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










Base de dados
Intervalo de ano de publicação
1.
Am J Obstet Gynecol ; 220(5): 498.e1-498.e9, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30849355

RESUMO

BACKGROUND: The transfer of pathogenic immunoglobulin G antibodies from mother to fetus is a critical step in the pathophysiology of alloimmune and autoimmune diseases of the fetus and neonate. Immunoglobulin G transfer across the human placenta to the fetus is mediated by the neonatal Fc receptor, and blockade of the neonatal Fc receptor may provide a therapeutic strategy to prevent or minimize pathological events associated with immune-mediated diseases of pregnancy. M281 is a fully human, aglycosylated monoclonal immunoglobulin G1 antineonatal Fc receptor antibody that has been shown to block the neonatal Fc receptor with high affinity in nonclinical studies and in a phase 1 study in healthy volunteers. OBJECTIVE: The objective of the study was to determine the transplacental transfer of M281 and its potential to inhibit transfer of immunoglobulin G from maternal to fetal circulation. STUDY DESIGN: To determine the concentration of M281 required for rapid cellular uptake and complete saturation of the neonatal Fc receptor in placental trophoblasts, primary human villous trophoblasts were incubated with various concentrations of M281 in a receptor occupancy assay. The placental transfer of M281, immunoglobulin G, and immunoglobulin G in the presence of M281 was studied using the dually perfused human placental lobule model. Immunoglobulin G transfer was established using a representative immunoglobulin G molecule, adalimumab, a human immunoglobulin G1 monoclonal antibody, at a concentration of 270 µg/mL. Inhibition of immunoglobulin G transfer by M281 was determined by cotransfusing 270 µg/mL of adalimumab with 10 µg/mL or 300 µg/mL of M281. Concentrations of adalimumab and M281 in sample aliquots from maternal and fetal circuits were analyzed using a sandwich enzyme-linked immunosorbent assay and Meso Scale Discovery assay, respectively. RESULTS: In primary human villous trophoblasts, the saturation of the neonatal Fc receptor by M281 was observed within 30-60 minutes at 0.15-5.0 µg/mL, suggesting rapid blockade of neonatal Fc receptor in placental cells. The transfer rate of adalimumab (0.23% ± 0.21%) across dually perfused human placental lobule was significantly decreased by 10 µg/mL and 300 µg/mL of M281 to 0.07 ± 0.01% and 0.06 ± 0.01%, respectively. Furthermore, the transfer rate of M281 was 0.002% ± 0.02%, approximately 100-fold lower than that of adalimumab. CONCLUSION: The significant inhibition of immunoglobulin G transfer across the human placental lobule by M281 and the minimal transfer of M281 supports the development of M281 as a novel agent for the treatment of fetal and neonatal diseases caused by transplacental transfer of alloimmune and autoimmune pathogenic immunoglobulin G antibodies.


Assuntos
Anticorpos Monoclonais/farmacologia , Imunoglobulina G/metabolismo , Troca Materno-Fetal/imunologia , Placenta/imunologia , Receptores Fc/imunologia , Adalimumab , Transporte Biológico , Feminino , Humanos , Imunoglobulina G/imunologia , Modelos Biológicos , Placenta/metabolismo , Gravidez , Trofoblastos/imunologia
2.
Mol Cancer Ther ; 18(2): 245-256, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30401693

RESUMO

Pancreatic cancer has an abysmal 5-year survival rate of 8%, making it a deadly disease with a need for novel therapies. Here we describe a multitargeting heparin-based mimetic, necuparanib, and its antitumor activity in both in vitro and in vivo models of pancreatic cancer. Necuparanib reduced tumor cell proliferation and invasion in a three-dimensional (3D) culture model; in vivo, it extended survival and reduced metastasis. Furthermore, proteomic analysis demonstrated that necuparanib altered the expression levels of multiple proteins involved in cancer-driving pathways including organ development, angiogenesis, proliferation, genomic stability, cellular energetics, and invasion and metastasis. One protein family known to be involved in invasion and metastasis and altered by necuparanib treatment was the matrix metalloprotease (MMP) family. Necuparanib reduced metalloproteinase 1 (MMP1) and increased tissue inhibitor of metalloproteinase 3 (TIMP3) protein levels and was found to increase RNA expression of TIMP3. MMP enzymatic activity was also found to be reduced in the 3D model. Finally, we confirmed necuparanib's in vivo activity by analyzing plasma samples of patients enrolled in a phase I/II study in patients with metastatic pancreatic cancer; treatment with necuparanib plus standard of care significantly increased TIMP3 plasma protein levels. Together, these results demonstrate necuparanib acts as a broad multitargeting therapeutic with in vitro and in vivo anti-invasive and antimetastatic activity.


Assuntos
Antineoplásicos/administração & dosagem , Heparitina Sulfato/análogos & derivados , Metaloproteinase 1 da Matriz/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Inibidor Tecidual de Metaloproteinase-3/metabolismo , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ensaios Clínicos Fase I como Assunto , Ensaios Clínicos Fase II como Assunto , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Heparitina Sulfato/administração & dosagem , Heparitina Sulfato/farmacologia , Humanos , Camundongos , Invasividade Neoplásica , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Proteômica/métodos , Esferoides Celulares/citologia , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/metabolismo , Células Estromais/efeitos dos fármacos , Inibidor Tecidual de Metaloproteinase-3/genética , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Proc Natl Acad Sci U S A ; 112(11): E1297-306, 2015 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-25733881

RESUMO

Despite the beneficial therapeutic effects of intravenous immunoglobulin (IVIg) in inflammatory diseases, consistent therapeutic efficacy and potency remain major limitations for patients and physicians using IVIg. These limitations have stimulated a desire to generate therapeutic alternatives that could leverage the broad mechanisms of action of IVIg while improving therapeutic consistency and potency. The identification of the important anti-inflammatory role of fragment crystallizable domain (Fc) sialylation has presented an opportunity to develop more potent Ig therapies. However, translating this concept to potent anti-inflammatory therapeutics has been hampered by the difficulty of generating suitable sialylated products for clinical use. Therefore, we set out to develop the first, to our knowledge, robust and scalable process for generating a well-qualified sialylated IVIg drug candidate with maximum Fc sialylation devoid of unwanted alterations to the IVIg mixture. Here, we describe a controlled enzymatic, scalable process to produce a tetra-Fc-sialylated (s4-IVIg) IVIg drug candidate and its qualification across a wide panel of analytic assays, including physicochemical, pharmacokinetic, biodistribution, and in vivo animal models of inflammation. Our in vivo characterization of this drug candidate revealed consistent, enhanced anti-inflammatory activity up to 10-fold higher than IVIg across different animal models. To our knowledge, this candidate represents the first s4-IVIg suitable for clinical use; it is also a valuable therapeutic alternative with more consistent and potent anti-inflammatory activity.


Assuntos
Anti-Inflamatórios/uso terapêutico , Desenho de Fármacos , Imunoglobulinas Intravenosas/uso terapêutico , Ácido N-Acetilneuramínico/metabolismo , Receptores Fc/metabolismo , Animais , Anti-Inflamatórios/farmacocinética , Anti-Inflamatórios/farmacologia , Artrite Experimental/tratamento farmacológico , Artrite Experimental/patologia , Vesícula/complicações , Vesícula/tratamento farmacológico , Vesícula/patologia , Modelos Animais de Doenças , Epidermólise Bolhosa Adquirida/complicações , Epidermólise Bolhosa Adquirida/tratamento farmacológico , Epidermólise Bolhosa Adquirida/patologia , Glicosilação/efeitos dos fármacos , Células HEK293 , Humanos , Fragmentos Fab das Imunoglobulinas/metabolismo , Imunoglobulinas Intravenosas/farmacocinética , Imunoglobulinas Intravenosas/farmacologia , Camundongos , Púrpura Trombocitopênica Idiopática/tratamento farmacológico , Púrpura Trombocitopênica Idiopática/patologia , Distribuição Tecidual/efeitos dos fármacos , Resultado do Tratamento
4.
PLoS One ; 6(6): e21106, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21698156

RESUMO

Heparan sulfate proteoglycans (HSPGs) play a key role in shaping the tumor microenvironment by presenting growth factors, cytokines, and other soluble factors that are critical for host cell recruitment and activation, as well as promoting tumor progression, metastasis, and survival. M402 is a rationally engineered, non-cytotoxic heparan sulfate (HS) mimetic, designed to inhibit multiple factors implicated in tumor-host cell interactions, including VEGF, FGF2, SDF-1α, P-selectin, and heparanase. A single s.c. dose of M402 effectively inhibited seeding of B16F10 murine melanoma cells to the lung in an experimental metastasis model. Fluorescent-labeled M402 demonstrated selective accumulation in the primary tumor. Immunohistological analyses of the primary tumor revealed a decrease in microvessel density in M402 treated animals, suggesting anti-angiogenesis to be one of the mechanisms involved in-vivo. M402 treatment also normalized circulating levels of myeloid derived suppressor cells in tumor bearing mice. Chronic administration of M402, alone or in combination with cisplatin or docetaxel, inhibited spontaneous metastasis and prolonged survival in an orthotopic 4T1 murine mammary carcinoma model. These data demonstrate that modulating HSPG biology represents a novel approach to target multiple factors involved in tumor progression and metastasis.


Assuntos
Progressão da Doença , Heparitina Sulfato/análogos & derivados , Heparitina Sulfato/farmacologia , Melanoma Experimental/patologia , Mimetismo Molecular , Metástase Neoplásica , Animais , Linhagem Celular Tumoral , Citometria de Fluxo , Melanoma Experimental/irrigação sanguínea , Camundongos , Ressonância de Plasmônio de Superfície
5.
PLoS One ; 5(2): e9364, 2010 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-20186331

RESUMO

Early detection of tumors can significantly improve the outcome of tumor treatment. One of the most frequently asked questions in cancer imaging is how many cells can be detected non-invasively in a live animal. Although many factors limit such detection, increasing the light emission from cells is one of the most effective ways of overcoming these limitations. Here, we describe development and utilization of a lentiviral vector containing enhanced firefly luciferase (luc2) gene. The resulting single cell clones of the mouse mammary gland tumor (4T1-luc2) showed stable light emission in the range of 10,000 photons/sec/cell. In some cases individual 4T1-luc2 cells inserted under the skin of a nu/nu mouse could be detected non-invasively using a cooled CCD camera in some cases. In addition, we showed that only few cells are needed to develop tumors in these mice and tumor progression can be monitored right after the cells are implanted. Significantly higher luciferase activity in these cells allowed us to detect micrometastases in both, syngeneic Balb/c and nu/nu mice.


Assuntos
Diagnóstico por Imagem/métodos , Luciferases/metabolismo , Medições Luminescentes/métodos , Neoplasias Mamárias Experimentais/metabolismo , Animais , Linhagem Celular Tumoral , Feminino , Vetores Genéticos/genética , Lentivirus/genética , Luciferases/genética , Medições Luminescentes/instrumentação , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/diagnóstico , Neoplasias Mamárias Experimentais/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Sensibilidade e Especificidade , Fatores de Tempo , Transfecção , Carga Tumoral
6.
J Med Chem ; 50(25): 6367-82, 2007 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-17994684

RESUMO

Indomethacin, a nonselective cyclooxygenase (COX) inhibitor, was modified in three distinct regions in an attempt both to increase cyclooxygenase-2 (COX-2) selectivity and to enhance drug safety by covalent attachment of an organic nitrate moiety as a nitric oxide donor. A human whole-blood COX assay shows the modifications on the 3-acetic acid part of the indomethacin yielding an amide-nitrate derivative 32 and a sulfonamide-nitrate derivative 61 conferred COX-2 selectivity. Along with their respective des-nitrate analogs, for example, 31 and 62, the nitrates 32 and 61 were effective antiinflammatory agents in the rat air-pouch model. After oral dosing, though, only 32 increased nitrate and nitrite levels in rat plasma, indicating that its nitrate tether served as a nitric oxide donor in vivo. In a rat gastric injury model, examples 31 and 32 both show a 98% reduction in gastric lesion score compared to that of indomethacin. In addition, the nitrated derivative 32 inducing 85% fewer gastric lesions when coadministered with aspirin as compared to the combination of aspirin and valdecoxib.


Assuntos
Inibidores de Ciclo-Oxigenase 2/síntese química , Indometacina/análogos & derivados , Indometacina/síntese química , Doadores de Óxido Nítrico/síntese química , Animais , Aspirina/efeitos adversos , Celecoxib , Inibidores de Ciclo-Oxigenase 2/efeitos adversos , Inibidores de Ciclo-Oxigenase 2/farmacologia , Desenho de Fármacos , Sinergismo Farmacológico , Feminino , Mucosa Gástrica/patologia , Humanos , Ácidos Hidroxâmicos/efeitos adversos , Ácidos Hidroxâmicos/síntese química , Ácidos Hidroxâmicos/farmacologia , Indometacina/efeitos adversos , Indometacina/farmacologia , Masculino , Doadores de Óxido Nítrico/efeitos adversos , Doadores de Óxido Nítrico/farmacologia , Pirazóis/farmacologia , Ratos , Ratos Sprague-Dawley , Úlcera Gástrica/induzido quimicamente , Úlcera Gástrica/patologia , Relação Estrutura-Atividade , Sulfonamidas/farmacologia
7.
Inflammopharmacology ; 12(5-6): 521-34, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16259719

RESUMO

Non-steroidal anti-inflammatory drugs (NSAIDs) are widely used to treat inflammation and to provide pain relief but suffer from a major liability concerning their propensity to cause gastric damage. As nitric oxide (NO) is known to be gastro-protective we have synthesized a NO-donating prodrug of naproxen named NMI-1182. We evaluated two cyclo-oxygenase (COX)-inhibiting nitric oxide donors (CINODs), NMI-1182 and AZD3582, for their ability to be gastro-protective compared to naproxen and for their anti-inflammatory activity. NMI-1182 and AZD3582 were found to produce similar inhibition of COX activity to that produced by naproxen. Both NMI-1182 and AZD3582 produced significantly less gastric lesions after oral administration than naproxen. All three compounds effectively inhibited paw swelling in the rat carrageenan paw edema model. In the carrageenan air pouch model all three compounds significantly reduced PGE2 levels in the pouch exudate but only NMI-1182 and naproxen inhibited leukocyte influx. These data demonstrate that NMI-1182 has comparable anti-inflammatory activity to naproxen but with a much reduced likelihood to cause gastric damage.


Assuntos
Inibidores de Ciclo-Oxigenase/farmacologia , Naftalenos/farmacologia , Doadores de Óxido Nítrico/farmacologia , Substâncias Protetoras/farmacologia , Animais , Anti-Inflamatórios/sangue , Anti-Inflamatórios/química , Anti-Inflamatórios/farmacologia , Aorta Abdominal/efeitos dos fármacos , Aorta Abdominal/fisiologia , Carragenina , Ciclo-Oxigenase 1/sangue , Ciclo-Oxigenase 2/sangue , Inibidores de Ciclo-Oxigenase/sangue , Inibidores de Ciclo-Oxigenase/química , Dinoprostona/antagonistas & inibidores , Dinoprostona/metabolismo , Relação Dose-Resposta a Droga , Edema/induzido quimicamente , Edema/prevenção & controle , Mucosa Gástrica/efeitos dos fármacos , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patologia , Humanos , Técnicas In Vitro , Inflamação/induzido quimicamente , Inflamação/prevenção & controle , Masculino , Estrutura Molecular , Naftalenos/sangue , Naftalenos/química , Naproxeno/sangue , Naproxeno/química , Naproxeno/farmacologia , Infiltração de Neutrófilos/efeitos dos fármacos , Doadores de Óxido Nítrico/sangue , Doadores de Óxido Nítrico/química , Substâncias Protetoras/química , Ratos , Ratos Sprague-Dawley , Vasodilatação/efeitos dos fármacos
8.
Biochem Pharmacol ; 70(9): 1343-51, 2005 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16168964

RESUMO

Cyclooxygenase (COX, EC 1.14.99.1) inhibitor-nitric oxide (NO) donor (CINOD) hybrid compounds represent an attractive alternative to NSAID and coxib therapy. This report compares two CINODs, NMI-1182 (naproxen-glyceryl dinitrate) and AZD3582 (naproxen-n-butyl nitrate), for their ability to inhibit COX-1 and -2, deliver bioavailable nitric oxide, and release naproxen, using in vitro biochemical and pharmacological methods. In human whole blood, both CINODs showed inhibition, comparable to naproxen, of both COX isozymes and slowly released naproxen. Both CINODs donated bioavailable NO, as detected by cGMP induction in the pig kidney transformed cell line, LLC-PK1, but NMI-1182 was more potent by 30-100 times than AZD3582, GTN, GDN, and ISDN and considerably faster in inducing cGMP synthesis than AZD3582. The nitrate groups of GTN, NMI-1182, and AZD3582 appeared to be bioactivated via a common pathway, since each compound desensitized LLC-PK1 cells to subsequent challenge with the other compounds. Similar cGMP induction also occurred in normal, untransformed cells (human renal proximal tubule epithelial cells and hepatocytes from man, rat, and monkey); again, NMI-1182 was superior to AZD3582. NMI-1182 was also the more metabolically labile compound, releasing more absolute nitrate and nitrite (total NO(x)) in human stomach (in which NO is salutary) and liver S9 homogenates. Naproxen was also more rapidly freed from NMI-1182 than AZD3582 in human stomach, although liver S9 hydrolyzed both CINODs with similar rates. These in vitro tests revealed that NMI-1182 may be a better CINOD than AZD3582 because of its superior NO donating and naproxen liberating properties.


Assuntos
Inibidores de Ciclo-Oxigenase/farmacologia , Naftalenos/farmacologia , Doadores de Óxido Nítrico/farmacologia , GMP Cíclico/biossíntese , Ciclo-Oxigenase 1/efeitos dos fármacos , Ciclo-Oxigenase 2/efeitos dos fármacos , Humanos , Fígado/metabolismo , Naftalenos/farmacocinética , Naproxeno/farmacologia , Óxido Nítrico/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...