Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 14(1): 11018, 2024 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-38744902

RESUMO

Antibody-drug conjugates (ADC) payloads are cleavable drugs that act as the warhead to exert an ADC's cytotoxic effects on cancer cells intracellularly. A simple and highly sensitive workflow is developed and validated for the simultaneous quantification of six ADC payloads, namely SN-38, MTX, DXd, MMAE, MMAF and Calicheamicin (CM). The workflow consists of a short and simple sample extraction using a methanol-ethanol mixture, followed by a fast liquid chromatography tandem mass spectrometry (LC-MS/MS) analysis. The results showed that well-validated linear response ranges of 0.4-100 nM for SN38, MTX and DXd, 0.04-100 nM for MMAE and MMAF, 0.4-1000 nM for CM were achieved in mouse serum. Recoveries for all six payloads at three different concentrations (low, medium and high) were more than 85%. An ultra-low sample volume of only 5 µL of serum is required due to the high sensitivity of the method. This validated method was successfully applied to a pharmacokinetic study to quantify MMAE in mouse serum samples.


Assuntos
Imunoconjugados , Espectrometria de Massas em Tandem , Animais , Camundongos , Cromatografia Líquida/métodos , Imunoconjugados/farmacocinética , Imunoconjugados/química , Espectrometria de Massas em Tandem/métodos , Fluxo de Trabalho , Espectrometria de Massa com Cromatografia Líquida
2.
Sci Rep ; 8(1): 11608, 2018 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-30072783

RESUMO

Monoclonal antibodies (mAbs) are used as targeted therapies against cancers. These mAbs kill cancer cells via various mechanisms of actions. In this study, human embryonic stem cells (hESCs) was used as the immunogen to generate a panel of antibodies. From this panel of mAbs, A19 was found to bind both hESC and various cancer cell lines. The antigen target of A19 was identified as Erbb-2 and glycan analysis showed that A19 binds to a N-glycan epitope on the antigen. A19 was elucidated to internalize into cancer cells following binding to Erbb-2 and hence developed as an antibody-drug conjugate (ADC). Using ADC as the mechanism of action, A19 was able to kill cancer cells in vitro and delayed the onset of tumour formation in mice xenograft model. When compared to Herceptin, A19 binds to different isoforms of Erbb-2 and does not compete with Herceptin for the same epitope. Hence, A19 has the potential to be developed as an alternative targeted therapeutic agent for cancers expressing Erbb-2.


Assuntos
Anticorpos Monoclonais Murinos , Antígenos de Neoplasias/imunologia , Antineoplásicos Imunológicos/farmacologia , Células-Tronco Embrionárias Humanas/imunologia , Neoplasias Experimentais , Animais , Anticorpos Monoclonais Murinos/imunologia , Anticorpos Monoclonais Murinos/farmacologia , Antineoplásicos Imunológicos/imunologia , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Oncotarget ; 9(17): 13206-13221, 2018 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-29568351

RESUMO

Monoclonal antibodies (mAbs) play an increasingly important role in cancer therapy. To address the wide heterogeneity of the disease, the identification of novel antigen targets and the development of mAbs against them are needed. Our lab previously generated a panel of mAbs against human embryonic stem cells (hESC) using a whole cell immunization approach in mice. These mAbs can potentially target oncofetal antigens and be repurposed for antibody or antibody drug conjugate (ADC) therapy. From this panel, the novel IgG1 2448 was found to bind surface antigens on hESC and multiple cancer cell lines. Here, we show 2448 targets a unique glycan epitope on annexin A2 (ANXA2) and can potentially monitor the Epithelial-Mesenchymal Transition (EMT) in ovarian and breast cancer. To evaluate 2448 as a potential drug, 2448 was engineered and expressed as a chimeric IgG1. Chimeric 2448 (ch2448) demonstrated efficient and specific killing when conjugated to cytotoxic payloads as an ADC. In addition, ch2448 elicited potent antibody-dependent cell-mediated cytotoxicity (ADCC) activity in vitro and in vivo. Further engineering of ch2448 to remove fucose in the Fc domain enhanced ADCC. Overall, these findings indicate that embryonic ANXA2 is an attractive target and suggest that ch2448 is a promising candidate for further therapeutic development.

4.
Tissue Eng Part A ; 18(9-10): 899-909, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22092252

RESUMO

One of the most pertinent concerns of using differentiated cells derived from human embryonic stem cells (hESC) is the presence of residual undifferentiated hESC, because they carry a risk of teratoma formation. A new cell-cell separation approach that eliminates teratoma-forming hESC in order to ensure safer cell therapy was developed. By combining antibodies (IgMs or IgGs) for the selective removal of undifferentiated hESC using magnetic activated cell sorting (MACS) followed by selective killing of residual hESC with the unique cytotoxic antibody mAb 84, the required purity of differentiated hESC can be achieved. The applicability and robustness of this separation strategy is shown here in a case study using pools of undifferentiated hESC and human fibroblast cells at different ratios (5%-50% hESC) to reflect the different scenario of contaminating hESC in a differentiated cell population. Notably, 97.2%-99.7% of the hESC were removed after the MACS step and 99.1%-100%, after the mAb 84 treatment step, which was confirmed by double-staining flow cytometry and RT-qPCR analysis. These in vitro findings were further validated in an in vivo severe combined immunodeficiency (SCID) mouse model. Importantly, we observed the absence of teratoma formation in eight out of nine SCID mice 28 weeks postinjection of cells after the MACS step, whereas teratomas were observed in all of the controls. Thus, the combination of MACS with the unique cytotoxic antibody mAb 84 constitutes an indispensible tool for successful and safe cell therapy.


Assuntos
Diferenciação Celular/fisiologia , Separação Celular/métodos , Células-Tronco Embrionárias/citologia , Adsorção , Animais , Linhagem Celular , Sobrevivência Celular/fisiologia , Feminino , Citometria de Fluxo , Humanos , Camundongos , Camundongos SCID
5.
Stem Cells ; 27(8): 1792-801, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19544435

RESUMO

The monoclonal antibody mAb 84, which binds to podocalyxin-like protein-1 (PODXL) on human embryonic stem cells (hESCs), was previously reported to bind and kill undifferentiated cells in in vitro and in vivo assays. In this study, we investigate the mechanism responsible for mAb 84-induced hESCs cytotoxicity. Apoptosis was likely not the cause of mAb 84-mediated cell death because no elevation of caspase activities or increased DNA fragmentation was observed in hESCs following incubation with mAb 84. Instead, it was preceded by cell aggregation and damage to cell membranes, resulting in the uptake of propidium iodide, and the leakage of intracellular sodium ions. Furthermore, examination of the cell surface by scanning electron microscopy revealed the presence of pores on the cell surface of mAb 84-treated cells, which was absent from the isotype control. This mechanism of cell death resembles that described for oncosis, a form of cell death resulting from membrane damage. Additional data suggest that the binding of mAb 84 to hESCs initiates a sequence of events prior to membrane damage, consistent with oncosis. Degradation of actin-associated proteins, namely, alpha-actinin, paxillin, and talin, was observed. The perturbation of these actin-associated proteins consequently permits the aggregation of PODXL, thus leading to the formation of pores. To our knowledge, this is the first report of oncotic cell death with hESCs as a model.


Assuntos
Anticorpos Monoclonais/farmacologia , Células-Tronco Embrionárias/efeitos dos fármacos , Actinina/metabolismo , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Agregação Celular/efeitos dos fármacos , Agregação Celular/imunologia , Morte Celular/efeitos dos fármacos , Morte Celular/imunologia , Diferenciação Celular/imunologia , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/imunologia , Proteínas do Citoesqueleto/análise , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Modelos Animais de Doenças , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/imunologia , Humanos , Camundongos , Camundongos SCID , Microscopia Eletrônica de Varredura , Paxilina/metabolismo , Sialoglicoproteínas/metabolismo , Talina/metabolismo , Teratoma/imunologia , Teratoma/prevenção & controle , Vinculina/metabolismo
6.
Stem Cells ; 26(6): 1454-63, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18356574

RESUMO

Future therapeutic applications of differentiated human embryonic stem cells (hESC) carry a risk of teratoma formation by contaminating undifferentiated hESC. We generated 10 monoclonal antibodies (mAbs) against surface antigens of undifferentiated hESC, showing strong reactivity against undifferentiated, but not differentiated hESC. The mAbs did not cross react with mouse fibroblasts and showed weak to no reactivity against human embryonal carcinoma cells. Notably, one antibody (mAb 84) is cytotoxic to undifferentiated hESC and NCCIT cells in a concentration-dependent, complement-independent manner. mAb 84 induced cell death of undifferentiated, but not differentiated hESC within 30 minutes of incubation, and immunoprecipitation of the mAb-antigen complex revealed that the antigen is podocalyxin-like protein-1. Importantly, we observed absence of tumor formation when hESC and NCCIT cells were treated with mAb 84 prior to transplantation into severe combined immunodeficiency mice. Our data indicate that mAb 84 may be useful in eliminating residual hESC from differentiated cells populations for clinical applications. Disclosure of potential conflicts of interest is found at the end of this article.


Assuntos
Células-Tronco Embrionárias/citologia , Sialoglicoproteínas/análise , Animais , Anticorpos , Anticorpos Monoclonais , Diferenciação Celular , Linhagem Celular , Sobrevivência Celular , Células-Tronco Embrionárias/fisiologia , Citometria de Fluxo , Células HeLa , Humanos , Camundongos , Sialoglicoproteínas/imunologia
7.
J Biotechnol ; 130(3): 320-8, 2007 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-17544536

RESUMO

The maintenance of undifferentiated human embryonic stem cells (hESC) requires feeder cells, either in co-culture or feeder-free with conditioned medium (CM) from the feeders. In this study, we compared the CM of a supporting primary mouse embryonic feeder (MEF) and an isogenic but non-supporting MEF line (DeltaE-MEF) in order to gain an insight to the differential expression profile of secreted factors. Using two-dimensional electrophoresis (2-DE) and matrix-assisted laser desorption/ionization-time of flight (MALDI) tandem mass spectrometry, 13 protein identities were found to be downregulated in DeltaE-MEF compared to MEF, of which 4 were found to be soluble factors and 3 proteins were membrane-associated or related to the extracellular matrix. In addition, four other proteins were identified to be differentially expressed in MEF-CM using high pressure liquid chromatography (HPLC) and cytokine arrays. In functional experiments where CM was replaced with six of the factors identified, hESC were able to proliferate for five continuous passages whilst maintaining 68-82% and 74-98% expression of pluripotent markers, Oct-4 and Tra-1-60, respectively. Using proteomic tools, important proteins from CM that supports hESC culture have been identified, which when replaced with recombinant proteins, continue to support undifferentiated hESC growth in a feeder-free culture platform.


Assuntos
Células-Tronco Embrionárias/metabolismo , Proteínas/análise , Animais , Células Cultivadas , Meios de Cultivo Condicionados , Meios de Cultura Livres de Soro , Citocinas/metabolismo , Eletroforese em Gel Bidimensional , Células-Tronco Embrionárias/citologia , Fibroblastos/citologia , Citometria de Fluxo , Perfilação da Expressão Gênica , Humanos , Camundongos , Fatores de Transcrição de Octâmero/metabolismo , Proteínas/genética , Proteínas Recombinantes/metabolismo
8.
Biotechnol Bioeng ; 91(5): 523-33, 2005 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-16044469

RESUMO

Embryonic stem cells (ESC) have the unique ability to differentiate into a variety of tissue types. However, the realization of regenerative medicine will require the production of large quantities of ESC which subsequently have to be differentiated into the final phenotype. Thus, we sought to develop a simple and scaleable bioprocess to increase densities of ESC to achieve this goal. Using mouse embryonic stem cells (mESC) as a model, by combining automated feeding and culture of mESC on petriperm dishes, cell densities were enhanced up to 6.4 x 10(6) cells/cm2 compared to conventional petri dish culture which only reached 0.2 to 1.4 x 10(6) cells/cm2. It was found that mESC from all experiments maintained excellent viability, pluripotency, and genetic stability after growing for 6 days in petriperm cultures with automated feeding. The expression of Oct-4 transcription factor was observed in all cultures, mESC formed embryoid bodies in differentiated cultures and teratomas in SCID mice, confirming their pluripotency, and karyotype of the cultures was normal. This culture method was stable for routine passaging and a second mESC cell line was shown to perform in a similar manner on petriperm with automated feeding. This work represents an important step towards achieving high density cultures of ESC.


Assuntos
Técnicas de Cultura de Células/métodos , Embrião de Mamíferos/citologia , Células-Tronco/citologia , Fosfatase Alcalina/metabolismo , Animais , Biomarcadores/metabolismo , Reatores Biológicos , Ciclo Celular , Diferenciação Celular , Linhagem Celular , Sobrevivência Celular , Células Cultivadas , Proteínas de Ligação a DNA/metabolismo , Citometria de Fluxo , Corantes Fluorescentes , Indóis , Cariotipagem , Antígenos CD15/metabolismo , Masculino , Camundongos , Camundongos SCID , Fator 3 de Transcrição de Octâmero , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/fisiologia , Transplante de Células-Tronco , Células-Tronco/metabolismo , Teratoma/patologia , Fatores de Transcrição/metabolismo
9.
Bioprocess Biosyst Eng ; 27(6): 381-387, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15928928

RESUMO

Human embryonic stem cells (hESC) are self-renewing pluripotent cells capable of differentiating into cells representative of all three embryonic germ layers. Hence, they hold great potential for regenerative medicine. However, significant cell numbers are required to fulfill their potential therapeutic applications. In this study, perfusion with supplemented conditioned media (SCM), produced by mouse embryonic fibroblasts (MEF), was adopted to improve cell densities of hESC cultures. Perfusion enhanced hESC numbers by 70% compared to static conditions, on both organ culture dish (OCD) and petri dish cultures. All cultures maintained healthy expression of the pluripotent marker, Oct-4 transcription factor. In vivo, perfused hESC formed teratomas in severe combined immunodeficiency (SCID) mice models that represent the three embryonic germ layers. When SCM was produced with lower concentrations of MEF, hESC densities and Oct-4 levels were reduced. Hence, perfusion with SCM is a potential feeding method for scale-up production of hESC.


Assuntos
Reatores Biológicos , Técnicas de Cultura de Células/métodos , Técnicas de Cocultura/métodos , Perfusão/métodos , Células-Tronco/citologia , Células-Tronco/fisiologia , Engenharia Tecidual/métodos , Animais , Técnicas de Cultura de Células/instrumentação , Linhagem Celular , Proliferação de Células , Sobrevivência Celular/fisiologia , Meios de Cultivo Condicionados/metabolismo , Humanos , Camundongos , Perfusão/instrumentação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...