagem Celular Tumoral , Endoglina/imunologia , Gangliosídeos/imunologia , Humanos , Células Matadoras Naturais/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Neuroblastoma/imunologia , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Cancer Res ; 66(5): 2691-9, 2006 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-16510589

RESUMO

The Wnt signaling transduction pathway plays a critical role in the pathogenesis of several murine and human epithelial cancers. Here, we have used mouse mammary tumor virus (MMTV)-Wnt1 transgenic mice, which develop spontaneous mammary adenocarcinoma, to examine whether matrix metalloproteinases (MMPs)--a family of extracellular proteases implicated in multiple steps of cancer progression--contributed to Wnt1-induced tumorigenesis. An analysis of the expression of several MMPs by RT-PCR and in situ hybridization revealed an increase in the expression of MMP-2, MMP-3, MMP-9, MMP-13, and MT1-MMP (MMP-14) in hyperplastic glands and in mammary tumors of MMTV-Wnt1 transgenic mice. Interestingly, whereas MMP-2, MMP-3, and MMP-9 were exclusively expressed by stromal cells in mammary tumors, MMP-13 and MT1-MMP were expressed by transformed epithelial cells in addition to the tumor stroma. To determine whether these MMPs contributed to tumorigenesis, MMTV-Wnt1 mice were crossed with transgenic mice overexpressing tissue inhibitor of metalloproteinase-2-a natural MMP inhibitor-in the mammary gland. In the double MMTV-Wnt1/tissue inhibitor of metalloproteinases-2 transgenic mice, we observed an increase in tumor latency and a 26.3% reduction in tumor formation. Furthermore, these tumors grew at a slower rate, exhibited an 18% decrease in proliferative rate, and a 12.2% increase in apoptotic rate of the tumor cells in association with a deficit in angiogenesis when compared with tumors from MMTV-Wnt1 mice. Thus, for the first time, the data provides evidence for the active role of MMPs in Wnt1-induced mammary tumorigenesis.


Assuntos
Neoplasias Mamárias Experimentais/enzimologia , Metaloproteinases da Matriz/biossíntese , Proteína Wnt1/biossíntese , Animais , Apoptose/fisiologia , Processos de Crescimento Celular/fisiologia , Feminino , Isoenzimas/biossíntese , Glândulas Mamárias Animais/enzimologia , Neoplasias Mamárias Experimentais/irrigação sanguínea , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/patologia , Vírus do Tumor Mamário do Camundongo/genética , Camundongos , Camundongos Transgênicos , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Regiões Promotoras Genéticas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Inibidor Tecidual de Metaloproteinase-2/biossíntese , Transgenes , Proteína Wnt1/genética
15.
Cancer Res ; 65(4): 1129-35, 2005 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-15734993

RESUMO

The bone is the third most common site of cancer metastasis. To invade the bone, tumor cells produce osteoclast-activating factors that increase bone resorption by osteoclasts. Here we report that human neuroblastoma cells that form osteolytic lesions in vivo do not produce osteoclast-activating factors but rather stimulate osteoclast activity in the presence of human bone marrow mesenchymal stem cells. This alternative pathway of osteoclast activation involves a nonadhesive interaction between neuroblastoma cells and bone marrow mesenchymal stem cells. Stimulated bone marrow mesenchymal stem cells express markedly increased levels of interleukin-6, which is then responsible for osteoclast activation. This report describes a critical role of bone marrow mesenchymal stem cells in bone destruction in cancer.


Assuntos
Células da Medula Óssea/patologia , Neoplasias Ósseas/secundário , Células-Tronco Mesenquimais/patologia , Neuroblastoma/patologia , Osteoclastos/patologia , Animais , Células da Medula Óssea/metabolismo , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Comunicação Celular , Linhagem Celular Tumoral , Humanos , Linfocinas/biossíntese , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Nus , Neuroblastoma/metabolismo , Osteoclastos/metabolismo
16.
Cancer Res ; 65(8): 3200-8, 2005 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15833851

RESUMO

The contribution of the tumor stroma to cancer progression has been increasingly recognized. We had previously shown that in human neuroblastoma tumors orthotopically implanted in immunodeficient mice, stromal-derived matrix metalloproteinase-9 (MMP-9) contributes to the formation of a mature vasculature by promoting pericyte recruitment along endothelial cells. Here we show that MMP-9 is predominantly expressed by bone marrow-derived CD45-positive leukocytes. Using a series of bone marrow transplantation experiments in MMP-9(+/+) and MMP-9(-/-) mice xenotransplanted with human neuroblastoma tumors, we show that bone marrow-derived MMP-9 is critical for the recruitment of leukocytes from bone marrow into the tumor stroma and for the integration of bone marrow-derived endothelial cells into the tumor vasculature. Expression of MMP-9 by bone marrow-derived cells in the tumor stroma is also critical for the formation of a mature vasculature and coverage of endothelial cells with pericytes. Furthermore, in primary human neuroblastoma tumor specimens of unfavorable histology, we observed a higher level of tumor infiltration with MMP-9 expressing phagocytic cells and a higher degree of coverage of endothelial cells by pericytes when compared with tumor specimens with a favorable histology. Taken together, the data show that in neuroblastoma, MMP-9 plays a critical role in the recruitment of bone marrow-derived cells to the tumor microenvironment where they positively contribute to angiogenesis and tumor progression.


Assuntos
Células da Medula Óssea/enzimologia , Metaloproteinase 9 da Matriz/biossíntese , Neovascularização Patológica/enzimologia , Neuroblastoma/irrigação sanguínea , Adolescente , Animais , Células da Medula Óssea/patologia , Transplante de Medula Óssea , Linhagem Celular Tumoral , Criança , Pré-Escolar , Humanos , Masculino , Metaloproteinase 9 da Matriz/deficiência , Metaloproteinase 9 da Matriz/genética , Camundongos , Camundongos Knockout , Transplante de Neoplasias , Neovascularização Patológica/patologia , Neuroblastoma/enzimologia , Neuroblastoma/patologia , Transplante Heterólogo
17.
J Vis Exp ; (129)2017 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-29286360

RESUMO

siRNA and shRNA-mediated knock down (KD) methods of regulating gene expression are invaluable tools for understanding gene and protein function. However, in the case that the KD of the protein of interest has a lethal effect on cells or the anticipated effect of the KD is time-dependent, unconditional KD methods are not appropriate. Conditional systems are more suitable in these cases and have been the subject of much interest. These include Ecdysone-inducible overexpression systems, Cytochrome P-450 induction system1, and the tetracycline regulated gene expression systems. The tetracycline regulated gene expression system enables reversible control over protein expression by induction of shRNA expression in the presence of tetracycline. In this protocol, we present an experimental design using functional Tet-ON system in human cancer cell lines for conditional regulation of gene expression. We then demonstrate the use of this system in the study of tumor cell-monocyte interaction.


Assuntos
Neoplasias da Mama/genética , Neoplasias do Colo/genética , Técnicas de Silenciamento de Genes/métodos , Macrófagos/patologia , Monócitos/patologia , Animais , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Neoplasias do Colo/patologia , Feminino , Células HCT116 , Humanos , RNA Interferente Pequeno/genética , Microambiente Tumoral
18.
Cancer Res ; 77(5): 1051-1059, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28209610

RESUMO

Over the past 10 years, the Tumor Microenvironment Network (TMEN), supported by the NCI (Bethesda, MD), has promoted collaborative research with the explicit goal of fostering multi-institutional and transdisciplinary groups that are capable of addressing complex issues involving the tumor microenvironment. The main goal of the TMEN was to generate novel information about the dynamic complexity of tumor-host interactions in different organ systems with emphasis on using human tissues and supplemented by experimental models. As this initiative comes to a close, members of the TMEN took time to examine what has been accomplished by the Network and importantly to identify the challenges and opportunities ahead. This consensus document summarizes for the broader scientific community discussions that occurred at the two final meetings of the TMEN in 2015 and 2016. Cancer Res; 77(5); 1051-9. ©2017 AACR.


Assuntos
Neoplasias/patologia , Microambiente Tumoral/fisiologia , Humanos
19.
J Extracell Vesicles ; 6(1): 1332941, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28717423

RESUMO

The bone marrow (BM) niche is a microenvironment promoting survival, dormancy and therapeutic resistance in tumor cells. Central to this function are mesenchymal stromal cells (MSCs). Here, using neuroblastoma (NB) as a model, we demonstrate that NB cells release an extracellular vesicle (EVs) whose protein cargo is enriched in exosomal proteins but lacks cytokines and chemokines. Using three different purification methods, we then demonstrate that NB-derived exosomes were captured by MSCs and induced the production of pro-tumorigenic cytokines and chemokines, including interleukin-6 (IL-6), IL-8/CXCL8, vascular endothelial cell growth factor and monocyte-chemotactic protein-1, with exosomes prepared by size exclusion chromatography having the highest activity. We found no correlation between the IL-6 and IL-8/CXCL8 stimulatory activity of exosomes from eight NB cell lines and their origin, degree of MYCN amplification, drug resistance and disease status. We then demonstrate that the uptake of NB exosomes by MSCs was associated with a rapid increase in ERK1/2 and AKT activation, and that blocking ERK1/2 but not AKT activation inhibited the IL-6 and IL-8/CXCL8 production by MSCs without affecting exosome uptake. Thus, we describe a new mechanism by which NB cells induce in MSCs an inflammatory reaction that contributes to a favorable microenvironment in the BM.

20.
Mol Cancer Ther ; 16(11): 2516-2527, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28716816

RESUMO

Drug resistance is a major barrier for the development of effective and durable cancer therapies. Overcoming this challenge requires further defining the cellular and molecular mechanisms underlying drug resistance, both acquired and environment-mediated drug resistance (EMDR). Here, using neuroblastoma (NB), a childhood cancer with high incidence of recurrence due to resistance to chemotherapy, as a model we show that human bone marrow-mesenchymal stromal cells induce tumor expression of sphingosine-1-phosphate receptor-1 (S1PR1), leading to their resistance to chemotherapy. Targeting S1PR1 by shRNA markedly enhances etoposide-induced apoptosis in NB cells and abrogates EMDR, while overexpression of S1PR1 significantly protects NB cells from multidrug-induced apoptosis via activating JAK-STAT3 signaling. Elevated S1PR1 expression and STAT3 activation are also observed in human NB cells with acquired resistance to etoposide. We show in vitro and in human NB xenograft models that treatment with FTY720, an FDA-approved drug and antagonist of S1PR1, dramatically sensitizes drug-resistant cells to etoposide. In summary, we identify S1PR1 as a critical target for reducing both EMDR and acquired chemoresistance in NB. Mol Cancer Ther; 16(11); 2516-27. ©2017 AACR.


Assuntos
Etoposídeo/administração & dosagem , Interação Gene-Ambiente , Neuroblastoma/tratamento farmacológico , Receptores de Lisoesfingolipídeo/genética , Fator de Transcrição STAT3/genética , Animais , Apoptose/efeitos dos fármacos , Células da Medula Óssea/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Cloridrato de Fingolimode/administração & dosagem , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Recidiva Local de Neoplasia/tratamento farmacológico , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/patologia , Neuroblastoma/genética , Neuroblastoma/patologia , RNA Interferente Pequeno , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Receptores de Esfingosina-1-Fosfato , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA