Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Cancer ; 127(8): 1246-1259, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33270904

RESUMO

BACKGROUND: CXCR4 mediates the retention and survival of acute myelogenous leukemia blasts in bone marrow and contributes to their resistance to chemotherapy. The authors evaluated a combination of the high-affinity CXCR4 antagonist BL-8040 with high-dose cytarabine (HiDAC) chemotherapy in a phase 2a study of patients with relapsed and refractory AML. METHODS: Forty-two patients received treatment with BL-8040 monotherapy for 2 days followed by a combination of BL-8040 with HiDAC for 5 days. Six escalating BL-8040 dose levels were investigated (0.5, 0.75, 1.0, 1.25, 1.5, and 2.0 mg/kg), and 1.5 mg/kg was selected as the dose for the expansion phase (n = 23). RESULTS: BL-8040 in combination with HiDAC was safe and well tolerated at all dose levels. Clinical response was observed with BL-8040 doses ≥1.0 mg/kg. The composite response rate (complete remissions plus complete remissions with incomplete hematologic recovery of platelets or neutrophils) was 29% (12 of 42) in all patients and 39% (9 of 23) in the 1.5-mg/kg phase. The median overall survival was 8.4 months for all patients, 10.8 months in the 1.5-mg/kg phase, and 21.8 months for responding patients in the 1.5-mg/kg cohort. Two days of BL-8040 monotherapy triggered the mobilization of blasts into peripheral blood, with significantly higher mean fold-changes in responders versus nonresponders. This was accompanied by a decrease in bone marrow blasts. CONCLUSIONS: The current results demonstrate the efficacy of CXCR4 targeting with BL-8040 and support continued clinical development in acute myelogenous leukemia.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Citarabina/administração & dosagem , Leucemia Mieloide Aguda/tratamento farmacológico , Peptídeos/administração & dosagem , Receptores CXCR4/antagonistas & inibidores , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Células da Medula Óssea/efeitos dos fármacos , Esquema de Medicação , Resistencia a Medicamentos Antineoplásicos , Feminino , Mobilização de Células-Tronco Hematopoéticas , Humanos , Leucemia Mieloide Aguda/sangue , Leucemia Mieloide Aguda/mortalidade , Masculino , Pessoa de Meia-Idade , Peptídeos/efeitos adversos , Peptídeos/farmacocinética , Recidiva , Indução de Remissão
2.
Cytokine ; 109: 11-16, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29903571

RESUMO

The chemokine receptor CXCR4 and its ligand stromal cell-derived factor-1 (SDF-1/CXCL12) are important players in the cross-talk among lymphoma, myeloma and leukemia cells and their microenvironments. In hematological malignancies and solid tumors, the overexpression of CXCR4 on the cell surface has been shown to be responsible for disease progression, increasing tumor cell survival and chemoresistance and metastasis to organs with high CXCL12 levels (e.g., lymph nodes and bone marrow (BM)). Furthermore, the overexpression of CXCR4 has been found to have prognostic significance for disease progression in many type of tumors including lymphoma, leukemia, glioma, and prostate, breast, colorectal, renal, and hepatocellular carcinomas. In leukemia, CXCR4 expression granted leukemic blasts a higher capacity to seed into BM niches, thereby protecting leukemic cells from chemotherapy-induced apoptosis, and was correlated with shorter disease-free survival. In contrast, neutralizing the interaction of CXCL12/CXCR4 with a variety of antagonists induced apoptosis and differentiation and increased the chemosensitivity of lymphoma, myeloma, and leukemia cells. The role of CXCL12 and CXCR4 in the pathogenesis of hematological malignancies and the clinical therapeutic potential of CXCR4 antagonists in these diseases is discussed.


Assuntos
Quimiocina CXCL12/metabolismo , Neoplasias Hematológicas/patologia , Leucemia Linfocítica Crônica de Células B/patologia , Leucemia Mieloide Aguda/patologia , Mieloma Múltiplo/patologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Receptores CXCR4/metabolismo , Apoptose/imunologia , Sobrevivência Celular/fisiologia , Progressão da Doença , Neoplasias Hematológicas/tratamento farmacológico , Humanos , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Prognóstico , Microambiente Tumoral/fisiologia
3.
Clin Immunol ; 183: 316-324, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28939272

RESUMO

Th-17 type immune response that occurs in multiple sclerosis (MS) is linked to CCR6-CCL20 interaction. We confirmed the dependency on CCR6 in EAE development. Vaccination of mice with hCCL20, but not mCCL20, produced anti-murine CCL20 and ameliorated EAE. The EAE clinical score negatively correlated with anti CCL20 levels. A beneficial effect was transferred by sera from hCCL20-immunized mice. Immunized mice with cyclic peptide that include a bacterial outer membrane protein A (ompA), that share homology sequence with hCCL20 produced anti CCL20, anti ompA and anti-cyclic peptide. Immunization of mice with ompA or the cyclic peptide ameliorated EAE. The cyclic peptide inhibited CCL20 activity in an adhesion assay. A significantly higher level of anti CCL20 were found in healthy individuals compared to RR-MS patients. There was no similar difference for anti-CXCL10. Natural or induced immunization against CCL20 confer protection against EAE and may be beneficial in MS.


Assuntos
Quimiocina CCL20/imunologia , Encefalite/prevenção & controle , Doença de Hashimoto/prevenção & controle , Esclerose Múltipla/prevenção & controle , Animais , Adesão Celular , Quimiocina CCL20/administração & dosagem , Biologia Computacional , Feminino , Imunização , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Receptores CCR6/genética , Receptores CCR6/metabolismo , Linfócitos T
4.
Hepatology ; 58(3): 1021-30, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23526353

RESUMO

UNLABELLED: Human hepatocellular carcinoma (HCC) is an inflammation-induced cancer, which is the third-leading cause of cancer mortality worldwide. We investigated the role of the chemokine receptors, CCR5 and CCR1, in regulating inflammation and tumorigenesis in an inflammation-induced HCC model in mice. Multidrug resistance 2 gene (Mdr2)-knockout (Mdr2-KO) mice spontaneously develop chronic cholestatic hepatitis and fibrosis that is eventually followed by HCC. We generated two new strains from the Mdr2-KO mouse, the Mdr2:CCR5 and the Mdr2:CCR1 double knockouts (DKOs), and set out to compare inflammation and tumorigenesis among these strains. We found that in Mdr2-KO mice lacking the chemokine receptor, CCR5 (Mdr2:CCR5 DKO mice), but not CCR1 (Mdr2:CCR1 DKO), macrophage recruitment and trafficking to the liver was significantly reduced. Furthermore, in the absence of CCR5, reduced inflammation was also associated with reduced periductal accumulation of CD24(+) oval cells and abrogation of fibrosis. DKO mice for Mdr2 and CCR5 exhibited a significant decrease in tumor incidence and size. CONCLUSIONS: Our results indicate that CCR5 has a critical role in both the development and progression of liver cancer. Therefore, we propose that a CCR5 antagonist can serve for HCC cancer prevention and treatment.


Assuntos
Carcinoma Hepatocelular/etiologia , Carcinoma Hepatocelular/fisiopatologia , Hepatite Crônica/complicações , Cirrose Hepática/complicações , Neoplasias Hepáticas/etiologia , Neoplasias Hepáticas/fisiopatologia , Receptores CCR5/fisiologia , Subfamília B de Transportador de Cassetes de Ligação de ATP/deficiência , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Subfamília B de Transportador de Cassetes de Ligação de ATP/fisiologia , Animais , Carcinoma Hepatocelular/epidemiologia , Quimiocina CCL5/fisiologia , Modelos Animais de Doenças , Progressão da Doença , Hepatite Crônica/genética , Incidência , Fígado/patologia , Fígado/fisiopatologia , Cirrose Hepática/genética , Neoplasias Hepáticas/epidemiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores CCR1/deficiência , Receptores CCR1/genética , Receptores CCR1/fisiologia , Receptores CCR5/deficiência , Receptores CCR5/genética , Membro 4 da Subfamília B de Transportadores de Cassetes de Ligação de ATP
5.
Br J Haematol ; 163(2): 248-59, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23906028

RESUMO

Platelets are the terminal differentiation product of megakaryocytes (MKs). Cytokines, such as thrombopoietin (TPO), are known to influence different steps in MK development; however, the complex differentiation and platelet localization processes are not fully understood. MKs express the receptor CXCR4 and have been shown to migrate in response to CXCL12 and to increase their platelet production. In this study, we studied the role of CXCR4 in platelet production with the high affinity CXCR4 antagonist, BKT140. Single and sequential administration of BKT140 significantly increased the number of MKs and haematopoietic progenitors (HPCs) within the bone marrow (BM). Increased megakaryopoiesis was associated with increased platelet production. Single and sequential administration of BKT140 also increased the number of HPCs in the blood. In a model of 5-fluorouracil-induced thrombocytopenia, BKT140 significantly reduced the severity and duration of thrombocytopenia and cytopenia when administered before and after chemotherapy. Our results demonstrated that the CXCR4 antagonist, BKT140, mediated unique beneficial effects by stimulating megakaryopoiesis and platelet production. These results provide evidence for the possible therapeutic use of BKT140 for modulating platelet numbers in thrombocytopenic conditions.


Assuntos
Plaquetas/citologia , Plaquetas/efeitos dos fármacos , Oligopeptídeos/administração & dosagem , Receptores CXCR4/antagonistas & inibidores , Trombopoese/efeitos dos fármacos , Trombopoese/fisiologia , Animais , Antineoplásicos/efeitos adversos , Plaquetas/metabolismo , Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Feminino , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Contagem de Leucócitos , Células Progenitoras de Megacariócitos/citologia , Células Progenitoras de Megacariócitos/efeitos dos fármacos , Camundongos , Contagem de Plaquetas , Trombocitopenia/induzido quimicamente , Trombocitopenia/tratamento farmacológico , Trombopoetina/farmacologia
6.
Nat Med ; 12(6): 627-35, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16715091

RESUMO

A major goal of immunotherapy for autoimmune diseases and transplantation is induction of regulatory T cells that mediate immunologic tolerance. The mucosal immune system is unique, as tolerance is preferentially induced after exposure to antigen, and induction of regulatory T cells is a primary mechanism of oral tolerance. Parenteral administration of CD3-specific monoclonal antibody is an approved therapy for transplantation in humans and is effective in autoimmune diabetes. We found that orally administered CD3-specific antibody is biologically active in the gut and suppresses autoimmune encephalomyelitis both before induction of disease and at the height of disease. Orally administered CD3-specific antibody induces CD4+ CD25- LAP+ regulatory T cells that contain latency-associated peptide (LAP) on their surface and that function in vitro and in vivo through a TGF-beta-dependent mechanism. These findings identify a new immunologic approach that is widely applicable for the treatment of human autoimmune conditions.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Complexo CD3/imunologia , Linfócitos T CD4-Positivos/imunologia , Encefalomielite Autoimune Experimental/terapia , Peptídeos/imunologia , Receptores de Interleucina-2/imunologia , Subpopulações de Linfócitos T/imunologia , Administração Oral , Transferência Adotiva , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Linfócitos T CD4-Positivos/citologia , Cricetinae , Encefalomielite Autoimune Experimental/imunologia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Peptídeos/genética , Receptores de Interleucina-2/genética , Subpopulações de Linfócitos T/citologia , Fator de Crescimento Transformador beta/imunologia
7.
Biomedicines ; 11(2)2023 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-36830899

RESUMO

Given that, even after multimodal therapy, early-stage lung cancer (LC) often recurs, novel prognostic markers to help guide therapy are highly desired. The mRNA levels of cell division cycle 25C (CDC25C), a phosphatase that regulates G2/M cell cycle transition in malignant cells, correlate with poor clinical outcomes in lung adenocarcinoma (LUAD). However, whether CDC25C protein detected by immunohistochemistry can serve as a prognostic marker in LUAD is yet unknown. We stained an LC tissue array and a cohort of 61 LUAD tissue sections for CDC25C and searched for correlations between CDC25C staining score and the pathological characteristics of the tumors and the patients' clinical outcomes. Clinical data were retrieved from our prospectively maintained departmental database. We found that high expression of CDC25C was predominant among poorly differentiated LUAD (p < 0.001) and in LUAD > 1cm (p < 0.05). Further, high expression of CDC25C was associated with reduced disease-free survival (p = 0.03, median follow-up of 39 months) and with a trend for reduced overall survival (p = 0.08). Therefore, high expression of CDC25C protein in LUAD is associated with aggressive histological features and with poor outcomes. Larger studies are required to further validate CDC25C as a prognostic marker in LUAD.

8.
J Hematol Oncol ; 15(1): 144, 2022 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-36217194

RESUMO

BACKGROUND: We assessed the mechanism by which multiple myeloma (MM) shapes the bone marrow (BM) microenvironment and affects MΦ polarization. METHODS: In vivo xenograft model of BM-disseminated human myeloma, as well as analysis of MM cell lines, stromal components, and primary samples from patients with MM, was utilized. RESULTS: Analysis of the BM from MM-bearing mice inoculated with human CXCR4-expressing RPMI8226 cells revealed a significant increase in M2 MΦ cell numbers (p < 0.01). CXCL13 was one of the most profoundly increased factors upon MM growth with increased levels in the blood of MM-bearing animals. Myeloid cells were the main source of the increased murine CXCL13 detected in MM-infiltrated BM. MM cell lines induced CXCL13 and concurrent expression of M2 markers (MERTK, CD206, CD163) in co-cultured human MΦ in vitro. Interaction with MΦ reciprocally induced CXCL13 expression in MM cell lines. Mechanistically, TGFß signaling was involved in CXCL13 induction in MM cells, while BTK signaling was implicated in MM-stimulated increase of CXCL13 in MΦ. Recombinant CXCL13 increased RANKL expression and induced TRAP+ osteoclast (OC) formation in vitro, while CXCL13 neutralization blocked these activities. Moreover, mice inoculated with CXCL13-silenced MM cells developed significantly lower BM disease. Reduced tumor load correlated with decreased numbers of M2 MΦ in BM, decreased bone disease, and lower expression of OC-associated genes. Finally, higher levels of CXCL13 were detected in the blood and BM samples of MM patients in comparison with healthy individuals. CONCLUSIONS: Altogether, our findings suggest that bidirectional interactions of MΦ with MM tumor cells result in M2 MΦ polarization, CXCL13 induction, and subsequent OC activation, enhancing their ability to support bone resorption and MM progression. CXCL13 may thus serve as a potential novel target in MM.


Assuntos
Quimiocina CXCL13 , Macrófagos , Mieloma Múltiplo , Animais , Quimiocina CXCL13/metabolismo , Humanos , Macrófagos/metabolismo , Camundongos , Mieloma Múltiplo/patologia , Fator de Crescimento Transformador beta/metabolismo , Microambiente Tumoral , c-Mer Tirosina Quinase/metabolismo
9.
Cytokine ; 54(3): 249-57, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21376626

RESUMO

Chemokines were shown to govern the trafficking of immune cells and may also play important roles in the survival and activation of these cells. We report here that under physiological conditions, the bone marrow (BM), spleen, blood and liver of Ccr5, but not of Ccr1-deficient mice, contain reduced numbers of NK cells. NK cells in the BM of Ccr5-deficient mice proliferate to a lesser extent compared to WT mice. Furthermore, spleen NK cells derived from Ccr5-deficient mice that were transplanted into irradiated recipients failed to proliferate in the host. Ccr5, but not Ccr1-deficient NK cells, failed to migrate in vitro in response to RANTES and MIP-1ß but not MIP-1ß or SDF-1 and had reduced activation, lower expression levels of NK cell markers and a slightly reduced capacity to adhere to target cells and stimulate their killing. Using the polyI:C mouse model for NK trafficking, we found that in the absence of Ccr5, but not Ccr1, NK cells failed to accumulate in the liver. In contrast, using the influenza viral infection as a model to evaluate NK cell proliferation, we found that Ccr5-deficient NK cells in the BM had a higher proliferation rate than WT NK cells. These results suggest a role for Ccr5 in NK cell proliferation and circulation under physiological conditions and a complex role for Ccr5 in determining the fate of NK cells under pathological conditions.


Assuntos
Regulação da Expressão Gênica , Células Matadoras Naturais/metabolismo , Receptores CCR5/deficiência , Receptores CCR5/fisiologia , Animais , Apoptose , Movimento Celular , Proliferação de Células , Feminino , Interferon gama/metabolismo , Células Matadoras Naturais/citologia , Fígado/metabolismo , Linfócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Fenótipo , Poli I-C/metabolismo , Baço/metabolismo , Viroses/metabolismo
10.
Bone Marrow Transplant ; 55(7): 1305-1316, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32433499

RESUMO

Graft versus host disease (GvHD) remains a limiting factor for successful hematopoietic stem cell transplantation (HSCT). T cells and antigen-presenting cells (APCs) are major components of the hematopoietic G-CSF mobilized peripheral blood cell (MPBC) graft. Here we show that a short incubation (2 h) of MPBCs with hexameric Fas ligand (FasL) selectively induces apoptosis of specific donor T cell subsets and APCs but not of CD34+ cells. FasL treatment preferentially induces apoptosis in mature T cell subsets which express high levels of Fas (CD95), such as T stem cell memory, T central memory, and T effector memory cells, as well as TH1 and TH17 cells. Anti-CD3/CD28 stimulated T cells derived from FasL-treated-MPBCs express lower levels of CD25 and secrete lower levels of IFN-γ as compared to control cells not treated with FasL. FasL treatment also induces apoptosis of transitional, naïve, memory and plasmablastoid B cells leading to a reduction in their numbers in the graft and following engraftment in transplanted mice. Most importantly, ex vivo treatment of MPBCs with FasL prior to transplant in conditioned NOD-scid IL2Rγnull (NSG) mice prevented GvHD while preserving graft versus leukemia (GvL) effects, and leading to robust stem cell engraftment.


Assuntos
Doença Enxerto-Hospedeiro , Transplante de Células-Tronco Hematopoéticas , Animais , Proteína Ligante Fas , Doença Enxerto-Hospedeiro/prevenção & controle , Ligantes , Camundongos , Camundongos Endogâmicos NOD , Células-Tronco
11.
J Immunol Res ; 2019: 8535273, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31467935

RESUMO

BACKGROUND: Age-related macular degeneration (AMD), the most common cause of blindness in the developed world, usually affects individuals older than 60 years of age. The majority of visual loss in this disease is attributable to the development of choroidal neovascularization (CNV). Mononuclear phagocytes, including monocytes and their tissue descendants, macrophages, have long been implicated in the pathogenesis of neovascular AMD (nvAMD). Current therapies for nvAMD are based on targeting vascular endothelial growth factor (VEGF). This study is aimed at assessing if perturbation of chemokine signaling and mononuclear cell recruitment may serve as novel complementary therapeutic targets for nvAMD. METHODS: A promiscuous chemokine antagonist (BKT130), aflibercept treatment, or combined BKT130+aflibercept treatment was tested in an in vivo laser-induced model of choroidal neovascularization (LI-CNV) and in an ex vivo choroidal sprouting assay (CSA). Quantification of CD11b+ cell in the CNV area was performed, and mRNA levels of genes implicated in CNV growth were measured in the retina and RPE-choroid. RESULTS: BKT130 reduced the CNV area and recruitment of CD11b+ cells by 30-35%. No effect of BKT130 on macrophages' proangiogenic phenotype was demonstrated ex vivo, but a lower VEGFA and CCR2 expression was found in the RPE-choroid and a lower expression of TNFα and NOS1 was found in both RPE-choroid and retinal tissues in the LI-CNV model under treatment with BKT130. CONCLUSIONS: Targeting monocyte recruitment via perturbation of chemokine signaling can reduce the size of experimental CNV and should be evaluated as a potential novel therapeutic modality for nvAMD.


Assuntos
Quimiocinas/antagonistas & inibidores , Neovascularização de Coroide/tratamento farmacológico , Monócitos/efeitos dos fármacos , Idoso , Idoso de 80 Anos ou mais , Animais , Antígeno CD11b/metabolismo , Movimento Celular/efeitos dos fármacos , Quimiocinas/metabolismo , Neovascularização de Coroide/etiologia , Neovascularização de Coroide/metabolismo , Neovascularização de Coroide/patologia , Feminino , Humanos , Lasers , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Monócitos/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Ratos , Ratos Long-Evans , Receptores CCR2/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/uso terapêutico , Proteínas Recombinantes de Fusão/uso terapêutico , Retina/metabolismo , Retina/patologia , Fator de Necrose Tumoral alfa/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
12.
Biochem Pharmacol ; 168: 412-428, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31325448

RESUMO

Although having promising anti-myeloma properties, the pan-histone deacetylase inhibitor (HDACi) panobinostat lacks therapeutic activity as a single agent. The aim of the current study was to elucidate the mechanisms underlying multiple myeloma (MM) resistance to panobinostat monotherapy and to define strategies to overcome it. Sensitivity of MM cell lines and primary CD138+ cells from MM patients to panobinostat correlated with reduced expression of the chemokine receptor CXCR4, whereas overexpression of CXCR4 in MM cell lines increased their resistance to panobinostat. Decreased sensitivity to HDACi was associated with reversible G0/G1 cell growth arrest while response was characterized by apoptotic cell death. Analysis of intra-cellular signaling mediators revealed the pro-survival mTOR pathway to be regulated by CXCR4 overexpression. Combining panobinostat with mTOR inhibitor everolimus abrogated the resistance to HDACi and induced synergistic cell death. The combination of panobinostat/everolimus resulted in sustained DNA damage and irreversible suppression of proliferation accompanied by robust apoptosis. Gene expression analysis revealed distinct genetic profiles of single versus combined agent exposure. Whereas panobinostat increased the expression of the cell cycle inhibitor p21, co-treatment with everolimus abrogated the increase in p21 and synergistically downregulated the expression of DNA repair genes and mitotic checkpoint regulators. Importantly, the combination of panobinostat with everolimus effectively targeted CXCR4-expressing resistant MM cells in vivo in the BM niche. In summary, our results uncover the mechanism responsible for the strong synergistic anti-MM activity of dual HDAC and mTOR inhibition and provide the rationale for a novel potential therapeutic approach to treat MM.


Assuntos
Antineoplásicos/administração & dosagem , Everolimo/administração & dosagem , Inibidores de Histona Desacetilases/administração & dosagem , Mitose/efeitos dos fármacos , Panobinostat/administração & dosagem , Receptores CXCR4/antagonistas & inibidores , Proteínas rho de Ligação ao GTP/antagonistas & inibidores , Animais , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Relação Dose-Resposta a Droga , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/fisiologia , Humanos , Camundongos , Mitose/fisiologia , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/metabolismo , Receptores CXCR4/biossíntese , Receptores CXCR4/genética , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/biossíntese , Serina-Treonina Quinases TOR/genética , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Proteínas rho de Ligação ao GTP/biossíntese , Proteínas rho de Ligação ao GTP/genética
13.
Front Biosci ; 13: 6820-33, 2008 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-18508696

RESUMO

Stem cells are rare, pluripotent, self-renewing cells that give rise to all mature cells during development and adult life. Due to their proliferative capabilities and their ability to home and contribute to the regeneration of damage tissue, stem cells can be transformed into established tumors. Stem cells can function as a double-edged sword--they have the ability to circulate and migrate throughout the developing and mature adult organism, which is essential for their normal function; however, transformed stem cells are also endowed with the machinery to metastasize into various organs. Chemokine and chemokine receptors play a critical role in directing the trafficking of these cells. It is therefore evident that understanding the role of chemokines and their receptors in stem cell circulation is critical for the successful use of these cells in therapy for a wide variety of pathological conditions.


Assuntos
Quimiocinas/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Receptores de Quimiocinas/fisiologia , Células-Tronco/fisiologia , Animais , Neoplasias da Mama/fisiopatologia , Quimiocina CCL3/fisiologia , Quimiocina CXCL2/fisiologia , Desenvolvimento Embrionário , Feminino , Mobilização de Células-Tronco Hematopoéticas , Humanos , Interleucina-8/fisiologia , Linfócitos/fisiologia
14.
Stem Cells ; 25(9): 2158-66, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17525235

RESUMO

An increase in the number of stem cells in blood following mobilization is required to enhance engraftment after high-dose chemotherapy and improve transplantation outcome. Therefore, an approach that improves stem cell mobilization is essential. The interaction between CXCL12 and its receptor, CXCR4, is involved in the retention of stem cells in the bone marrow. Therefore, blocking CXCR4 may result in mobilization of hematopoietic progenitor and stem cells. We have found that the CXCR4 antagonist known as 4F-benzoyl-TN14003 (T-140) can induce mobilization of hematopoietic stem cells and progenitors within a few hours post-treatment in a dose-dependent manner. Furthermore, although T-140 can also increase the number of white blood cells (WBC) in blood, including monocytes, B cells, and T cells, it had no effect on mobilizing natural killer cells. T-140 was found to efficiently synergize with granulocyte colony-stimulating factor (G-CSF) in its ability to mobilize WBC and progenitors, as well as to induce a 660-fold increase in the number of erythroblasts in peripheral blood. Comparison between the CXCR4 antagonists T-140 and AMD3100 showed that T-140 with or without G-CSF was significantly more potent in its ability to mobilize hematopoietic stem cells and progenitors into blood. These results demonstrate that different CXCR4 antagonists may have different therapeutic potentials.


Assuntos
Mobilização de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Peptídeos/farmacologia , Receptores CXCR4/antagonistas & inibidores , Animais , Proliferação de Células/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Sinergismo Farmacológico , Feminino , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Fator Estimulador de Colônias de Granulócitos/farmacologia , Transplante de Células-Tronco Hematopoéticas , Leucócitos/citologia , Leucócitos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Peptídeos/administração & dosagem
15.
J Neurol Sci ; 274(1-2): 9-12, 2008 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18804221

RESUMO

One of the major goals for the immunotherapy of autoimmune diseases is the induction of regulatory T cells that mediate immunologic tolerance. Parenteral administration of anti-CD3 monoclonal antibody is an approved therapy for transplantation in humans and is effective in autoimmune diabetes. We have found that oral administration of anti-CD3 monoclonal antibody is biologically active in the gut and suppresses experimental autoimmune encephalomyelitis both prior to disease induction and at the height of disease. Oral anti-CD3 antibody acts by inducing a unique type of regulatory T cell characterized by latency-associated peptide (LAP) on its cell surface that functions in vivo and in vitro via TGF-beta dependent mechanism. Orally delivered antibody would not have side effects including cytokine release syndromes, thus oral anti-CD3 antibody is clinically applicable for chronic therapy. These findings identify a novel and powerful immunologic approach that is widely applicable for the treatment of human autoimmune conditions.


Assuntos
Anticorpos/administração & dosagem , Autoimunidade/efeitos dos fármacos , Complexo CD3/imunologia , Encefalomielite Autoimune Experimental/terapia , Administração Oral , Animais , Autoimunidade/imunologia , Diabetes Mellitus Tipo 1/terapia , Humanos , Camundongos
16.
Cancer Res ; 78(6): 1471-1483, 2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29259008

RESUMO

CXCR4 expression in neuroblastoma tumors correlates with disease severity. In this study, we describe mechanisms by which CXCR4 signaling controls neuroblastoma tumor growth and response to therapy. We found that overexpression of CXCR4 or stimulation with CXCL12 supports neuroblastoma tumorigenesis. Moreover, CXCR4 inhibition with the high-affinity CXCR4 antagonist BL-8040 prevented tumor growth and reduced survival of tumor cells. These effects were mediated by the upregulation of miR-15a/16-1, which resulted in downregulation of their target genes BCL-2 and cyclin D1, as well as inhibition of ERK. Overexpression of miR-15a/16-1 in cells increased cell death, whereas antagomirs to miR-15a/16-1 abolished the proapoptotic effects of BL-8040. CXCR4 overexpression also increased miR-15a/16-1, shifting their oncogenic dependency from the BCL-2 to the ERK signaling pathway. Overall, our results demonstrate the therapeutic potential of CXCR4 inhibition in neuroblastoma treatment and provide a rationale to test combination therapies employing CXCR4 and BCL-2 inhibitors to increase the efficacy of these agents.Significance: These results provide a mechanistic rationale for combination therapy of CXCR4 and BCL-2 inhibitors to treat a common and commonly aggressive pediatric cancer.Cancer Res; 78(6); 1471-83. ©2017 AACR.


Assuntos
Neoplasias Encefálicas/patologia , MicroRNAs/metabolismo , Neuroblastoma/patologia , Receptores CXCR4/metabolismo , Animais , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Linhagem Celular Tumoral , Ciclina D1/genética , Ciclina D1/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Camundongos , MicroRNAs/genética , Neuroblastoma/tratamento farmacológico , Neuroblastoma/genética , Peptídeos/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptores CXCR4/antagonistas & inibidores , Receptores CXCR4/genética , Ensaios Antitumorais Modelo de Xenoenxerto
17.
J Thorac Cardiovasc Surg ; 155(5): 2164-2175.e1, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29429629

RESUMO

OBJECTIVE: Past studies are inconsistent with regard to the role of matrix metalloproteinase 12 in lung tumorigenesis. This is due, in part, to differential tumorigenesis based on tumor-derived versus immune-derived matrix metalloproteinase 12 expression. Our study aims to thoroughly dissect the role of matrix metalloproteinase 12 in lung tumorigenesis. METHODS: We tested matrix metalloproteinase 12 expression and the association with prognosis using a tissue array and a published non-small cell lung cancer gene expression database. In addition, we characterized the contribution of matrix metalloproteinase 12 to tumor propagation in the lung using a series of in vitro and in vivo studies. RESULTS: Tumor cells of a diverse set of human lung cancers stained positive for matrix metalloproteinase 12, and high matrix metalloproteinase 12 mRNA levels in the tumor were associated with reduced survival. The lung microenvironment stimulated endogenous production of matrix metalloproteinase 12 in lung cancer cells (human 460 lung cancer cell line, Lewis lung carcinoma). In vitro, matrix metalloproteinase 12 knockout Lewis lung carcinoma and Lewis lung carcinoma cells had the same proliferation rate, but Lewis lung carcinoma showed increased invasiveness. In vivo, deficiency of matrix metalloproteinase 12 in Lewis lung carcinoma cells, but not in the host, reduced tumor growth and invasiveness. CONCLUSIONS: We suggest that tumor cell-derived matrix metalloproteinase 12 promotes tumor propagation in the lung and that in the context of pulmonary malignancies matrix metalloproteinase 12 should further be tested as a potential novel therapeutic target.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma Pulmonar de Lewis/enzimologia , Carcinoma Pulmonar de Células não Pequenas/enzimologia , Movimento Celular , Neoplasias Pulmonares/enzimologia , Metaloproteinase 12 da Matriz/metabolismo , Animais , Biomarcadores Tumorais/genética , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/patologia , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Feminino , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Masculino , Metaloproteinase 12 da Matriz/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Invasividade Neoplásica , Transdução de Sinais
18.
Front Immunol ; 8: 1432, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29218043

RESUMO

Chemokines and their receptors play critical roles in the progression of autoimmunity and inflammation. Typically, multiple chemokines are involved in the development of these pathologies. Indeed, targeting single chemokines or chemokine receptors has failed to achieve significant clinical benefits in treating autoimmunity and inflammation. Moreover, the binding of host atypical chemokine receptors to multiple chemokines as well as the binding of chemokine-binding proteins secreted by various pathogens can serve as a strategy for controlling inflammation. In this work, promiscuous chemokine-binding peptides that could bind and inhibit multiple inflammatory chemokines, such as CCL2, CCL5, and CXCL9/10/11, were selected from phage display libraries. These peptides were cloned into human mutated immunoglobulin Fc-protein fusions (peptibodies). The peptibodies BKT120Fc and BKT130Fc inhibited the ability of inflammatory chemokines to induce the adhesion and migration of immune cells. Furthermore, BKT120Fc and BKT130Fc also showed a significant inhibition of disease progression in a variety of animal models for autoimmunity and inflammation. Developing a novel class of antagonists that can control the courses of diseases by selectively blocking multiple chemokines could be a novel way of generating effective therapeutics.

19.
Clin Cancer Res ; 23(7): 1733-1747, 2017 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-27697999

RESUMO

Purpose: To explore the functional consequences of possible cross-talk between the CXCR4/CXCL12 and the sphingosine-1-phosphate (S1P) pathways in multiple myeloma (MM) cells and to evaluate the effect of S1P targeting with the FTY720 modulator as a potential anti-MM therapeutic strategy.Experimental Design and Results: S1P targeting with FTY720 induces MM cell apoptosis. The combination of FTY720 with the SPHK1 inhibitor SKI-II results in synergistic inhibition of MM growth. CXCR4/CXCL12-enhanced expression correlates with reduced MM cell sensitivity to both FTY720 and SKI-II inhibitors, and with SPHK1 coexpression in both cell lines and primary MM bone marrow (BM) samples, suggesting regulative cross-talk between the CXCR4/CXCL12 and SPHK1 pathways in MM cells. FTY720 was found to directly target CXCR4. FTY720 profoundly reduces CXCR4 cell-surface levels and abrogates the CXCR4-mediated functions of migration toward CXCL12 and signaling pathway activation. Moreover, FTY720 cooperates with bortezomib, inducing its cytotoxic activity and abrogating the bortezomib-mediated increase in CXCR4 expression. FTY720 effectively targets bortezomib-resistant cells and increases their sensitivity to bortezomib, promoting DNA damage. Finally, in a recently developed novel xenograft model of CXCR4-dependent systemic MM with BM involvement, FTY720 treatment effectively reduces tumor burden in the BM of MM-bearing mice. FTY720 in combination with bortezomib demonstrates superior tumor growth inhibition and abrogates bortezomib-induced CXCR4 increase on MM cells.Conclusions: Altogether, our work identifies a cross-talk between the S1P and CXCR4 pathways in MM cells and provides a preclinical rationale for the therapeutic application of FTY720 in combination with bortezomib in patients with MM. Clin Cancer Res; 23(7); 1733-47. ©2016 AACR.


Assuntos
Quimiocina CXCL12/genética , Mieloma Múltiplo/tratamento farmacológico , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Receptores CXCR4/genética , Animais , Apoptose/efeitos dos fármacos , Bortezomib/administração & dosagem , Dano ao DNA/efeitos dos fármacos , Sinergismo Farmacológico , Cloridrato de Fingolimode/administração & dosagem , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Lisofosfolipídeos/genética , Lisofosfolipídeos/metabolismo , Camundongos , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Esfingosina/análogos & derivados , Esfingosina/genética , Esfingosina/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Clin Cancer Res ; 23(22): 6790-6801, 2017 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-28835380

RESUMO

Purpose: The potential of the high-affinity CXCR4 antagonist BL-8040 as a monotherapy-mobilizing agent and its derived graft composition and quality were evaluated in a phase I clinical study in healthy volunteers (NCT02073019).Experimental Design: The first part of the study was a randomized, double-blind, placebo-controlled dose escalation phase. The second part of the study was an open-label phase, in which 8 subjects received a single injection of BL-8040 (1 mg/kg) and approximately 4 hours later underwent a standard leukapheresis procedure. The engraftment potential of the purified mobilized CD34+ cells was further evaluated by transplanting the cells into NSG immunodeficient mice.Results: BL-8040 was found safe and well tolerated at all doses tested (0.5-1 mg/kg). The main treatment-related adverse events were mild to moderate. Transient injection site and systemic reactions were mitigated by methylprednisolone, paracetamol, and promethazine pretreatment. In the first part of the study, BL-8040 triggered rapid and substantial mobilization of WBCs and CD34+ cells in all tested doses. Four hours postdose, the count rose to a mean of 8, 37, 31, and 35 cells/µL (placebo, 0.5, 0.75, and 1 mg/kg, respectively). FACS analysis revealed substantial mobilization of immature dendritic, T, B, and NK cells. In the second part, the mean CD34+ cells/kg collected were 11.6 × 106 cells/kg. The graft composition was rich in immune cells.Conclusions: The current data demonstrate that BL-8040 is a safe and effective monotherapy strategy for the collection of large amounts of CD34+ cells and immune cells in a one-day procedure for allogeneic HSPC transplantation. Clin Cancer Res; 23(22); 6790-801. ©2017 AACR.


Assuntos
Antígenos CD34/metabolismo , Mobilização de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Peptídeos/administração & dosagem , Receptores CXCR4/antagonistas & inibidores , Animais , Biomarcadores , Feminino , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Voluntários Saudáveis , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Humanos , Imunofenotipagem , Leucaférese , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Modelos Animais , Transplante Homólogo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa