Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 13(1): 12130, 2023 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-37495742

RESUMO

The in-depth analysis of the ADME profiles of drug candidates using in vitro models is essential for drug development since a drug's exposure in humans depends on its ADME properties. In contrast to efforts in developing human in vitro absorption models, only a limited number of studies have explored models using rats, the most frequently used species in in vivo DMPK studies. In this study, we developed a monolayer model with an effective barrier function for ADME assays using rat duodenal organoids as a cell source. At first, we developed rat duodenal organoids according to a previous report, but they were not able to generate a confluent monolayer. Therefore, we modified organoid culture protocols and developed cyst-enriched organoids; these strongly promoted the formation of a confluent monolayer. Furthermore, adding valproic acid to the culture accelerated the differentiation of the monolayer, which possessed an effective barrier function and apicobasal cell polarity. Drug transporter P-gp function as well as CYP3A activity and nuclear receptor function were confirmed in the model. We expect our novel monolayer model to be a useful tool for elucidating drug absorption processes in detail, enabling the development of highly absorbable drugs.


Assuntos
Organoides , Ratos , Humanos , Animais , Diferenciação Celular
2.
Sci Rep ; 11(1): 24305, 2021 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-34934075

RESUMO

Tumor nests in lung squamous cell carcinoma (LUSC) have a hierarchical structure resembling squamous epithelium. The nests consist of basal-like cells on the periphery and layers of keratinocyte-like cells that differentiate towards the center of the nest, forming keratin pearls. Reproducing this spatial heterogeneity in in vitro models would be useful for understanding the biology of LUSC. Here, we established a three-dimensional (3D) culture model with a squamous epithelial structure using LUSC cell lines PLR327F-LD41 and MCC001F, established in-house. When PLR327F-LD41 cells were cultured in a mixture of Matrigel and collagen I, they generated 3D colonies (designated cancer organoids, or COs) with involucrin (IVL)-positive keratinizing cells in the center (IVLinner COs). COs with uniform size were generated by seeding PLR327F-LD41 cells in a form of small cell aggregates. Since Notch signaling induces the differentiation of squamous epithelium, we confirmed the effect of γ-secretase inhibitor in inhibiting Notch signaling in IVLinner COs. Surprisingly, γ-secretase inhibitor did not block induction of IVL-positive cells; however, cells residing between the CK5-positive basal-like layer and IVL-positive layer decreased significantly. Thus, our 3D culture model with uniform size and structure promises to be a useful tool for elucidating the biology of LUSC and for screening drug-candidates.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Técnicas de Cultura de Células , Neoplasias Pulmonares/metabolismo , Modelos Biológicos , Animais , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Feminino , Humanos , Neoplasias Pulmonares/patologia , Camundongos
3.
Lab Invest ; 101(1): 12-25, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32728120

RESUMO

New cancer characteristics can be discovered by focusing on the process of tumor formation. Cancer stem cells (CSCs) are a key subpopulation, as they are theorized to be at the apex of the tumor hierarchy. We can better understand their function in the tumor hierarchy by using sectioned samples to observe the growth of tumors from their origins as CSCs. In this study, we evaluated the growth of moderate differentiated colorectal cancer from LGR5-positive cells, which is a CSC marker of colorectal cancer, using xenograft and three-dimensional culture models spatiotemporally. These cells express LGR5 at high levels and show CSC phenotypes. To detect them, we used a previously generated antibody that specifically targets LGR5, and were therefore able to observe LGR5-positive cells aggregating into small clusters (sCLs) over the course of tumor growth. Because these LGR5-expressing sCLs formed continuously during growth mainly in the invasive front, we concluded that the structure must contribute significantly to the expansion of CSCs and to tumor growth overall. We confirmed the formation of sCLs from gland structures using a three-dimensional culture model. In addition, sCLs exhibited upregulated genes related to stress response and partial/hybrid epithelial-mesenchymal transition (EMT), as well as genes reported to be prognosis factors. Finally, sCLs with high LGR5 expression were identified in clinical samples. Based on these results, we elucidate how sCLs are an important contributors to tumor growth and the expansion of CSCs.


Assuntos
Neoplasias Colorretais/patologia , Células-Tronco Neoplásicas/fisiologia , Receptores Acoplados a Proteínas G/metabolismo , Adenocarcinoma/patologia , Animais , Linhagem Celular Tumoral , Técnicas de Cocultura , Colo/patologia , Neoplasias Colorretais/metabolismo , Transição Epitelial-Mesenquimal , Fibroblastos , Humanos , Camundongos , Transplante de Neoplasias , Neoplasias Experimentais
4.
J Toxicol Pathol ; 33(2): 67-76, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32425339

RESUMO

Desmoglein-3 (DSG3) is a potential target of cytotoxic antibody therapy for squamous cell carcinomas but is also expressed in various normal squamous epithelia. We obtained information about DSG3 distribution in mouse tissues by immunohistochemistry and conducted an intravenous multiple-dose study in mouse to estimate the toxic potential of anti-DSG3 therapy. DSG3 was expressed in the squamous epithelium of several organs including the skin, esophagus, tongue, forestomach, eye, and vagina. It was expressed at all estrous cycles of the vagina with changes in distribution patterns along with the structural changes in each cycle, and expression was reduced in ovariectomized (OVX) mice. On the administration of the antibody, there was disarrangement of the vaginal mucosal epithelium with formation of miroabscess, increased granulocyte infiltration, and single cell necrosis. Despite similar expression levels of DSG3 in other tissues, histopathological changes were limited to the vagina. The severity of the changes was reduced by ovariectomy. From these findings, the lesions were thought to be related to the drastic change in the histological structure of the vaginal mucosa accompanying the estrous cycle. Thus, we have shown that the changing expression of target antigen distribution and its relationship with physiological changes in tissue structure are important features for estimating the toxic potential of cytotoxic antibody therapy.

5.
Sci Rep ; 10(1): 3156, 2020 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-32081957

RESUMO

Colorectal cancer demonstrates intra-tumour heterogeneity formed by a hierarchical structure comprised of cancer stem cells (CSCs) and their differentiated progenies. The mechanism by which CSCs are maintained and differentiated needs to be further elucidated, and there is evidence that the tumour microenvironment governs cancer stemness. Using PLR123, a colon cancer cell line with CSC properties, we determined the culture conditions necessary to establish a pair of three-dimensional (3D) culture models grown in Matrigel, designated stemCO and diffCO. The conditions were determined by comparing the phenotypes in the models with PLR123 mouse xenografts colonising lung and liver. StemCO resembled LGR5-positive undifferentiated tumours in the lung, and diffCO had lumen structures composed of polarised cells that were similar to the ductal structures found in differentiated tumours in the liver. In a case using the models for biomedical research, treatment with JAG-1 peptide or a γ-secretase inhibitor modified the Notch signaling and induced changes indicating that the signal participates in lumen formation in the models. Our results demonstrate that culture conditions affect the stemness of 3D culture models generated from CSCs and show that comparing models with different phenotypes is useful for studying how the tumour environment regulates cancer.


Assuntos
Técnicas de Cultura de Células , Neoplasias do Colo/patologia , Células-Tronco Neoplásicas/citologia , Microambiente Tumoral , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Animais , Diferenciação Celular , Linhagem Celular Tumoral , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hepáticas/secundário , Neoplasias Pulmonares/secundário , Camundongos , Transplante de Neoplasias , Fenótipo , Transdução de Sinais
6.
J Biochem ; 164(6): 471-481, 2018 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-30239818

RESUMO

It is ideal for the target antigen of a cytotoxic therapeutic antibody against cancer to be cancer-specific, but such antigens are rare. Thus an alternative strategy for target selection is necessary. Desmoglein 3 (DSG3) is highly expressed in lung squamous cell carcinoma, while it is well-known that anti-DSG3 antibodies cause pemphigus vulgaris, an autoimmune disease. We evaluated DSG3 as a novel target by selecting an epitope that exerts efficacy against cancer with no pathogenic effects in normal tissues. Pathogenic anti-DSG3 antibodies induce skin blisters by inhibiting the cell-cell interaction in a Ca2+-dependent manner. We screened anti-DSG3 antibodies that bind DGS3 independent of Ca2+ and have high antibody-dependent cell cytotoxicity (ADCC) activity against DSG3-expressing cells. These selected antibodies did not inhibit cell-cell interaction and showed ADCC activity against squamous cell carcinoma cell lines. Furthermore, one of the DSG3 antibodies showed anti-tumour activity in tumour mouse models but did not induce adverse effects such as blister formation in the skin. Thus it was possible to generate an antibody against DSG3 by using an appropriate epitope that retained efficacy with no pathogenicity. This approach of epitope selection may expand the variety of druggable target molecules.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos Imunológicos/uso terapêutico , Carcinoma de Células Escamosas/tratamento farmacológico , Desmogleína 3/antagonistas & inibidores , Desenho de Fármacos , Queratinócitos/efeitos dos fármacos , Animais , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/farmacologia , Especificidade de Anticorpos , Antineoplásicos Imunológicos/efeitos adversos , Antineoplásicos Imunológicos/farmacologia , Células CHO , Carcinoma de Células Escamosas/imunologia , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cricetulus , Desmogleína 3/química , Desmogleína 3/genética , Desmogleína 3/metabolismo , Epitopos/química , Epitopos/genética , Epitopos/metabolismo , Humanos , Queratinócitos/imunologia , Queratinócitos/metabolismo , Queratinócitos/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos SCID , Camundongos Transgênicos , Fragmentos de Peptídeos/antagonistas & inibidores , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
7.
J Toxicol Pathol ; 31(1): 81-85, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29479145

RESUMO

In vitro-cultured 3D structures called organoids have become important tools for biological research, but there is little information concerning simple and efficient methods to evaluate organoid morphology. To address this issue, we attempted to establish a simple method by applying conventional histopathology that enables observation of multiple organoids on a single cross section, maintains good morphology, and is applicable to various histopathological stains. By centrifugation in unsolidified agarose solution, we were able to accumulate the organoids onto a single plane. The morphology was well preserved, and the various morphological types and sizes of organoid structures were identified. This method was also applicable for special staining, immunohistochemistry, and immunofluorescence staining. This method makes it possible to utilize the advantages of conventional pathological methods when studying organoids.

8.
J Biochem ; 161(4): 361-368, 2017 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-28013222

RESUMO

Leucine-rich repeat-containing G protein-coupled receptor 6 (LGR6) is a seven-pass transmembrane protein known to be a marker of stem cells in several organs. To deepen our understanding of the cell biology of LGR6-positive cells, including stem cells, we generated monoclonal antibodies (mAbs) against human LGR6. DNA immunization followed by whole-cell immunization with LGR6-expressing transfectants was performed to obtain mAbs that recognized the native form of LGR6. Hybridomas were screened by flow cytometry using LGR6-transfected cells. Because the molecules of LGR4, LGR5, and LGR6 are 50% homologous at the amino acid level, specificity of the mAbs was confirmed by transfectants expressing LGR4, LGR5, or LGR6. Three LGR6-specific mAbs were generated. Two of the three mAbs (designated 43A6 and 43D10) recognized the large N-terminal extracellular domain of LGR6, and competitively blocked the binding of R-spondin 1, which is known to be the ligand for LGR6. The other mAb, 43A25, recognized the seven-pass transmembrane domain of LGR6, and was able to be used for immunoblot analysis. In addition, mAbs 43A6 and 43D10 detected endogenous expression of LGR6 in cancer cell lines. We expect that our mAbs will contribute to widening our understanding of LGR6-positive cells in humans.


Assuntos
Anticorpos Monoclonais/imunologia , Afinidade de Anticorpos/imunologia , Especificidade de Anticorpos/imunologia , Receptores Acoplados a Proteínas G/imunologia , Animais , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/metabolismo , Ligação Competitiva , Linhagem Celular , Linhagem Celular Tumoral , Citometria de Fluxo , Humanos , Hibridomas , Immunoblotting , Camundongos Endogâmicos BALB C , Ligação Proteica , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Trombospondinas/metabolismo
9.
PLoS One ; 8(12): e83905, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24386306

RESUMO

The immunomodulatory drug lenalidomide (Len) has drawn attention to potentiate antibody-dependent cellular cytotoxicity (ADCC)-mediated immunotherapies. We developed the defucosylated version (YB-AHM) of humanized monoclonal antibody against HM1.24 (CD317) overexpressed in multiple myeloma (MM) cells. In this study, we evaluated ADCC by YB-AHM and Len in combination against MM cells and their progenitors. YB-AHM was able to selectively kill via ADCC MM cells in bone marrow samples from patients with MM with low effector/target ratios, which was further enhanced by treatment with Len. Interestingly, Len also up-regulated HM1.24 expression on MM cells in an effector-dependent manner. HM1.24 was found to be highly expressed in a drug-resistant clonogenic "side population" in MM cells; and this combinatory treatment successfully reduced SP fractions in RPMI 8226 and KMS-11 cells in the presence of effector cells, and suppressed a clonogenic potential of MM cells in colony-forming assays. Collectively, the present study suggests that YB-AHM and Len in combination may become an effective therapeutic strategy in MM, warranting further study to target drug-resistant MM clonogenic cells.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Antígenos CD/imunologia , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Talidomida/análogos & derivados , Idoso , Idoso de 80 Anos ou mais , Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Monoclonais Humanizados/metabolismo , Antígenos CD/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica , Linhagem Celular Tumoral , Sinergismo Farmacológico , Feminino , Proteínas Ligadas por GPI/imunologia , Proteínas Ligadas por GPI/metabolismo , Glicosilação , Humanos , Imunoterapia , Lenalidomida , Masculino , Pessoa de Meia-Idade , Células-Tronco Neoplásicas/imunologia , Células-Tronco Neoplásicas/patologia , Células da Side Population/efeitos dos fármacos , Células da Side Population/patologia , Talidomida/farmacologia , Regulação para Cima/efeitos dos fármacos
10.
Cancer Sci ; 101(10): 2227-33, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20701608

RESUMO

The humanized monoclonal antibody (mAb) against CD317 antigen (anti-HM1.24 antibody; AHM), which is highly expressed on multiple myeloma (MM), induces antibody-dependent cellular cytotoxicity (ADCC). However, the antitumor activity of AHM in the clinical setting has not been clearly demonstrated. In this study, we produced defucosylated AHM and evaluated its potency for clinical application by performing autologous ADCC assays against primary MM cells from patients. Defucosylated AHM that was produced in rat myeloma YB2/0 cells expressing a low level of fucosyltransferase (FUT8) showed significant ADCC activity against three out of six primary MM cells in the presence of autologous PBMC, whereas conventional AHM did not. The results indicate that the potency of AHM to induce ADCC against primary MM cells was insufficient, but was significantly augmented by defucosylation. To generate more homogenous defucosylated monoclonal antibodies (mAb) for fermentation, we disrupted the GFT gene that encodes a GDP-fucose transporter in a CHO/DXB11 cell line by sequential homologous recombination. Analysis of the N-linked oligosaccharide in the defucosylated AHM produced by the established GFT(-/-)CHO cell line showed that a majority (93.4%) of the oligosaccharide was fucose free. The GFT(-/-) cells stably produced defucosylated mAb over passages. These results demonstrate that GTF(-/-)CHO-produced defucosylated AHM (GFTKO-AHM) will be a promising new therapeutic antibody against MM in the clinical setting.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Citotoxicidade Celular Dependente de Anticorpos , Antígenos CD/imunologia , Glicoproteínas de Membrana/imunologia , Mieloma Múltiplo/tratamento farmacológico , Animais , Células CHO , Cricetinae , Cricetulus , Proteínas Ligadas por GPI , Glucosiltransferases/fisiologia , Humanos , Proteínas de Transporte de Monossacarídeos/fisiologia , Mieloma Múltiplo/imunologia
11.
Cancer Sci ; 99(12): 2461-6, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19032371

RESUMO

A murine (mAHM) and a humanized (AHM) monoclonal antibody against CD317 (also called tetherin, BST2, or HM1.24 antigen), expressed preferentially in neoplastic B cells such as multiple myeloma, exhibited antitumor effects as a result of antibody-dependent cellular cytotoxicity (ADCC). The putative interferon (IFN) response elements IRF-1/2 and ISGF3 are present in the promoter of the CD317 gene, and IFN has been used for the treatment of not only myeloproliferative diseases but also solid tumors such as renal cell carcinoma (RCC) and melanoma. Therefore, we examined the effects of IFN on the expression of CD317 and on the antitumor activity of AHM and mAHM in RCC and melanoma. Flow cytometry and in vitro ADCC assays with human or mouse effector cells demonstrated that IFN-alpha markedly increased the amount of cell surface CD317 and augmented the ADCC activity of mAHM and AHM in RCC cells and to a lesser extent in melanoma cells. Administration of IFN-alpha to mice bearing RCC xenografts also increased the expression of CD317 in tumor cells. When coadministered with IFN-alpha, mAHM exhibited more profound antitumor activity in both IFN-alpha-sensitive and -insensitive RCC xenograft models. Thus, AHM in combination with IFN-alpha may be an effective therapy for the treatment of RCC.


Assuntos
Anticorpos Anti-Idiotípicos/farmacologia , Antígenos CD/imunologia , Carcinoma de Células Renais/terapia , Interferon-alfa/farmacologia , Neoplasias Renais/terapia , Glicoproteínas de Membrana/imunologia , Animais , Anticorpos Anti-Idiotípicos/imunologia , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Antígenos CD/genética , Carcinoma de Células Renais/imunologia , Linhagem Celular Tumoral , Fluoresceína-5-Isotiocianato/metabolismo , Corantes Fluorescentes/metabolismo , Proteínas Ligadas por GPI , Humanos , Imuno-Histoquímica , Interferon-alfa/imunologia , Neoplasias Renais/imunologia , Masculino , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Nus , Camundongos SCID , Carga Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Clin Cancer Res ; 13(9): 2745-50, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17473208

RESUMO

PURPOSE: L612, a human IgM monoclonal antibody produced by an EBV-transformed human B-cell line, binds to ganglioside GM3 and kills GM3-positive human melanoma cells in the presence of complement. It has been shown to be effective in some patients with late-stage melanoma. L612 consists of hexameric IgM (about 20%), pentameric IgM (about 74%), and other minor IgM molecules. Because hexameric IgM activates complement more effectively than pentameric IgM, we developed and evaluated a hexamer-dominant recombinant IgM for clinical applications. EXPERIMENTAL DESIGN: Chinese hamster ovary (CHO) cells were transfected with heavy- and light-chain genes of L612, with or without the joining-chain gene. Antitumor effects of the recombinant IgM secreted from CHO cells were evaluated in vitro and in vivo. RESULTS: Recombinant IgM secreted from CHO cells without the joining chain (designated CA19) was approximately 80% hexameric, whereas recombinant IgM from CHO cells transfected with heavy-, light-, and joining-chain genes (designated CJ45) was about 90% pentameric. Both CA19 and CJ45 recombinant IgMs caused complement-dependent cytotoxicity against human and mouse melanoma cell lines, but the amount of CA19 required for 50% specific cytotoxicity was 5 to 10 times smaller. I.v. injection of CA19 compared with CJ45 or native L612 elicited more profound antitumor activity in nude rats bearing a GM3-positive mouse melanoma xenograft. CONCLUSIONS: A hexamer-dominant human IgM against GM3 may provide a more potent treatment option for patients with GM3-positive melanoma.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Gangliosídeo G(M3)/imunologia , Melanoma/tratamento farmacológico , Neoplasias Cutâneas/tratamento farmacológico , Animais , Anticorpos Monoclonais Humanizados , Células CHO , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Humanos , Melanoma/química , Melanoma/patologia , Proteínas Recombinantes/uso terapêutico , Neoplasias Cutâneas/química , Neoplasias Cutâneas/patologia
13.
Cancer Res ; 67(3): 1184-92, 2007 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-17283154

RESUMO

Cross-linked human leukocyte antigen (HLA) class I molecules have been shown to mediate cell death in neoplastic lymphoid cells. However, clinical application of an anti-HLA class I antibody is limited by possible side effects due to widespread expression of HLA class I molecules in normal tissues. To reduce the unwanted Fc-mediated functions of the therapeutic antibody, we have developed a recombinant single-chain Fv diabody (2D7-DB) specific to the alpha2 domain of HLA-A. Here, we show that 2D7-DB specifically induces multiple myeloma cell death in the bone marrow environment. Both multiple myeloma cell lines and primary multiple myeloma cells expressed HLA-A at higher levels than normal myeloid cells, lymphocytes, or hematopoietic stem cells. 2D7-DB rapidly induced Rho activation and robust actin aggregation that led to caspase-independent death in multiple myeloma cells. This cell death was completely blocked by Rho GTPase inhibitors, suggesting that Rho-induced actin aggregation is crucial for mediating multiple myeloma cell death. Conversely, 2D7-DB neither triggered Rho-mediated actin aggregation nor induced cell death in normal bone marrow cells despite the expression of HLA-A. Treatment with IFNs, melphalan, or bortezomib enhanced multiple myeloma cell death induced by 2D7-DB. Furthermore, administration of 2D7-DB resulted in significant tumor regression in a xenograft model of human multiple myeloma. These results indicate that 2D7-DB acts on multiple myeloma cells differently from other bone marrow cells and thus provide the basis for a novel HLA class I-targeting therapy against multiple myeloma.


Assuntos
Antígenos HLA-A/imunologia , Imunização Passiva/métodos , Fragmentos de Imunoglobulinas/farmacologia , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/terapia , Animais , Células da Medula Óssea/imunologia , Células da Medula Óssea/patologia , Morte Celular/imunologia , Linhagem Celular Tumoral , Citocinas/farmacologia , Ativação Enzimática , Antígenos HLA-A/biossíntese , Humanos , Células Jurkat , Camundongos , Camundongos SCID , Mieloma Múltiplo/enzimologia , Mieloma Múltiplo/patologia , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas rho de Ligação ao GTP/metabolismo
14.
Clin Cancer Res ; 12(11 Pt 1): 3257-64, 2006 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-16740745

RESUMO

PURPOSE: Hepatocellular carcinoma is the most common primary malignancy of the liver and accounts for as many as one million deaths annually worldwide. The present study was done to identify new transmembrane molecules for antibody therapy in hepatocellular carcinoma. EXPERIMENTAL DESIGN: Gene expression profiles of pooled total RNA from three tissues each of moderately differentiated and poorly differentiated hepatocellular carcinoma were compared with those of normal liver, noncancerous liver tissue in hepatocellular carcinoma patients, 30 normal tissue samples, and five fetal tissue samples. Target genes up-regulated specifically in hepatocellular carcinoma were validated by immunohistochemical analysis and complement-dependent cytotoxicity assay using monoclonal antibodies generated against target molecules. RESULTS: The human homologue of the Drosophila Roundabout gene, axon guidance receptor homologue 1, ROBO1/DUTT1, a member of the immunoglobulin superfamily, was highly expressed in hepatocellular carcinoma, whereas it showed only a limited distribution in normal tissues. On immunohistochemical analysis using a newly generated anti-ROBO1 monoclonal antibody, positive signals were observed in 83 of 98 cases of hepatocellular carcinoma (84.7%). The mAb B2318C induced complement-dependent cytotoxicity in ROBO1-expressing cell lines and in the liver cancer cell line PLC/PRF/5. Strikingly, the ectodomain of ROBO1 was detected not only in the culture medium of liver cancer cell lines (PLC/PRF/5, HepG2, etc.) but also in sera from hepatocellular carcinoma patients (6 of 11). CONCLUSIONS: This is the first report that ROBO1 is overexpressed in hepatocellular carcinoma and shed into serum in humans. These observations suggest that ROBO1 is a potential new serologic marker for hepatocellular carcinoma and may represent a new therapeutic target.


Assuntos
Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Proteínas do Tecido Nervoso/genética , Receptores Imunológicos/genética , Animais , Anticorpos Monoclonais/farmacologia , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/sangue , Biomarcadores Tumorais/genética , Células COS , Carcinoma Hepatocelular/sangue , Carcinoma Hepatocelular/diagnóstico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Chlorocebus aethiops , Testes Imunológicos de Citotoxicidade , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Neoplasias Hepáticas/sangue , Neoplasias Hepáticas/diagnóstico , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/sangue , Estrutura Terciária de Proteína , RNA Mensageiro/biossíntese , Receptores Imunológicos/biossíntese , Receptores Imunológicos/sangue , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Regulação para Cima/genética , Proteínas Roundabout
15.
Leuk Res ; 30(8): 949-56, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16473407

RESUMO

A humanized monoclonal antibody (mAb) against HM1.24 (AHM) caused antibody-dependent cellular cytotoxicity (ADCC) against multiple myeloma (MM) cells. Here, we constructed a conventional non-radioisotope method that quantifies the amount of HM1.24 using fluorescein-labeled AHM. More than 10(4) molecules/cell of HM1.24 were detected in 12 out of 14 patients' MM cells, and a linear correlation was found between ADCC by AHM and the amounts of HM1.24. Thus, AHM is likely to be more efficacious against MM cells with high levels of HM1.24. This conventional non-RI method to quantify HM1.24 will be useful to select patients most likely to respond to AHM.


Assuntos
Anticorpos Monoclonais/imunologia , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Antígenos CD/análise , Glicoproteínas de Membrana/análise , Mieloma Múltiplo/imunologia , Reações Antígeno-Anticorpo , Antígenos CD/imunologia , Linhagem Celular Tumoral , Fluoresceína/química , Proteínas Ligadas por GPI , Humanos , Leucócitos Mononucleares/imunologia , Glicoproteínas de Membrana/imunologia , Valores de Referência , Reprodutibilidade dos Testes
16.
Oncol Rep ; 15(2): 361-7, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16391855

RESUMO

A humanized monoclonal antibody against HM1.24 antigen (AHM), which is highly expressed on multiple myeloma (MM) cells, induced antibody-dependent cellular cytotoxicity (ADCC) in vitro. In this study, we further characterized AHM and evaluated its potency for clinical application. AHM bound to HM1.24 antigen with a dissociation constant of 0.35 nM, and its epitope resided between Leu116 and Leu127 of the HM1.24 antigen. Single intravenous injection of AHM significantly inhibited tumor growth in both orthotopic and ectopic human MM xenograft models. AHM reduced serum M protein levels and prolonged survival of mice intravenously inoculated with KPMM2 and ARH-77 cells. The number of KPMM2 cells in bone marrow or tumor volume of subcutaneously inoculated RPMI 8226 cells was also inhibited by AHM. The antitumor activity of AHM against tumor cells in bone marrow was diminished when the mice were pretreated with anti-Fcgamma receptor III/II antibody, demonstrating that antitumor activity by AHM requires effector cell functions in vivo. Experiments involving in vitro ADCC assays indicated that NK cells and monocytes/macrophages serve as effector cells for AHM-induced ADCC in mouse and human. Thus, AHM will provide an additional treatment option for MM.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Especificidade de Anticorpos/imunologia , Glicoproteínas de Membrana/imunologia , Mieloma Múltiplo/tratamento farmacológico , Animais , Afinidade de Anticorpos/imunologia , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Antígenos CD , Mapeamento de Epitopos , Citometria de Fluxo , Proteínas Ligadas por GPI , Humanos , Células Matadoras Naturais/imunologia , Leucócitos Mononucleares/imunologia , Masculino , Camundongos , Camundongos SCID , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Int Immunopharmacol ; 5(12): 1731-40, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16102523

RESUMO

To characterize the biological activity of tocilizumab, a humanized anti-human interleukin-6 receptor (IL-6R) monoclonal antibody, we examined its binding activity to both soluble IL-6R (sIL-6R) and membrane bound IL-6R (mIL-6R) and its neutralizing activity to other IL-6 family cytokines. ELISA assay demonstrated that tocilizumab bound to sIL-6R and inhibited IL-6 binding to sIL-6R in a dose-dependent manner. The dissociation constant (Kd value) for IL-6R was determined to be 2.54+/-0.12 nmol/L by Scatchard analysis. In addition, tocilizumab had the ability to dissociate IL-6 and sIL-6R from their preformed complex. The immune complex of tocilizumab and sIL-6R did not transmit signaling. Moreover, tocilizumab suppressed the IL-6/sIL-6R complex-induced proliferation of human gp130-transfected cell, BAF-h130. In addition, tocilizumab had the ability to bind to human IL-6R expressing COS-7 cells and to suppress the growth of the IL-6-dependent myeloma cell line, KPMM2. Finally, to analyze the specificity of this antibody, the effects on signal transduction of IL-6 family cytokines such as interleukin-11 (IL-11), oncostatin M (OSM), leukemia inhibitory factor (LIF), and ciliary neurotrophic factor (CNTF) were examined using murine transfectant cell lines (BaF/IL-6R, BaF/IL-11R, BaF/OSMR, BaF/LIFR and BaF/CNTFR) that proliferate depending on IL-6, IL-11, OSM, LIF and human CNTF, respectively. Tocilizumab inhibited the proliferation of BaF/IL-6R induced by IL-6, but did not inhibit the proliferation of BaF/IL-11R, BaF/OSMR, BaF/LIFR and BaF/CNTFR cells induced by their corresponding cytokines. These lines of evidence indicate that tocilizumab is able to bind to both sIL-6R and mIL-6R and to inhibit IL-6 binding to its receptors, leading to the blockade of the IL-6 signaling through both sIL-6R and mIL-6R, but not block the signaling of other IL-6 family cytokines.


Assuntos
Anticorpos Monoclonais/farmacologia , Citocinas/imunologia , Receptores de Interleucina-6/imunologia , Transdução de Sinais/efeitos dos fármacos , Animais , Anticorpos Monoclonais Humanizados , Células COS , Proliferação de Células/efeitos dos fármacos , Chlorocebus aethiops , Citocinas/metabolismo , Relação Dose-Resposta a Droga , Humanos , Interleucina-6/imunologia , Interleucina-6/metabolismo , Ligação Proteica , Receptores de Citocinas/imunologia , Receptores de Citocinas/metabolismo , Receptores de Interleucina-6/metabolismo , Transdução de Sinais/imunologia , Transfecção
18.
Clin Cancer Res ; 11(9): 3377-84, 2005 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15867238

RESUMO

PURPOSE: The purpose of this work was to test the suitability of the HM1.24 antigen as a CTL target for immunotherapy of patients with multiple myeloma. EXPERIMENTAL DESIGN: Antigen-specific T cells were generated from patients with multiple myeloma using stimulation with protein-pulsed dendritic cells and tested in ELISPOT and CTL assays. RESULTS: HM1.24-primed T cells responded selectively to HM1.24-loaded autologous peripheral blood mononuclear cells (PBMC) in an IFN-gamma ELISPOT assay (median, 342; range, 198-495 IFN-gamma-producing cells/10(5) cf. unloaded PBMC median, 98; range, 7-137; P < 0.05, n = 5) and also to autologous malignant plasma cells (MPC; median, 227; range, 153-335; P < 0.05 when compared with the response to allogeneic MPC median, 57; range, 22-158; n = 5). HM1.24-primed T cells lysed autologous MPC (at 20:1 E/T ratio: median, 48% specific killing; range, 23-88%; at 10:1 E/T ratio: median, 43%; range, 15-80%; n = 12) but not allogeneic MPC. Lysis of autologous MPC was inhibited by anti-MHC class I but not anti-MHC class II antibodies and was blocked by Concanamycin A. Lysis of autologous MPC was blocked by competition with autologous HM1.24-transfected dendritic cells (10:1 ratio with autologous MPC). Unmanipulated, or control plasmid-transfected dendritic cells had no effect on lysis of autologous MPC. CONCLUSION: Our results indicate that HM1.24 is a promising target for immunotherapy of multiple myeloma.


Assuntos
Glicoproteínas de Membrana/imunologia , Mieloma Múltiplo/imunologia , Linfócitos T Citotóxicos/imunologia , Antígenos CD , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Testes Imunológicos de Citotoxicidade , Citotoxicidade Imunológica/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Citometria de Fluxo/métodos , Proteínas Ligadas por GPI , Humanos , Imunofenotipagem , Interferon gama/metabolismo , Antígenos Comuns de Leucócito/imunologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiologia , Mieloma Múltiplo/patologia , Perforina , Plasmócitos/imunologia , Proteínas Citotóxicas Formadoras de Poros , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Transfecção
19.
Biochem Biophys Res Commun ; 325(4): 1201-9, 2004 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-15555554

RESUMO

A mouse monoclonal antibody (2D7 mAb), which specifically bound to the alpha2 domain of HLA class I, rapidly induces cell aggregation accompanied by weak cytotoxicity against ARH-77 cells, suggesting that 2D7 mAb had a potential for agonist antibody. In order to enhance this cytotoxicity, 2D7 mAb was engineered to be a small bivalent antibody fragment, 2D7 diabody. The resultant 2D7 diabody showed a strong cytotoxicity against ARH-77 cells. As a notable characteristic feature, the lethal effect of 2D7 diabody was quite rapid, mediated by a caspase-independent death pathway. Furthermore, 2D7 diabody also showed cytotoxicity against several leukemia and lymphoma cell lines, and mitogen-activated peripheral blood mononuclear cells (PBMC), but not for normal resting PBMC and adherent cell lines such as HUVEC. These results suggest that 2D7 diabody could be expected as a novel therapeutic antibody for hematological malignancies as well as inflammatory diseases.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/imunologia , Apoptose/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Mieloma Múltiplo/imunologia , Animais , Apoptose/efeitos dos fármacos , Células CHO , Caspases/imunologia , Linhagem Celular Tumoral , Células Cultivadas , Cricetinae , Cricetulus , Relação Dose-Resposta a Droga , Humanos , Mieloma Múltiplo/patologia , Ligação Proteica , Estrutura Terciária de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...