Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
AAPS J ; 26(1): 9, 2023 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-38114736

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a progressive fatal interstitial lung disease that affects three million patients worldwide and currently without an effective cure. Zinpentraxin alfa, a recombinant human pentraxin-2 (rhPTX-2) protein, has been evaluated as a potential drug candidate for the treatment of IPF. Clinical pharmacokinetic analysis of zinpentraxin alfa has been challenging historically due to interference from serum amyloid P component (SAP), an endogenous human pentraxin-2 protein. These molecules share an identical primary amino acid sequence and glycan composition; however, zinpentraxin alfa possesses α2,3-linked terminal sialic acid residues while SAP is an α2,6-linked isomer. By taking advantage of this only structural difference, we developed a novel assay strategy where α2,3-sialidase was used to selectively hydrolyze α2,3-linked sialic acid residues, resulting in desialylated zinpentraxin alfa versus unchanged sialylated SAP, following an immunoaffinity capture step. Subsequent tryptic digestion produced a unique surrogate asialo-glycopeptide from zinpentraxin alfa and allowed specific quantification of the biotherapeutic in human plasma. In addition, a common peptide shared by both molecules was selected as a surrogate to determine total hPTX-2 concentrations, i.e., sum of zinpentraxin alfa and SAP. The quantification methods for both zinpentraxin alfa and total hPTX-2 were validated and used in pharmacokinetic assessment in IPF patients. The preliminary results suggest that endogenous SAP levels remained largely constant in IPF patients throughout the treatment with zinpentraxin alfa. Our novel approach provides a general bioanalytical strategy to selectively quantify α2,3-sialylated glycoproteins in the presence of their corresponding α2,6-linked isomers.


Assuntos
Fibrose Pulmonar Idiopática , 60705 , Humanos , Cromatografia Líquida , Ácido N-Acetilneuramínico/análise , Ácido N-Acetilneuramínico/química , Espectrometria de Massas em Tandem , Fibrose Pulmonar Idiopática/tratamento farmacológico
2.
J Cancer Res Ther ; 19(5): 1160-1169, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37787279

RESUMO

Aim: To evaluate the use of this new technique, surface-guided radiotherapy (SGRT), for patient setup and motion management in various cancers. Materials and Methods: Data was collected from 533 patients, who received treatment in our hospital for various malignancies using SGRT from October 2019 to April 2021. We studied patient setup, interfraction position, and patient position during the breath-hold (BH) technique. The main advantage of SGRT is that, it is completely non-invasive and uses visible light to compare the patient's skin surface in the treatment room and planned treatment position. In this analysis, Monaco 5.51.10 (Elekta) treatment planning system, Versa HD Linear Accelerator, and AlignRT 6.2 (Vision RT) SGRT system were used. Results: With SGRT, treatment setup time can be reduced with more precision and techniques like Deep inspiration breathhold (DIBH) can be done with very good compliance. Conclusion: SGRT has shown improved accuracy in patient setup compared to conventional laser setup. The daily kilo voltage imaging frequency can be reduced; it helps in reducing additional radiation exposure due to imaging. SGRT has demonstrated reproducibility with adequate accuracy in BH treatments in DIBH for breast and SBRT.


Assuntos
Neoplasias da Mama , Neoplasias , Radioterapia Guiada por Imagem , Feminino , Humanos , Mama , Neoplasias da Mama/radioterapia , Suspensão da Respiração , Neoplasias/radioterapia , Dosagem Radioterapêutica , Planejamento da Radioterapia Assistida por Computador/métodos , Radioterapia Guiada por Imagem/métodos , Reprodutibilidade dos Testes , Tomografia Computadorizada por Raios X
3.
Sci Transl Med ; 14(675): eabp9159, 2022 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-36516271

RESUMO

The epidermis is a barrier that prevents water loss while keeping harmful substances from penetrating the host. The impermeable cornified layer of the stratum corneum is maintained by balancing continuous turnover driven by epidermal basal cell proliferation, suprabasal cell differentiation, and corneal shedding. The epidermal desquamation process is tightly regulated by balance of the activities of serine proteases of the Kallikrein-related peptidases (KLK) family and their cognate inhibitor lymphoepithelial Kazal type-related inhibitor (LEKTI), which is encoded by the serine peptidase inhibitor Kazal type 5 gene. Imbalance of proteolytic activity caused by a deficiency of LEKTI leads to excessive desquamation due to increased activities of KLK5, KLK7, and KLK14 and results in Netherton syndrome (NS), a debilitating condition with an unmet clinical need. Increased activity of KLKs may also be pathological in other dermatoses such as atopic dermatitis (AD). Here, we describe the discovery of inhibitory antibodies against murine KLK5 and KLK7 that could compensate for the deficiency of LEKTI in NS. These antibodies are protective in mouse models of NS and AD and, when combined, promote improved skin barrier integrity and reduced inflammation. To translate these findings, we engineered a humanized bispecific antibody capable of potent inhibition of human KLK5 and KLK7. A crystal structure of KLK5 bound to the inhibitory Fab revealed that the antibody binds distal to its active site and uses a relatively unappreciated allosteric inhibition mechanism. Treatment with the bispecific anti-KLK5/7 antibody represents a promising therapy for clinical development in NS and other inflammatory dermatoses.


Assuntos
Dermatite Atópica , Síndrome de Netherton , Dermatopatias , Camundongos , Humanos , Animais , Síndrome de Netherton/genética , Síndrome de Netherton/metabolismo , Síndrome de Netherton/patologia , Dermatite Atópica/patologia , Inibidor de Serinopeptidase do Tipo Kazal 5/metabolismo , Epiderme/patologia , Dermatopatias/metabolismo , Anticorpos/metabolismo , Calicreínas/metabolismo
4.
Am J Physiol Cell Physiol ; 320(2): C162-C174, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33206546

RESUMO

Proteolytic processing of procollagens is a central step during collagen fibril formation. Bone morphogenic protein 1 (BMP1) is a metalloprotease that plays an important role in the cleavage of carboxy-terminal (COOH-terminal) propeptides from procollagens. Although the removal of propeptides is required to generate mature collagen fibrils, the contribution of BMP1 to this proteolytic process and its action site remain to be fully determined. In this study, using postnatal lung fibroblasts as a model system, we showed that genetic ablation of Bmp1 in primary murine lung fibroblasts abrogated COOH-terminal cleavage from type I procollagen as measured by COOH-terminal propeptide of type I procollagen (CICP) production. We also showed that inhibition of BMP1 by siRNA-mediated knockdown or small-molecule inhibitor reduced the vast majority of CICP production and collagen deposition in primary human lung fibroblasts. Furthermore, we discovered and characterized two antibody inhibitors for BMP1. In both postnatal lung fibroblast and organoid cultures, BMP1 blockade prevented CICP production. Together, these findings reveal a nonredundant role of extracellular BMP1 to process CICP in lung fibroblasts and suggest that development of antibody inhibitors is a viable pharmacological approach to target BMP1 proteinase activity in fibrotic diseases.


Assuntos
Proteína Morfogenética Óssea 1/metabolismo , Líquido Extracelular/metabolismo , Fibroblastos/metabolismo , Pulmão/metabolismo , Fragmentos de Peptídeos/metabolismo , Pró-Colágeno/metabolismo , Proteólise , Sequência de Aminoácidos , Animais , Proteína Morfogenética Óssea 1/antagonistas & inibidores , Proteína Morfogenética Óssea 1/genética , Células CHO , Cricetinae , Cricetulus , Líquido Extracelular/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Células HEK293 , Humanos , Pulmão/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Técnicas de Cultura de Órgãos , Organoides , Oxidiazóis/farmacologia , Fragmentos de Peptídeos/genética , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/metabolismo , Pró-Colágeno/genética , Inibidores de Proteases/farmacologia , Proteólise/efeitos dos fármacos , Coelhos
5.
PLoS One ; 15(12): e0244158, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33347473

RESUMO

The proactive generation of anti-idiotypic antibodies (anti-IDs) against therapeutic antibodies with desirable properties is an important step in pre-clinical and clinical assay development supporting their bioanalytical programs. Here, we describe a robust platform to generate anti-IDs using rabbit single B cell sorting-culture and cloning technology by immunizing rabbits with therapeutic drug Fab fragment and sorting complementarity determining regions (CDRs) specific B cells using designed framework control as a negative gate to exclude non-CDRs-specific B cells. The supernatants of cultured B cells were subsequently screened for binding to drug-molecule by enzyme-linked immunosorbent assay and the positive hits of B cell lysates were selected for cloning of their immunoglobulin G (IgG) variable regions. The recombinant monoclonal anti-IDs generated with this method have high affinity and specificity with broad epitope coverage and different types. The recombinant anti-IDs were available for assay development to support pharmacokinetic (PK) and immunogenicity studies within 12 weeks from the start of rabbit immunization. Using this novel rapid and efficient in-house approach we have generated a large panel of anti-IDs against a series of 11 therapeutic antibody drugs and successfully applied them to the clinical assay development.


Assuntos
Anticorpos Anti-Idiotípicos/imunologia , Anticorpos Monoclonais/imunologia , Afinidade de Anticorpos , Linfócitos B/imunologia , Separação Celular/métodos , Epitopos/imunologia , Animais , Anticorpos Anti-Idiotípicos/genética , Anticorpos Monoclonais/genética , Linfócitos B/classificação , Células Cultivadas , Clonagem Molecular/métodos , Epitopos/genética , Humanos , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/imunologia , Coelhos
6.
Hum Vaccin Immunother ; 13(5): 1094-1104, 2017 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-28059624

RESUMO

Patients with refractory or recurrent B-lineage hematologic malignancies have less than 50% of chance of cure despite intensive therapy and innovative approaches are needed. We hypothesize that gene modification of haematopoietic stem cells (HSC) with an anti-CD19 chimeric antigen receptor (CAR) will produce a multi-lineage, persistent immunotherapy against B-lineage malignancies that can be controlled by the HSVsr39TK suicide gene. High-titer third-generation self-inactivating lentiviral constructs were developed to deliver a second-generation CD19-specific CAR and the herpes simplex virus thymidine kinase HSVsr39TK to provide a suicide gene to allow ablation of gene-modified cells if necessary. Human HSC were transduced with such lentiviral vectors and evaluated for function of both CAR and HSVsr39TK. Satisfactory transduction efficiency was achieved; the addition of the suicide gene did not impair CAR expression or antigen-specific cytotoxicity, and determined marked cytotoxicity to ganciclovir. NSG mice transplanted with gene-modified human HSC showed CAR expression not significantly different between transduced cells with or without HSVsr39TK, and expression of anti-CD19 CAR conferred anti-tumor survival advantage. Treatment with ganciclovir led to significant ablation of gene-modified cells in mouse tissues. Haematopoietic stem cell transplantation is frequently part of the standard of care for patients with relapsed and refractory B cell malignancies; following HSC collection, a portion of the cells could be modified to express the CD19-specific CAR and give rise to a persistent, multi-cell lineage, HLA-independent immunotherapy, enhancing the graft-versus-malignancy activity.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas , Linfoma de Células B/terapia , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Animais , Linfócitos B/imunologia , Antígenos CD28/imunologia , Ganciclovir/administração & dosagem , Humanos , Imunoterapia , Células Jurkat , Lentivirus/genética , Linfoma de Células B/imunologia , Camundongos , Recidiva Local de Neoplasia/terapia , Receptores de Antígenos de Linfócitos T/uso terapêutico , Simplexvirus/enzimologia , Simplexvirus/genética , Timidina Quinase/genética , Transdução Genética
7.
Breast Cancer Res Treat ; 149(3): 655-68, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25663548

RESUMO

Epigenetic therapy by DNA demethylating agent 5-aza-2'-deoxycytidine (5-aza 2'dC) is clinically effective in acute myeloid leukemia; however, it has shown limited results in treatment of breast cancer and has significant toxicity to normal cells. Green tea polyphenol (-)-epigallocatechin-3-gallate (EGCG) has anti-cancer and DNA demethylating properties with no significant toxicity toward normal cells. Therefore, the objective of this study was to evaluate the therapeutic efficacy of a combination of non-toxic, low dose of 5-aza 2' dC with EGCG, on growth inhibition of breast cancer cells. Human breast cancer cell lines (MCF-7, MDA-MB 231) and non-tumorigenic MCF-10A breast epithelial cells were treated with either 5-aza 2' dC, EGCG, or their combination for 7 days. Cell growth inhibition was determined by cell count, cell viability, cell cycle, and soft agar assay, whereas genes expression changes were determined by quantitative real-time PCR and/or Western blot analysis. Histone modifications and global DNA methylation changes were determined by Western blot and RAPD-PCR, respectively. The results revealed significantly greater inhibition of growth of breast cancer cells by co-treatment with 5-aza 2' dC and EGCG compared to individual treatments, whereas it has no significant toxicity to MCF-10A cells. This was further confirmed by gene expression analysis. Changes in DNA methylation and histone modifications were also greater in cells with combination treatment. Findings of this study suggest that potentiation of growth inhibition of breast cancer cells by 5-aza 2' dC and EGCG combination treatment, at least in part, is mediated by epigenetic mechanism.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Catequina/análogos & derivados , Proliferação de Células/efeitos dos fármacos , Epigênese Genética/efeitos dos fármacos , Extratos Vegetais/administração & dosagem , Azacitidina/administração & dosagem , Azacitidina/análogos & derivados , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Catequina/administração & dosagem , Catequina/química , Sobrevivência Celular/efeitos dos fármacos , Metilação de DNA/efeitos dos fármacos , Decitabina , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Histonas/genética , Humanos , Células MCF-7 , Extratos Vegetais/química , Técnica de Amplificação ao Acaso de DNA Polimórfico , Chá/química
8.
Prostate ; 73(15): 1660-72, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23804311

RESUMO

BACKGROUND: Chronic exposure to arsenic and estrogen is associated with risk of prostate cancer, but their mechanism is not fully understood. Additionally, the carcinogenic effects of their co-exposure are not known. Therefore, the objective of this study was to evaluate the effects of chronic exposure to arsenic, estrogen, and their combination, on cell growth and transformation, and identify the mechanism behind these effects. METHODS: RWPE-1 human prostate epithelial cells were chronically exposed to arsenic and estrogen alone and in combination. Cell growth was measured by cell count and cell cycle, whereas cell transformation was evaluated by colony formation assay. Gene expression was measured by quantitative real-time PCR and confirmed at protein level by Western blot analysis. MLH1 promoter methylation was determined by pyrosequencing method. RESULTS: Exposure to arsenic, estrogen, and their combinations increases cell growth and transformation in RWPE-1 cells. Increased expression of Cyclin D1 and Bcl2, whereas decreased expression of mismatch repair genes MSH4, MSH6, and MLH1 was also observed. Hypermethylation of MLH1 promoter further suggested the epigenetic inactivation of MLH1 expression in arsenic and estrogen treated cells. Arsenic and estrogen combination caused greater changes than their individual treatments. CONCLUSIONS: Findings of this study for the first time suggest that arsenic and estrogen exposures cause increased cell growth and survival potentially through epigenetic inactivation of MLH1 resulting in decreased MLH1-mediated apoptotic response, and consequently increased cellular transformation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Arsênio/administração & dosagem , Crescimento Celular/efeitos dos fármacos , Transformação Celular Neoplásica/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Estrogênios/administração & dosagem , Proteínas Nucleares/genética , Próstata/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Expressão Gênica/efeitos dos fármacos , Inativação Gênica , Humanos , Masculino , Metilação , Proteína 1 Homóloga a MutL , Proteínas Nucleares/metabolismo , Próstata/metabolismo
9.
PLoS One ; 7(8): e43880, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22952798

RESUMO

Chronic exposures to arsenic and estrogen are known risk factors for prostate cancer. Though the evidence suggests that exposure to arsenic or estrogens can disrupt normal DNA methylation patterns and histone modifications, the mechanisms by which these chemicals induce epigenetic changes are not fully understood. Moreover, the epigenetic effects of co-exposure to these two chemicals are not known. Therefore, the objective of this study was to evaluate the effects of chronic exposure to arsenic and estrogen, both alone and in combination, on the expression of epigenetic regulatory genes, their consequences on DNA methylation, and histone modifications. Human prostate epithelial cells, RWPE-1, chronically exposed to arsenic and estrogen alone and in combination were used for analysis of epigenetic regulatory genes expression, global DNA methylation changes, and histone modifications at protein level. The result of this study revealed that exposure to arsenic, estrogen, and their combination alters the expression of epigenetic regulatory genes and changes global DNA methylation and histone modification patterns in RWPE-1 cells. These changes were significantly greater in arsenic and estrogen combination treated group than individually treated group. The findings of this study will help explain the epigenetic mechanism of arsenic- and/or estrogen-induced prostate carcinogenesis.


Assuntos
Arsênio/farmacologia , Epigênese Genética/efeitos dos fármacos , Epigênese Genética/genética , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Estrogênios/farmacologia , Próstata/citologia , Linhagem Celular , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA/efeitos dos fármacos , Metilação de DNA/genética , Interações Medicamentosas , Estradiol/farmacologia , Histonas/metabolismo , Humanos , Masculino , Fatores de Tempo
10.
Cancer Lett ; 316(1): 62-9, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22082530

RESUMO

Prolonged exposure to elevated levels of estrogen is a risk factor for breast cancer. Though increased cell growth and loss of DNA repair capacity is one of the proposed mechanisms for estrogen-induced cancers, the mechanism through which estrogen induces cell growth and decreases DNA repair capacity is not clear. DNA hypermethylation is known to inactivate DNA repair genes and apoptotic response in cancer cells. Therefore, the objective of this study was to determine the role of DNA hypermethylation in estrogen-induced cell growth and regulation of DNA repair genes expression in breast cancer cells. To achieve this objective, the estrogen-responsive MCF-7 cells either pretreated with 5-aza-2-deoxycytidine (5-aza-dC) or untreated (as control) were exposed to 17 beta-estradiol (E2), and its effect on cell growth and expression of DNA repair genes were measured. The result revealed that 5-aza-dC abrogates the E2-induced growth in MCF-7 cells. An increased expression of OGG1, MSH4, and MLH1 by 5-aza-dC treatment alone, suggest the DNA hypermethylation as a potential cause for decreased expression of these genes in MCF-7 cells. The decreased expression of ERCC1, XPC, OGG1, and MLH1 by E2 alone and its restoration by co-treatment with 5-aza-dC further suggest that E2 reduces the expression of these DNA repair genes potentially through promoter hypermethylation. Reactivation of mismatch repair (MMR) gene MLH1 and abrogation of E2-induced cell growth by 5-aza-dC treatment suggest that estrogen causes increased growth in breast cancer cells potentially through the inhibition of MMR-mediated apoptotic response. In summary, this study suggests that estrogen increases cell growth and decreases the DNA repair capacity in breast cancer cells, at least in part, through epigenetic mechanism.


Assuntos
Azacitidina/análogos & derivados , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Metilação de DNA/efeitos dos fármacos , Reparo do DNA , Estradiol/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Azacitidina/farmacologia , Neoplasias da Mama/patologia , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Processos de Crescimento Celular/efeitos dos fármacos , Processos de Crescimento Celular/genética , Reparo de Erro de Pareamento de DNA , Decitabina , Interações Medicamentosas , Epigênese Genética/efeitos dos fármacos , Epigênese Genética/genética , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Regiões Promotoras Genéticas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...