Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Pharm Biol ; 60(1): 2253-2265, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36428240

RESUMO

CONTEXT: Rongjin Niantong Fang (RJNTF) is a Traditional Chinese Medicine formulation with a good therapeutic effect on osteoarthritis (OA). However, the underlying mechanisms remain unclear. OBJECTIVE: This study investigates whether RJNTF could delay OA cartilage degeneration by regulating the SDF-1/CXCR4-p38MAPK signalling pathway. MATERIALS AND METHODS: The Sprague-Dawley (SD) rats were used to establish the OA model by a modified Hulth's method. SD rats were divided into three groups (n = 10): blank group, model group (0.9% saline, 10 mL/kg/day), and treatment group (RJNTF, 4.5 g/kg/day). After 12 weeks of treatment, each group was analysed by H&E, Safranine-O solid green, ELISA, Immunohistochemistry, and Western blot. An in vitro model was induced with 100 ng/mL SDF-1 by ELISA, the blank group, model group, RJNTF group, and inhibitor group with intervention for 12 h, each group was analysed by Immunofluorescence staining and Western blot. RESULTS: SDF-1 content in the synovium was reduced in RJNTF treatment group compared to non-treatment model group (788.10 vs. 867.32 pg/mL) and down-regulation of CXCR4, MMP-3, MMP-9, MMP-13 protein expression, along with p38 protein phosphorylated were observed in RJNTF treatment group. In vitro results showed that RJNTF (IC50 = 8.925 mg/mL) intervention could down-regulate SDF-1 induced CXCR4 and p38 protein phosphorylated and reduce the synthesis of MMP-3, MMP-9, and MMP-13 proteins of chondrocytes from SD rat cartilage tissues. DISCUSSION AND CONCLUSION: RJNTF alleviates OA cartilage damage by SDF-1/CXCR4-p38MAPK signalling pathway inhibition. Our ongoing research focuses on Whether RJNTF treats OA through alternative pathways.


Assuntos
Cartilagem Articular , Osteoartrite , Ratos , Animais , Metaloproteinase 3 da Matriz/metabolismo , Metaloproteinase 3 da Matriz/farmacologia , Metaloproteinase 3 da Matriz/uso terapêutico , Metaloproteinase 9 da Matriz/metabolismo , Metaloproteinase 13 da Matriz , Ratos Sprague-Dawley , Osteoartrite/tratamento farmacológico , Receptores CXCR4/metabolismo , Receptores CXCR4/uso terapêutico
2.
ACS Biomater Sci Eng ; 8(10): 4486-4496, 2022 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-36178141

RESUMO

Stromal cell-derived factor-1 alpha (SDF-1α, CXCL12) mediates the migration of circulating cells to desired sites for tissue development, homeostasis, and regeneration and can be used to promote cardiac regeneration by recruiting stem cells. However, the use of SDF-1α in the injured heart necessitates not only higher binding affinity to its receptor, CXCR4+, but also better robustness against enzymatic degradation than other SDF-1 isoforms. Here, we conduct a screening of SDF-1α analog peptides that were designed by structure-based drug design (SBDD), a type of computer-aided drug design (CADD). We have developed in vitro and in vivo methods that enable us to estimate the effect of peptides on the migration of human mesenchymal stem cells (hMSCs) and cardiac regeneration in acute myocardial infarction (AMI)-induced animals, respectively. We demonstrate that one type of SDF-1α analog peptide, SDP-4, among the four analog peptides preselected by SBDD, is more potent than native SDF-1α for cardiac regeneration in myocardial infarction. It is interesting to note that the migratory effects of SDP-4 determined by a wound healing assay, a Transwell assay, and a 2D migration assay are comparable to those of SDF-1α. These results suggest that in vivo, as well as in vitro, screening of peptides developed by SBDD is a quintessential process to the development of a novel therapeutic compound for cardiac regeneration. Our finding also has an implication that the SDP-4 peptide is an excellent candidate for use in the regeneration of an AMI heart.


Assuntos
Quimiocina CXCL12 , Infarto do Miocárdio , Animais , Movimento Celular , Quimiocina CXCL12/química , Quimiocina CXCL12/farmacologia , Quimiocina CXCL12/uso terapêutico , Desenho de Fármacos , Humanos , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/metabolismo , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Receptores CXCR4/metabolismo , Receptores CXCR4/uso terapêutico
3.
Drug Deliv ; 29(1): 1384-1397, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35532120

RESUMO

Colorectal cancer (CRC) remains the third cause of cancer-related mortality in Western countries, metastases are the main cause of death. CRC treatment remains limited by systemic toxicity and chemotherapy resistance. Therefore, nanoparticle-mediated delivery of cytotoxic agents selectively to cancer cells represents an efficient strategy to increase the therapeutic index and overcome drug resistance. We have developed the T22-PE24-H6 therapeutic protein-only nanoparticle that incorporates the exotoxin A from Pseudomonas aeruginosa to selectively target CRC cells because of its multivalent ligand display that triggers a high selectivity interaction with the CXCR4 receptor overexpressed on the surface of CRC stem cells. We here observed a CXCR4-dependent cytotoxic effect for T22-PE24-H6, which was not mediated by apoptosis, but instead capable of inducing a time-dependent and sequential activation of pyroptotic markers in CRC cells in vitro. Next, we demonstrated that repeated doses of T22-PE24-H6 inhibit tumor growth in a subcutaneous CXCR4+ CRC model, also through pyroptotic activation. Most importantly, this nanoparticle also blocked the development of lymphatic and hematogenous metastases, in a highly aggressive CXCR4+ SW1417 orthotopic CRC model, in the absence of systemic toxicity. This targeted drug delivery approach supports for the first time the clinical relevance of inducing GSDMD-dependent pyroptosis, a cell death mechanism alternative to apoptosis, in CRC models, leading to the selective elimination of CXCR4+ cancer stem cells, which are associated with resistance, metastases and anti-apoptotic upregulation.


Assuntos
Antineoplásicos , Neoplasias Colorretais , Proteínas de Ligação a Fosfato , Proteínas Citotóxicas Formadoras de Poros , Piroptose , Receptores CXCR4 , Antineoplásicos/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Humanos , Metástase Neoplásica/prevenção & controle , Proteínas de Ligação a Fosfato/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Receptores CXCR4/metabolismo , Receptores CXCR4/uso terapêutico , Transdução de Sinais
4.
Skin Therapy Lett ; 27(2): 1-5, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35385630

RESUMO

Warts, Hypogammaglobulinemia, Infections and Myelokathexis (WHIM) is a primary immunodeficiency syndrome. Patients with WHIM syndrome are more susceptible to human papillomavirus (HPV) infections and commonly present to a dermatologist with recalcitrant to treatment warts. Other cardinal features of WHIM syndrome include recurrent sinopulmonary bacterial infections, neutropenia/lymphopenia, low levels of immunoglobulins (IgG, IgA, IgM) and myelokathexis. Research demonstrated that truncating gain-of-function mutations of the C-X-C chemokine receptor type 4 gene (CXCR4) are responsible for this disease. Plerixafor, a specific small molecule antagonist of CXCR4, is currently used for peripheral blood hematopoietic stem cell (HSC) mobilization in stem cell transplant recipients. It has recently shown promise for the treatment of WHIM syndrome in phase I/II clinical trials. In this paper we review the emerging patient clinical data for this medication and highlight the role of CXCR4 in other important skin diseases including keratinocyte carcinomas, psoriasis and cutaneous T-cell lymphoma.


Assuntos
Agamaglobulinemia , Compostos Heterocíclicos , Neutropenia , Infecções por Papillomavirus , Verrugas , Agamaglobulinemia/tratamento farmacológico , Benzilaminas , Ciclamos , Fantasia , Mobilização de Células-Tronco Hematopoéticas , Compostos Heterocíclicos/farmacologia , Compostos Heterocíclicos/uso terapêutico , Humanos , Neutropenia/tratamento farmacológico , Doenças da Imunodeficiência Primária , Receptores CXCR4/uso terapêutico , Síndrome , Verrugas/tratamento farmacológico , Verrugas/patologia
5.
Neurotherapeutics ; 19(2): 616-634, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35267171

RESUMO

Temozolomide (TMZ) monotherapy is known to be insufficient for resistant/relapsed glioblastoma (GBM), thus seeking a sensitization agent for TMZ is necessary. It was found that regorafenib may improve the overall survival of relapsed GBM patients. We aimed to discover whether regorafenib can enhance the anti-GBM effects of TMZ, and elucidate underlying mechanism. Our analysis of The Cancer Genome Atlas database revealed that the increased expression of CXCR4 is linked to poor survival of GBM patients. Additionally, TMZ treatment may trigger CXCR4/CXCL12 axis of GBM. We used two GBM cell lines, two primary GBM cells, and animal model to identify underlying mechanism and treatment efficacy of regorafenib combined with TMZ by cytotoxicity, apoptosis, reporter gene and invasion/migration assays, chemokine array, Western blotting, MRI, microarray, and immunohistochemistry. We observed that the chemokine CXCL-12 and its receptor CXCR4 regulate the resistance to TMZ, whereas the inhibition of CXCL-12/CXCR4 signaling sensitizes GBM cells to TMZ. The TMZ-induced CXCL-12/CXCR4 signaling, phosphor-extracellular signal-regulated kinases 1 and 2 (ERK1/2) and nuclear factor kappa light chain enhancer of activated B cells (NF-κB), and NF-κB-related proteins can effectively diminish when combining with regorafenib. Regorafenib significantly enhanced the TMZ-induced extrinsic/intrinsic apoptotic pathways, and facilitated the suppression of invasion and migration potential in GBM. Orthotopic tumor experiments demonstrated tumor size reduction and prolonged survival in combination group even with half-dose of TMZ. Our findings provide promising evidence that regorafenib may sensitize GBM to TMZ treatment through inhibition of the CXCL12/CXCR4/ERK/NF-κB signaling.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Compostos de Fenilureia , Piridinas , Animais , Apoptose , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Quimiocina CXCL12/farmacologia , Quimiocina CXCL12/uso terapêutico , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , NF-kappa B/metabolismo , Compostos de Fenilureia/farmacologia , Compostos de Fenilureia/uso terapêutico , Piridinas/farmacologia , Piridinas/uso terapêutico , Receptores CXCR4/uso terapêutico , Temozolomida/farmacologia , Temozolomida/uso terapêutico
6.
Int J Biol Sci ; 18(3): 1271-1287, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35173552

RESUMO

Rationale: Heart failure with preserved ejection fraction (HFpEF) can arise from hypertension-induced cardiac remodeling. Monocyte/macrophage accumulation and inflammation are crucial elements in the pathogenesis of hypertension-induced cardiac remodeling. The C-X-C chemokine receptor 4 (CXCR4) is a critical regulator of the macrophage-mediated immune response. Nevertheless, the contribution of CXCR4 to macrophage phenotype and function during the progression of HFpEF remains unclear. Herein, we aimed to determine the role of macrophagic CXCR4 in heart failure with preserved ejection fraction (HFpEF). Methods: As a HFpEF model, wild type mice and myeloid-specific CXCR4 deficiency mice were subjected to pressure overload for 30 days to assess the function of macrophagic CXCR4 on cardiac function. Medium from macrophages was used to treat cardiac fibroblasts to study macrophage-to-fibroblast signaling. Results: We found circulatory CXCR4+ immune cells, mainly monocytes, markedly increased in HFpEF patients with hypertension. In the experimental HFpEF mice model, macrophages but not neutrophils represent the main infiltrating inflammatory cells in the heart, abundantly expressing CXCR4. Myeloid-specific CXCR4 deficient impeded macrophage infiltration and inflammatory response in the heart of HFpEF mice, thus ameliorating cardiac fibrosis and improving cardiac diastolic function. Furthermore, transcriptomic profiling data revealed that CXCR4 loss in macrophages exhibited a decreased transcriptional signature associated with the regulation of inflammatory response. Notably, CXCR4 significantly augmented chemokine (C­X­C) motif ligand (CXCL3) expression, which at least partly contributed to fibrosis by promoting myofibroblast differentiation. Mechanistically, the increased production of pro-inflammatory cytokines in CXCR4 expressed macrophages could be attributed to the suppression of the peroxisome proliferator-activated receptor γ (PPARγ) activity. Conclusions: Collectively, our data supported that the infiltration of CXCR4+ macrophages in the heart exacerbates hypertension-induced diastolic function by promoting pro-inflammatory cytokines production and thus may serve as a potential therapeutic target for hypertension-induced HFpEF.


Assuntos
Cardiomiopatias , Insuficiência Cardíaca , Hipertensão , Animais , Citocinas , Fibroblastos/metabolismo , Hipertensão/complicações , Macrófagos/metabolismo , Camundongos , Receptores CXCR4/genética , Receptores CXCR4/uso terapêutico , Volume Sistólico/fisiologia , Função Ventricular Esquerda , Remodelação Ventricular/fisiologia
10.
Eur J Pharm Sci ; 157: 105606, 2021 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-33131745

RESUMO

Insufficient T cell infiltration in triple-negative breast cancer (TNBC) has limited its response rate to immune checkpoint blockade (ICB) therapies and motivated the development of immunostimulatory approaches to enhance the ICB therapy. CXCR4 is a chemokine receptor highly upregulated both on cell surface and cytoplasm in tumor tissues. Activating CXCR4 has been associated with increased immunosuppression in the tumor microenvironment. Here, we developed a CXCR4-targeted liposomal formulation (Liposomal-AMD3100) to enhance therapeutic efficacy of AMD3100, a CXCR4 antagonist. Particularly, AMD3100 is not only encapsulated into the liposome but coated on the surface of the formulation to serve as a targeting moiety and a dual blocker capable of inhibiting CXCR4 activation extracellularly and intracellularly. The Liposomal-AMD3100 remodeled both immune and stromal microenvironment more efficiently compared with free AMD3100, indicating better pharmacodynamic profile of AMD3100 achieved by liposomal formulation. The combination of anti-PD-L1 with Liposomal-AMD3100 formulation exhibited an increased antitumor effect and prolonged survival time compared with monotherapies in a murine TNBC model (4T1). This work proves that immune activation via liposomal delivery of CXCR4 inhibitors has a great potential to expand ICB therapies to originally ICB-insensitive cancer types.


Assuntos
Neoplasias de Mama Triplo Negativas , Animais , Quimiocina CXCL12/farmacologia , Quimiocina CXCL12/uso terapêutico , Humanos , Inibidores de Checkpoint Imunológico , Camundongos , Receptores CXCR4/uso terapêutico , Transdução de Sinais , Linfócitos T , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Microambiente Tumoral
11.
Cell Transplant ; 29: 963689720929992, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32452221

RESUMO

Mesenchymal stem cells (MSCs) are thought to have great potential in the therapy of acute liver injury. It is possible that these cells may be regulated by the stromal cell-derived factor-1 (SDF-1)/CXC chemokine receptor-4 (CXCR4) signaling axis, which has been shown to promote stem cells migration in the inflammation-associated diseases. However, the effects of SDF-1/CXCR4 axis on the MSCs-transplantation-based treatment for acute liver injury and the underlying mechanisms are largely unknown. In this study, we sought to determine whether SDF-1/CXCR4 would augment the therapeutic effect of bone marrow mesenchymal stem cells (BMSCs) by promoting their migration, which may result from activating the phosphoinositide 3-kinase (PI3K)/Akt signaling pathway, in a rat acute liver injury model induced by lipopolysaccharide (LPS). We found that BMSCs transplantation markedly attenuated liver injury and improved the survival of LPS-treated rats. Of interest, overexpression of CXCR4 in BMSCs could substantially promote their migration both in vitro and in vivo, and result in even better therapeutic effects. This might be attributed to the activation of PI3K/Akt signaling pathway in BMSCs that is downstream of CXCR4, as demonstrated by the use of the CXCR4 antagonist AMD3100 and PI3K pathway inhibitor LY294002 assays in vitro and in vivo. Together, our results unraveled a novel molecular mechanism for the therapeutic effect of BMSCs for the treatment of acute liver injury, which may shed a new light on the clinical application of BMSCs for acute liver failure.


Assuntos
Células da Medula Óssea/metabolismo , Transplante de Medula Óssea/métodos , Doença Hepática Induzida por Substâncias e Drogas/tratamento farmacológico , Lipopolissacarídeos/efeitos adversos , Células-Tronco Mesenquimais/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores CXCR4/uso terapêutico , Animais , Movimento Celular , Feminino , Ratos , Ratos Sprague-Dawley , Transdução de Sinais
12.
J Neurol Sci ; 414: 116837, 2020 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-32334273

RESUMO

Glioblastoma multiforme (GBM) is the most common primary malignancy in the adult central nervous, and is characterized by high aggressiveness and a high mortality rate. The high mortality rate is largely due to the development of drug resistance. Temozolomide (TMZ) resistance is considered to be one of the major reasons responsible for GBM therapy failure. CXCL12/CXCR4 has been demonstrated to be involved in cell proliferation, migration, invasion, angiogenesis, and radioresistance in GBM. However, its role in TMZ resistance in GBM is unknown. In this study, we aimed to evaluate the role of CXCL12/CXCR4 in mediating the TMZ resistance to GBM cells and explore the underlying mechanisms. We found that the CXCL12/CXCR4 axis enhanced TMZ resistance in GBM cells. Further study showed that CXCL12/CXCR4 conferred TMZ resistance and promoted the migration and invasion of GBM cells by up-regulating FOXM1. This resistance was partially reversed by suppressing CXCL12/CXCR4 and FOXM1 silencing. Our study revealed the vital role of CXCL12/CXCR4 in mediating the resistance of GBM cells to TMZ, and suggested that targeting CXCL12/CXCR4 axis may attenuate the resistance to TMZ in GBM.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Antineoplásicos Alquilantes/farmacologia , Antineoplásicos Alquilantes/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Linhagem Celular Tumoral , Quimiocina CXCL12/genética , Resistencia a Medicamentos Antineoplásicos/genética , Proteína Forkhead Box M1/genética , Proteína Forkhead Box M1/uso terapêutico , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Humanos , Receptores CXCR4/genética , Receptores CXCR4/uso terapêutico , Temozolomida/farmacologia , Temozolomida/uso terapêutico , Regulação para Cima
13.
Arch Med Res ; 51(2): 135-144, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32113784

RESUMO

BACKGROUND: Chronic myocardial infarction (CMI), represents a public health and a financial burden. Since stem cell transplant is used to regenerate cardiac tissue after acute myocardial infarction. AIM OF THE STUDY: To determine if autologous CXCR4 stem cells could restore damaged myocardial tissue in patients with CMI lesions. METHODS: 20 NYHA grade III male patients with CMI defined by clinical, biochemical, ECG and echocardiographic parameters were included. Patients were treated with G-CSF for 6 d before isolating their autologous stem cells from PBMCs. Cell phenotyping was done by cytofluorometry using monoclonal antibodies (anti-CXCR4, -CD34, -48, -117, -133, -Ki67, -SDF1 and CXCR4); CXCR4 cell subpopulations isolated by sorting were adjusted to 1 × 108 cells by subpopulation and injected in a circular pattern into the cicatrix previously defined by echocardiography. RESULTS: Patients were followed for 6 and 12 months. Six months after cell implant improvements in left ventricle ejection fraction (from 33-50%), stress rate values (from -3/-9% to -18/-22%), stress tests (from 4-12 METS), and the quantity of left ventricle affected segments (3-9) disappeared according to the G-SPECT images. 12 months evaluations did not show significant differences. Interestingly, 3 months after cell implant the ECG showed normal electrical activity in 9 patients whereas after 6 months it was normal in all the patients. CONCLUSIONS: These results ratify that locally injected autologous CXCR4+ bone marrow-derived stem cells have a physiological and a clinical impact in patients with CMI.


Assuntos
Células-Tronco Hematopoéticas/metabolismo , Infarto do Miocárdio/terapia , Receptores CXCR4/uso terapêutico , Idoso , Humanos , Masculino , Pessoa de Meia-Idade
14.
Lima; IETSI; 2019.
Não convencional em Espanhol | BRISA/RedTESA | ID: biblio-1009658

RESUMO

INTRODUCCIÓN: El presente dictamen expone la evaluación de la eficacia y seguridad del esquema combinado de plerixafor 0.24 mg/Kg/día administrado vía subcutánea + factor estimulante de colonias-granulocitos (FEC-G), en comparación con el tratamiento con FEC-G solo, para la movilización de células CD34+ en pacientes con linfoma no Hodgkin (LNH) candidatos a trasplante autólogo con falla de uno o más esquemas de movilización de células progenitoras hematopoyéticas (CPH). Se denomina linfoma no-Hodgkin (LNH) a un grupo heterogéneo de neoplasias malignas linfoproliferativas con diferentes patrones histológicos, manifestaciones clínicas y respuesta al tratamiento. Los LNH representan cerca del 90% de los linfomas. Solo en el Instituto Nacional de Enfermedades Neoplásica de Perú se registran anualmente más de 500 casos de LNH. La mortalidad en los casos registrados en Lima metropolitana asciende a 4.82 por cada 100,000 casos. El tratamiento del LNH varía, según el grado de la enfermedad. Entre el 20 y 30% de los pacientes con LNH no logra una remisión completa de la enfermedad con el tratamiento inicial. En los casos de LNH refractario o recidivante, el tratamiento estándar es el trasplante autólogo de células progenitoras hematopoyéticas (TA-CPH) con quimioterapia a altas dosis. TECNOLOGÍA SANITARIA DE INTERÉS: Plerixafor inhibe selectivamente el receptor de quimiocina CXCR4; facilitando el flujo de las CPH hacia la circulación periférica. Plerixafor está aprobado por la Food and Drug Administration y la European Medicines Agency, en combinación con FEC-G, para la movilización de CPH a la circulación periférica para la realización de TA-CPH en pacientes con LNH o mieloma múltiple. METODOLOGÍA: Se realizó una búsqueda sistemática de la evidencia sobre la eficacia de plerixafor + FEC-G, comparado FEC-G solo, en la re-movilización de CPH; de acuerdo con la pregunta PICO validada con la especialista. La revisión de la información con respecto a los esquemas de removilización en pacientes con LNH y mieloma múltiple mostró otros esquemas de re-movilización; los cuales incluyen FEC-G + quimioterapia. Dado que EsSalud dispone de quimioterápicos para el manejo de LNH (por ejemplo, ciclofosfamida), se ampliaron los criterios de elegibilidad para incluir, además, evidencia que compare plerixafor + FEC-G versus FEC-G + quimioterapia. Adicionalmente, se agregó la sobrevida global como desenlace de interés. RESULTADOS: Tras una búsqueda sistemática de literatura científica publicada hasta la actualidad, no se encontraron revisiones sistemáticas o ensayos clínicos aleatorizados que respondan directamente a la pregunta PICO de interés. Por este motivo, se optó por incluir también estudios observacionales. Se incluyeron cinco guías de práctica clínica (GPC) elaboradas por: la American Society for Blood and Marrow Transplantation (SBMT), el British Society for Haematology (BSH), un equipo de trabajo del Stem Cell Transplant Steering Committee (SCTSC), un grupo de expertos brasileños (Duarte 2016) y un grupo de clínicos representantes de Reino Unido e Irlanda (Douglas 2017). Estas GPC trataron sobre el manejo de pacientes que fallaron a algún esquema de movilización de CPH. Además, se incluyó una evaluación de tecnología sanitaria (ETS) realizada por la Canadian Agency for Drugs and Technologies in Health (CADTH); la cual evaluó la eficacia del plerixafor como parte de esquemas de re-movilización de CPH. Por último, se incluyeron dos estudios retrospectivos (Pusic 2008 y Perkins 2012) que compararon el esquema FEC-G + plerixafor con otros esquemas de re-movilización, dentro de los cuales se incluyen FEC-G solo y FEC-G + quimioterapia. CONCLUSIONES: El presente dictamen preliminar tuvo como objetivo evaluar la evidencia disponible sobre la eficacia de plerixafor usado junto con FEC-G (filgrastim) como esquema de re-movilización de CPH en pacientes adultos con LNH, candidatos a TA-CPH con falla de uno o más intentos de movilización Tras la búsqueda sistemática de literatura, no se encontró evidencia que compare directamente, plerixafor + FEC-G con FEC-G solo, como esquema de re-movilización de CPH en la población de interés del presente dictamen preliminar (pacientes adultos con LNH, candidatos a TA-CPH con falla de uno o más intentos de movilización previos). Por otro lado, aunque se encontró evidencia indirecta que compara plerixafor + FEC-G con FEC-G solo, la toma de decisión a partir de esta evidencia presenta las siguientes limitaciones: (i) Los estudios realizados únicamente en pacientes con LNH son pocos, y en los casos de poblaciones más grandes, la proporción de pacientes con LNH es muy pequeña. Por lo tanto, los resultados obtenidos no pueden generalizarse con total confianza a la población de interés del presente dictamen, (ii) Los resultados de esta evidencia indirecta sugieren que los esquemas de re-movilización que incluyen plerixafor son tan eficaces como los esquemas de re-movilización que no incluyen plerixafor. Sin embargo, dado que esta evidencia proviene de poblaciones con patologías distintas a LNH (en términos de fisiopatología, severidad y pronóstico), la capacidad para generalizar estos resultados es limitada, (iii) Pocos estudios han reportado la frecuencia de eventos adversos; y la hacerlo, ha sido de manera selectiva o sin el objetivo de comparar esquemas de re-movilización. Todo ello impide realizar una conclusión sobre la seguridad de plerixafor. En consecuencia, con la evidencia disponible hasta la fecha, no se tienen argumentos suficientes para concluir que el esquema de re-movilización con FEC-G (filgrastim) y plerixafor es más eficaz y seguro que otros esquemas de re-movilización disponibles en EsSalud. Además, al considerar su alto costo, tampoco es posible asumir un perfil de costo-oportunidad favorable para sistemas públicos de servicios de salud como el nuestro. El Instituto de Evaluaciones de Tecnologías en Salud e Investigación - IETSI, no aprueba el uso de plerixafor junto con FEC-G (filgrastim) como esquema de movilización de CPH en pacientes adultos con LNH, candidatos a TA-CPH con falla de uno o más intentos de movilización previos.


Assuntos
Humanos , Linfoma não Hodgkin/tratamento farmacológico , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Receptores CXCR4/uso terapêutico , Avaliação da Tecnologia Biomédica , Análise Custo-Eficiência
15.
Proc Natl Acad Sci U S A ; 115(16): 4270-4275, 2018 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-29610328

RESUMO

Epilepsy is a neurological disorder often associated with seizure that affects ∼0.7% of pregnant women. During pregnancy, most epileptic patients are prescribed antiepileptic drugs (AEDs) such as valproic acid (VPA) to control seizure activity. Here, we show that prenatal exposure to VPA in mice increases seizure susceptibility in adult offspring through mislocalization of newborn neurons in the hippocampus. We confirmed that neurons newly generated from neural stem/progenitor cells (NS/PCs) are integrated into the granular cell layer in the adult hippocampus; however, prenatal VPA treatment altered the expression in NS/PCs of genes associated with cell migration, including CXC motif chemokine receptor 4 (Cxcr4), consequently increasing the ectopic localization of newborn neurons in the hilus. We also found that voluntary exercise in a running wheel suppressed this ectopic neurogenesis and countered the enhanced seizure susceptibility caused by prenatal VPA exposure, probably by normalizing the VPA-disrupted expression of multiple genes including Cxcr4 in adult NS/PCs. Replenishing Cxcr4 expression alone in NS/PCs was sufficient to overcome the aberrant migration of newborn neurons and increased seizure susceptibility in VPA-exposed mice. Thus, prenatal exposure to an AED, VPA, has a long-term effect on the behavior of NS/PCs in offspring, but this effect can be counteracted by a simple physical activity. Our findings offer a step to developing strategies for managing detrimental effects in offspring exposed to VPA in utero.


Assuntos
Anticonvulsivantes/toxicidade , Neurogênese/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal , Convulsões/etiologia , Ácido Valproico/toxicidade , Animais , Anticonvulsivantes/administração & dosagem , Anticonvulsivantes/farmacologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Células Cultivadas , Giro Denteado/efeitos dos fármacos , Giro Denteado/embriologia , Giro Denteado/patologia , Suscetibilidade a Doenças , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Idade Gestacional , Hipocampo/embriologia , Hipocampo/patologia , Hipocampo/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Células-Tronco Neurais/efeitos dos fármacos , Neurônios/patologia , Esforço Físico , Gravidez , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Distribuição Aleatória , Ratos , Receptores CXCR4/biossíntese , Receptores CXCR4/genética , Receptores CXCR4/uso terapêutico , Convulsões/induzido quimicamente , Convulsões/embriologia , Transcriptoma , Ácido Valproico/administração & dosagem , Ácido Valproico/farmacologia
16.
Santiago; MINSAL; 2018. tab.
Não convencional em Espanhol | BRISA/RedTESA | ID: biblio-1022165

RESUMO

INTRODUCCIÓN: El mieloma múltiple (MM) se caracteriza por la proliferación neoplásica de células plasmáticas que producen una inmunoglobulina monoclonal, estas células plasmáticas proliferan en la médula ósea y frecuentemente, dan como resultado una extensa destrucción esquelética con lesiones osteolíticas, osteopenia y / o fracturas patológicas. La sospecha diagnóstica se inicia, generalmente debido a la presencia de dolor óseo con lesiones líticas, aumento de la concentración sérica total de proteínas o presencia de una proteína monoclonal en orina o suero, signos o síntomas sistémicos sugestivos de malignidad como anemia inexplicada, hipercalcemia, insuficiencia renal aguda con un urinálisis suave o raramente el síndrome nefrótico debido a la amiloidosis de cadena ligera de inmunoglobulina concurrente, pudiendo presentarse de forma copulativa. Es importante distinguir MM tanto de otras causas de las presentaciones clínicas anteriores, como de otras discrasias de células plasmáticas, para fines de pronóstico y tratamiento. Se considerarán para su evaluación aquellas solicitudes realizadas conforme al Reglamento que establece el proceso destinado a determinar los diagnósticos y tratamientos de alto costo con sistema de protección financiera, según lo establecido en los artículos 7° y 8° de la Ley N° 20.850. Estas solicitudes no son vinculantes para el Ministerio de Salud, debiendo, sin embargo, tomar especialmente en cuenta aquellas solicitudes y opiniones que hayan sido realizadas por sus comisiones técnicas asesoras y por las asociaciones de pacientes incluidas en el Registro de Asociaciones de Pacientes que crea la Ley 20.850. De igual forma, para ser incorporadas en el proceso de evaluación científica de la evidencia, cada intervención debe cumplir con los criterios establecidos en el Artículo 6o del Reglamento mencionado, según lo indicado en el Numeral 9 del presente informe. TECNOLOGÍAS SANITARIA DE INTERÉS: Bortezomib es un fármaco que actúa por inhibición reversible del proteasoma. Al inhibir los proteasomas (complejos multi-enzimáticos presentes en todas las células), Bortezomib interfiere con el ciclo celular que conduce a la muerte celular . Plerixafor es un antagonista potente, selectivo y transitorio del receptor de quimioquinas CXCR4 y bloquea la unión de su ligando afín, el factor 1 derivado de células estromales (SDF-1), también conocido como CXCL12. Moviliza las células madre de la médula ósea aumentando su número en sangre periférica. Lenalidomida es un agente inmunomodulador que pertenece a una clase de agentes a menudo denominados derivados inmunomoduladores, que son todos derivados estructurales de talidomida. El mecanismo de acción exacto de la lenalidomida no se comprende del todo actualmente, pero tiene propiedades anti-neoplásicas, anti-angiogénicas y pro-eritropoyéticas. Bendamustina: El clorhidrato de Bendamustina es un agente antitumoral alquilante con actividad única. El efecto antineoplásico y citocida del clorhidrato de bendamustina se basa esencialmente en una reticulación de cadenas simples y dobles de ADN por alquilación. Como resultado, las funciones de la matriz de ADN y la síntesis y reparación del ADN se deterioran. EFICACIA DE LOS TRATAMIENTOS: Bortezomib: Encontramos 23 revisiones sistemáticas que incluyen 6 estudios primarios todos correspondientes a ensayos controlados aleatorizados. Solo 2 de estos estudios consideraron el uso de bortezomib post trasplante autólogo de células madre y 4 de estos estudios consideraron el uso de bortezomib como terapia de inducción pre trasplante autólogo de células madre. Se puede concluir que la adición de bortezomib al tratamiento del mieloma múltiple disminuiría la mortalidad, pero la certeza en la evidencia es baja. Plerixafor: Encontramos 2 revisiones sistemáticas, las cuales incluyen 3 estudios primarios, todos correspondientes a ensayos controlados aleatorizados. En estos ensayos, el Plerixafor podría tener poco o nulo efecto sobre la mortalidad, pero la certeza de la evidencia es baja y aumenta la proporción de pacientes que alcanzan más de 6x10


Assuntos
Humanos , Receptores CXCR4/uso terapêutico , Bortezomib/uso terapêutico , Cloridrato de Bendamustina/uso terapêutico , Lenalidomida/uso terapêutico , Mieloma Múltiplo/tratamento farmacológico , Avaliação da Tecnologia Biomédica , Análise Custo-Benefício/economia
17.
Nanomedicine (Lond) ; 11(18): 2387-98, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27529439

RESUMO

AIM: Lactococcus lactis is a Gram-positive (endotoxin-free) food-grade bacteria exploited as alternative to Escherichia coli for recombinant protein production. We have explored here for the first time the ability of this platform as producer of complex, self-assembling protein materials. MATERIALS & METHODS: Biophysical properties, cell penetrability and in vivo biodistribution upon systemic administration of tumor-targeted protein nanoparticles produced in L. lactis have been compared with the equivalent material produced in E. coli. RESULTS: Protein nanoparticles have been efficiently produced in L. lactis, showing the desired size, internalization properties and biodistribution. CONCLUSION: In vitro and in vivo data confirm the potential and robustness of the production platform, pointing out L. lactis as a fascinating cell factory for the biofabrication of protein materials intended for therapeutic applications.


Assuntos
Lactococcus lactis/genética , Nanopartículas/química , Neoplasias/tratamento farmacológico , Proteínas Recombinantes/genética , Escherichia coli/genética , Humanos , Nanopartículas/administração & dosagem , Transporte Proteico/genética , Receptores CXCR4/química , Receptores CXCR4/genética , Receptores CXCR4/uso terapêutico , Proteínas Recombinantes/química , Proteínas Recombinantes/uso terapêutico , Distribuição Tecidual
18.
Cytometry B Clin Cytom ; 90(2): 209-19, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26384735

RESUMO

BACKGROUND: Receptor occupancy (RO) assays provide a means to measure the direct interaction of therapeutics with their cell surface targets. Free receptor assays quantify cell-surface receptors not bound by a therapeutic while total receptor assays quantify the amount of target on the cell surface. METHODS: We developed both a flow cytometry-based free RO assay to detect free surface CXCR4, and a total surface CXCR4 assay. In an effort to evaluate potential displacement interference, we performed in vitro experiments to compare on-cell affinity with the IC50 values from in vitro and in vivo from the free CXCR4 assay. We determined free and total surface CXCR4 on circulating blood cells in cynomolgus monkeys dosed with MEDI3185, a fully human monoclonal antibody to CXCR4. RESULTS: We devised an approach to evaluate displacement interference during assay development and showed that our free assay demonstrated little to no displacement interference. After dosing cynomolgus monkeys with MEDI3185, we observed dose-dependence in the magnitude and duration of receptor occupancy and found CXCR4 to increase on lymphocytes, monocytes, and granulocytes. In a multiple dose study, we observed time points where surface CXCR4 appeared fully occupied but MEDI3185 was not detectable in serum. These paradoxical results represented a type of assay interference, and by comparing pharmacokinetic, ADA and total CXCR4 results, the most likely reason for the free CXCR4 results was the emergence of neutralizing anti-drug antibodies (ADA). The total CXCR4 assay was unaffected by ADA and provided a reliable marker of target modulation in both in vivo studies.


Assuntos
Anticorpos Anti-Idiotípicos/administração & dosagem , Anticorpos Monoclonais/administração & dosagem , Citometria de Fluxo , Receptores CXCR4/uso terapêutico , Animais , Anticorpos Anti-Idiotípicos/imunologia , Anticorpos Monoclonais/imunologia , Granulócitos/efeitos dos fármacos , Granulócitos/imunologia , Humanos , Imunoglobulinas Intravenosas/administração & dosagem , Linfócitos/efeitos dos fármacos , Linfócitos/imunologia , Macaca fascicularis/imunologia , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Receptores CXCR4/imunologia
19.
Belo Horizonte; CCATES; 2016. tab.
Não convencional em Português | BRISA/RedTESA | ID: biblio-876493

RESUMO

CONTEXTO: A MW (CID-10 C88.0) é um tipo de câncer caracterizado por distúrbio linfoproliferativo de linfócitos B, levando a uma produção monoclonal da imunoglobulina M (IgM), o que ocasiona a hipergamaglobulinemia (aumento de IgM no plasma sanguíneo) e infiltração de linfoma linfoplasmocítico na medula óssea. De acordo com a classificação dos tumores hematopoiéticos e tecidos linfóides da Organização Mundial da Saúde (OMS), elaborado em 2008, é classificado como um tipo de linfoma linfoplasmocítico. Atualmente é uma doença incurável, sendo objetivo do tratamento o aumento da sobrevida do paciente. TECNOLOGIA: Plerixafor. PERGUNTA: Existe evidência para o uso de plerixafor em pacientes com macroglobulinemia de Waldenström? EVIDÊNCIAS: Não foram encontrados ensaios clínicos randomizados, revisões sistemáticas e estudos de avaliação econômica do plerixafor para pacientes com macroglobulinemia de Waldenström (MW). CONCLUSÕES: Apesar de estar disponível no mercado internacional desde 2008, não foram encontrados evidências, protocolos e diretrizes que recomendem o uso do plerixafor em pacientes com MW.


Assuntos
Humanos , Macroglobulinemia de Waldenstrom/tratamento farmacológico , Análise Custo-Benefício/economia , Receptores CXCR4/uso terapêutico , Avaliação da Tecnologia Biomédica , Resultado do Tratamento
20.
Gene Ther ; 21(4): 444-9, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24572792

RESUMO

Resting human CD4 T cells are highly resistant to transfection or infection with lentiviral vectors derived from the human immunodeficiency virus. We now describe a flexible and efficient approach involving virus-like particles containing simian immunodeficiency virus lentiviral gene product protein X and pseudotyping with CXCR4-tropic HIV Env. This method permits effective genetic manipulation of these cells while preserving their naturally quiescent state. This technology can also be extended to primary lymphoid cultures where authentic cellular composition and functional relationships are preserved.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Técnicas de Transferência de Genes , Terapia Genética , Proteínas Virais/genética , Animais , Linfócitos T CD4-Positivos/patologia , Vetores Genéticos , HIV-1/genética , Humanos , Lentivirus/genética , Receptores CXCR4/genética , Receptores CXCR4/uso terapêutico , Vírus da Imunodeficiência Símia , Proteínas Virais/administração & dosagem , Proteínas Virais/uso terapêutico , Produtos do Gene env do Vírus da Imunodeficiência Humana/genética , Produtos do Gene env do Vírus da Imunodeficiência Humana/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...