Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Exp Neurol ; 347: 113918, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34748756

RESUMEN

In temporal lobe epilepsy (TLE), abnormal axon guidance and synapse formation lead to sprouting of mossy fibers in the hippocampus, which is one of the most consistent pathological findings in patients and animal models with TLE. Glypican 4 (Gpc4) belongs to the heparan sulfate proteoglycan family, which play an important role in axon guidance and excitatory synapse formation. However, the role of Gpc4 in the development of mossy fibers sprouting (MFS) and its underlying mechanism remain unknown. Using a pilocarpine-induced mice model of epilepsy, we showed that Gpc4 expression was significantly increased in the stratum granulosum of the dentate gyrus at 1 week after status epilepticus (SE). Using Gpc4 overexpression or Gpc4 shRNA lentivirus to regulate the Gpc4 level in the dentate gyrus, increased or decreased levels of netrin-1, SynI, PSD-95, and Timm score were observed in the dentate gyrus, indicating a crucial role of Gpc4 in modulating the development of functional MFS. The observed effects of Gpc4 on MFS were significantly antagonized when mice were treated with L-leucine or rapamycin, an agonist or antagonist of the mammalian target of rapamycin (mTOR) signal, respectively, demonstrating that mTOR pathway is an essential requirement for Gpc4-regulated MFS. Additionally, the attenuated spontaneous recurrent seizures (SRSs) were observed during chronic stage of the disease by suppressing the Gpc4 expression after SE. Altogether, our findings demonstrate a novel control of neuronal Gpc4 on the development of MFS through the mTOR pathway after pilocarpine-induced SE. Our results also strongly suggest that Gpc4 may serve as a promising target for antiepileptic studies.


Asunto(s)
Glipicanos/biosíntesis , Fibras Musgosas del Hipocampo/metabolismo , Pilocarpina/toxicidad , Transducción de Señal/fisiología , Estado Epiléptico/metabolismo , Serina-Treonina Quinasas TOR/biosíntesis , Animales , Células Cultivadas , Glipicanos/antagonistas & inhibidores , Masculino , Ratones , Fibras Musgosas del Hipocampo/efectos de los fármacos , Agonistas Muscarínicos/toxicidad , Transducción de Señal/efectos de los fármacos , Estado Epiléptico/inducido químicamente , Serina-Treonina Quinasas TOR/antagonistas & inhibidores
2.
Am J Physiol Cell Physiol ; 321(5): C846-C858, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34550795

RESUMEN

Glypican-1 (GPC1) is one of the six glypican family members in humans. It is composed of a core protein with three heparan sulfate chains and attached to the cell membrane by a glycosyl-phosphatidylinositol anchor. GPC1 modulates various signaling pathways including fibroblast growth factors (FGF), vascular endothelial growth factor-A (VEGF-A), transforming growth factor-ß (TGF-ß), Wnt, Hedgehog (Hh), and bone morphogenic protein (BMP) through specific interactions with pathway ligands and receptors. The impact of these interactions on signaling pathways, activating or inhibitory, is dependent upon specific GPC1 domain interaction with pathway components, as well as cell surface context. In this review, we summarize the current understanding of the structure of GPC1, as well as its role in regulating multiple signaling pathways. We focus on the functions of GPC1 in cancer cells and how new insights into these signaling processes can inform its translational potential as a therapeutic target in cancer.


Asunto(s)
Glipicanos/metabolismo , Transducción de Señal , Animales , Antineoplásicos Inmunológicos/uso terapéutico , Glipicanos/antagonistas & inhibidores , Glipicanos/química , Humanos , Inmunoconjugados/uso terapéutico , Inmunoterapia Adoptiva , Ligandos , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/terapia , Conformación Proteica , Relación Estructura-Actividad
3.
Mol Cancer Ther ; 20(9): 1713-1722, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34224365

RESUMEN

Cholangiocarcinoma is a highly malignant cancer. Many patients need systemic chemotherapy to prevent tumor development and recurrence; however, their prognosis is poor due to the lack of effective therapy. Therefore, a new treatment option is urgently required. We recently identified glypican-1 (GPC1) as a novel cancer antigen of esophageal squamous cell carcinoma. We also demonstrated the efficacy and safety of GPC1-targeted ADC (GPC1-ADC) conjugating anti-GPC1 mAb possessing high internalization activity with monomethyl auristatin F (MMAF), which is a potent tubulin polymerizing inhibitor. In this study, we confirmed that GPC1 was highly expressed in cholangiocarcinoma cells and tissues. IHC analysis of 49 extrahepatic cholangiocarcinoma patient tumor specimens revealed high expression of GPC1 in 47% of patients. These patients demonstrated significantly poorer prognosis compared with the low-expression group in terms of disease-free survival and overall survival (P < 0.05). GPC1 was also expressed in tumor vessels of cholangiocarcinoma, but not on the vessels of nontumor tissues. MMAF-conjugated GPC1-ADC showed potent tumor growth inhibition against GPC1-positive cholangiocarcinoma cells in vitro and in vivo In a GPC1 knockout xenograft model, GPC1-ADC partially inhibited tumor growth. Vascular endothelial cells in tumor tissues of GPC1-negative xenograft mice expressed GPC1 and were arrested in the G2-M phase of cell cycle by GPC1-ADC. GPC1-ADC exhibits direct as well as indirect antitumor effects via inhibition of tumor angiogenesis. Our preclinical data highlight GPC1-ADC as a promising therapy for GPC1-positive cholangiocarcinoma.


Asunto(s)
Neoplasias de los Conductos Biliares/tratamiento farmacológico , Colangiocarcinoma/tratamiento farmacológico , Glipicanos/antagonistas & inhibidores , Inmunoconjugados/farmacología , Neovascularización Patológica/prevención & control , Animales , Apoptosis , Neoplasias de los Conductos Biliares/irrigación sanguínea , Neoplasias de los Conductos Biliares/metabolismo , Neoplasias de los Conductos Biliares/patología , Proliferación Celular , Colangiocarcinoma/irrigación sanguínea , Colangiocarcinoma/metabolismo , Colangiocarcinoma/patología , Femenino , Glipicanos/inmunología , Humanos , Ratones , Ratones SCID , Neovascularización Patológica/inmunología , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Pronóstico , Tasa de Supervivencia , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Cell Rep Med ; 2(6): 100297, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34195677

RESUMEN

Targeting solid tumors must overcome several major obstacles, in particular, the identification of elusive tumor-specific antigens. Here, we devise a strategy to help identify tumor-specific epitopes. Glypican 2 (GPC2) is overexpressed in neuroblastoma. Using RNA sequencing (RNA-seq) analysis, we show that exon 3 and exons 7-10 of GPC2 are expressed in cancer but are minimally expressed in normal tissues. Accordingly, we discover a monoclonal antibody (CT3) that binds exons 3 and 10 and visualize the complex structure of CT3 and GPC2 by electron microscopy. The potential of this approach is exemplified by designing CT3-derived chimeric antigen receptor (CAR) T cells that regress neuroblastoma in mice. Genomic sequencing of T cells recovered from mice reveals the CAR integration sites that may contribute to CAR T cell proliferation and persistence. These studies demonstrate how RNA-seq data can be exploited to help identify tumor-associated exons that can be targeted by CAR T cell therapies.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Glipicanos/genética , Neoplasias del Sistema Nervioso/terapia , Neuroblastoma/terapia , Receptores de Antígenos de Linfocitos T/genética , Receptores Quiméricos de Antígenos/genética , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/metabolismo , Línea Celular Tumoral , Proliferación Celular , Exones , Femenino , Expresión Génica , Glipicanos/antagonistas & inhibidores , Glipicanos/química , Glipicanos/inmunología , Humanos , Inmunoterapia Adoptiva/métodos , Ratones , Ratones Desnudos , Modelos Moleculares , Neoplasias del Sistema Nervioso/genética , Neoplasias del Sistema Nervioso/mortalidad , Neoplasias del Sistema Nervioso/patología , Neuroblastoma/genética , Neuroblastoma/mortalidad , Neuroblastoma/patología , Unión Proteica , Conformación Proteica , Receptores de Antígenos de Linfocitos T/inmunología , Receptores Quiméricos de Antígenos/inmunología , Análisis de Secuencia de ARN , Análisis de Supervivencia , Linfocitos T/inmunología , Linfocitos T/metabolismo , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Neoplasia ; 23(9): 939-950, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34332450

RESUMEN

An antibody-drug conjugate (ADC) is a promising therapeutic modality because selective and effective delivery of an anti-cancer drug is achieved by drug-conjugated antibody-targeting cancer antigen. Glypican 1 (GPC1) is highly expressed in malignant tumors, including pancreatic ductal adenocarcinoma (PDAC) and esophageal squamous cell carcinoma (ESCC). Herein, we describe the usefulness of GPC1-targeting ADC. Humanized anti-GPC1 antibody (clone T2) was developed and conjugated with monomethyl auristatin E (MMAE) via maleimidocaproyl-valine-citrulline-p-aminobenzyloxycarbonyl (mc-vc-PABC) linkers (humanized GPC1-ADC[MMAE]). Humanized GPC1-ADC(MMAE) inhibited the growth of GPC1-positive PDAC and ESCC cell lines via inducing cycle arrest in the G2/M phase and apoptosis in vitro. The binding activity of humanized GPC1-ADC(MMAE) with GPC1 was comparable with that of the unconjugated anti-GPC1 antibody. The humanized GPC1-ADC(MMAE) was effective in GPC1-positive BxPC-3 subcutaneously xenografted mice but not in GPC1-negative BxPC-3-GPC1-KO xenografted mice. To assess the bystander killing activity of the humanized GPC1-ADC(MMAE), a mixture of GPC1-positive BxPC-3 and GPC1-negative BxPC-3-GPC1-KO-Luc cells were subcutaneously inoculated, and a heterogenous GPC1-expressing tumor model was developed. The humanized GPC1-ADC(MMAE) inhibited the tumor growth and decreased the luciferase signal, measured with an in vivo imaging system (IVIS), which suggests that the suppression of the BxPC-3-GPC1-KO-Luc population. The humanized GPC1-ADC(MMAE) also inhibited the established liver metastases of BxPC-3 cells and significantly improved the overall survival of the mice. It exhibited a potent antitumor effect on the GPC1-positive PDAC and ESCC patient-derived xenograft (PDX) models. Our preclinical data demonstrate that GPC1 is a promising therapeutic target for ADC.


Asunto(s)
Anticuerpos Monoclonales Humanizados/metabolismo , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas de Esófago/metabolismo , Glipicanos/metabolismo , Inmunoconjugados/metabolismo , Neoplasias Pancreáticas/metabolismo , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales Humanizados/administración & dosificación , Antígenos de Neoplasias/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/inmunología , Carcinoma de Células Escamosas de Esófago/tratamiento farmacológico , Carcinoma de Células Escamosas de Esófago/inmunología , Glipicanos/antagonistas & inhibidores , Inhibidores de Crecimiento/administración & dosificación , Inhibidores de Crecimiento/metabolismo , Humanos , Inmunoconjugados/administración & dosificación , Ratones , Ratones Noqueados , Ratones SCID , Ratones Transgénicos , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
6.
J Immunother Cancer ; 9(4)2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33833049

RESUMEN

BACKGROUND: Glypican-3 (GPC3), a cell surface glycoprotein that is pathologically highly expressed in hepatocellular carcinoma (HCC), is an attractive target for immunotherapies, including chimeric antigen receptor (CAR) T cells. The serum GPC3 is frequently elevated in HCC patients due to the shedding effect of cell surface GPC3. The shed GPC3 (sGPC3) is reported to block the function of cell-surface GPC3 as a negative regulator. Therefore, it would be worth investigating the potential influence of antigen shedding in anti-GPC3 CAR-T therapy for HCC. METHODS: In this study, we constructed two types of CAR-T cells targeting distinct epitopes of GPC3 to examine how sGPC3 influences the activation and cytotoxicity of CAR-T cells in vitro and in vivo by introducing sGPC3 positive patient serum or recombinant sGPC3 proteins into HCC cells or by using sGPC3-overexpressing HCC cell lines. RESULTS: Both humanized YP7 CAR-T cells and 32A9 CAR-T cells showed GPC3-specific antitumor functions in vitro and in vivo. The existence of sGPC3 significantly inhibited the release of cytokines and the cytotoxicity of anti-GPC3 CAR-T cells in vitro. In animal models, mice carrying Hep3B xenograft tumors expressing sGPC3 exhibited a worse response to the treatment with CAR-T cells under both a low and high tumor burden. sGPC3 bound to CAR-T cells but failed to induce the effective activation of CAR-T cells. Therefore, sGPC3 acted as dominant negative regulators when competed with cell surface GPC3 to bind anti-GPC3 CAR-T cells, leading to an inhibitory effect on CAR-T cells in HCC. CONCLUSIONS: We provide a proof-of-concept study demonstrating that GPC3 shedding might cause worse response to CAR-T cell treatment by competing with cell surface GPC3 for CAR-T cell binding, which revealed a new mechanism of tumor immune escape in HCC, providing a novel biomarker for patient enrolment in future clinical trials and/or treatments with GPC3-targeted CAR-T cells.


Asunto(s)
Biomarcadores de Tumor/antagonistas & inhibidores , Carcinoma Hepatocelular/terapia , Glipicanos/antagonistas & inhibidores , Inmunoterapia Adoptiva , Neoplasias Hepáticas/terapia , Receptores Quiméricos de Antígenos/genética , Linfocitos T/trasplante , Animales , Unión Competitiva , Biomarcadores de Tumor/sangre , Biomarcadores de Tumor/inmunología , Carcinoma Hepatocelular/sangre , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Citocinas/metabolismo , Citotoxicidad Inmunológica , Femenino , Glipicanos/sangre , Glipicanos/inmunología , Neoplasias Hepáticas/sangre , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/patología , Activación de Linfocitos , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones Desnudos , Prueba de Estudio Conceptual , Unión Proteica , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Sci Rep ; 11(1): 9262, 2021 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-33927256

RESUMEN

Cancer-associated fibroblasts (CAFs) are the most abundant stromal cell type in the tumor microenvironment. CAFs orchestrate tumor-stromal interactions, and contribute to cancer cell growth, metastasis, extracellular matrix (ECM) remodeling, angiogenesis, immunomodulation, and chemoresistance. However, CAFs have not been successfully targeted for the treatment of cancer. The current study elucidates the significance of glypican-1 (GPC-1), a heparan sulfate proteoglycan, in regulating the activation of human bone marrow-derived stromal cells (BSCs) of fibroblast lineage (HS-5). GPC-1 inhibition changed HS-5 cellular and nuclear morphology, and increased cell migration and contractility. GPC-1 inhibition also increased pro-inflammatory signaling and CAF marker expression. GPC-1 induced an activated fibroblast phenotype when HS-5 cells were exposed to prostate cancer cell conditioned media (CCM). Further, treatment of human bone-derived prostate cancer cells (PC-3) with CCM from HS-5 cells exhibiting GPC-1 loss increased prostate cancer cell aggressiveness. Finally, GPC-1 was expressed in mouse tibia bone cells and present during bone loss induced by mouse prostate cancer cells in a murine prostate cancer bone model. These data demonstrate that GPC-1 partially regulates the intrinsic and extrinsic phenotype of human BSCs and transformation into activated fibroblasts, identify novel functions of GPC-1, and suggest that GPC-1 expression in BSCs exerts inhibitory paracrine effects on the prostate cancer cells. This supports the hypothesis that GPC-1 may be a novel pharmacological target for developing anti-CAF therapeutics to control cancer.


Asunto(s)
Fibroblastos Asociados al Cáncer/patología , Fibroblastos/patología , Glipicanos/antagonistas & inhibidores , Células Madre Mesenquimatosas/patología , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/secundario , Microambiente Tumoral/inmunología , Animales , Fibroblastos Asociados al Cáncer/inmunología , Fibroblastos Asociados al Cáncer/metabolismo , Movimiento Celular , Fibroblastos/metabolismo , Glipicanos/metabolismo , Humanos , Masculino , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Fenotipo , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/metabolismo
8.
Artículo en Inglés | MEDLINE | ID: mdl-33418242

RESUMEN

Screening active components targeting membrane proteins is important for drug discovery from traditional Chinese medicine. Cell membrane chromatography (CMC) has achieved a wide application in screening active components on pathological cells due to its high sensitivity and effectiveness. However, it is hard to clarify the specific target protein through simply using pathological and normal cells. In this study, a novel comparative two-dimensional (2D) cell membrane chromatography system was established. Based on the construction of hepatocellular carcinoma cell line SK-Hep1-GPC3 with high expression of protein Glypican-3 (GPC3), SK-Hep1-GPC3/CMC column was loaded to screen selective antitumor components from Scutellariae Radix according to the retention behaviors on column. Viscidulin I was retained on SK-Hep1-GPC3/CMC column, and showed 4.33 µM affinity to GPC3 according to surface plasmon resonance (SPR). The IC50 of viscidulin I on SK-Hep1-GPC3 cells was 18.01 µM in cell proliferation assay. Thus, this method can be applied to screen complex herbal medicines for ligands bound to specific target protein receptor related to hepatic carcinoma.


Asunto(s)
Antineoplásicos , Membrana Celular/metabolismo , Cromatografía de Afinidad/métodos , Glipicanos , Scutellaria baicalensis/química , Antineoplásicos/análisis , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Línea Celular Tumoral , Descubrimiento de Drogas , Medicamentos Herbarios Chinos/análisis , Medicamentos Herbarios Chinos/metabolismo , Medicamentos Herbarios Chinos/farmacología , Flavonoides/análisis , Flavonoides/metabolismo , Flavonoides/farmacología , Glipicanos/antagonistas & inhibidores , Glipicanos/metabolismo , Humanos , Neoplasias Hepáticas/metabolismo , Reproducibilidad de los Resultados
9.
Mol Ther ; 29(4): 1572-1584, 2021 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-33429083

RESUMEN

Glypican-3 (GPC3) is a well-characterized hepatocellular carcinoma (HCC)-associated antigen, yet anti-GPC3 therapies have achieved only minimal clinical progress. CD47 is a ubiquitously expressed innate immune checkpoint that promotes evasion of tumors from immune surveillance. Given both the specific expression of GPC3 in HCC and the known phagocytosis inhibitory effect of CD47 in liver cancer, we hypothesized that a bispecific antibody (BsAb) that co-engages with GPC3 and CD47 may offer excellent antitumor efficacy with minimal toxicity. Here, we generated a novel BsAb: GPC3/CD47 biAb. With the use of both in vitro and in vivo assays, we found that GPC3/CD47 biAb exerts strong antitumor activity preferentially against dual antigen-expressing tumor cells. In hCD47/human signal regulatory protein alpha (hCD47/hSIRPα) humanized mice, GPC3/CD47 biAb had an extended serum half-life without causing systemic toxicity. Importantly, GPC3/CD47 biAb induced enhanced Fc-mediated effector functions to dual antigen-expressing HCC cells in vitro, and both macrophages and neutrophils are required for its strong efficacy against xenograft HCC tumors. Notably, GPC3/CD47 biAb outperformed monotherapies and a combination therapy with anti-CD47 and anti-GPC3 monoclonal antibodies (mAbs) in a xenograft HCC model. Our study illustrates a strategy for improving HCC treatment by boosting innate immune responses and presents new insights to inform antibody design for the future development of innovative immune therapies.


Asunto(s)
Antígeno CD47/genética , Carcinoma Hepatocelular/tratamiento farmacológico , Glipicanos/genética , Neoplasias Hepáticas/tratamiento farmacológico , Animales , Anticuerpos Biespecíficos/farmacología , Anticuerpos Monoclonales/farmacología , Antígeno CD47/antagonistas & inhibidores , Antígeno CD47/inmunología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Glipicanos/antagonistas & inhibidores , Glipicanos/inmunología , Humanos , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/inmunología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/patología , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Invest New Drugs ; 39(3): 615-626, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33215325

RESUMEN

The acceleration of therapeutic antibody development has been motivated by the benefit to and their demand for human health. In particular, humanized transgenic antibody discovery platforms, combined with immunization, hybridoma fusion and/or single cell DNA sequencing are the most reliable and rapid methods for mining the human monoclonal antibodies. Human GPC3 protein is an oncofetal antigen, and it is highly expressed in most hepatocellular carcinomas and some types of squamous cell carcinomas. Currently, no fully human anti-GPC3 therapeutic antibodies have been reported and evaluated in extensive tumor tissues. Here, we utilized a new humanized transgenic mouse antibody discovery platform (CAMouse) that contains large V(D)J -regions and human gamma-constant regions of human immunoglobulin in authentic configurations to generate fully human anti-GPC3 antibodies. Our experiments resulted in four anti-GPC3 antibodies with high-specific binding and cytotoxicity to GPC3 positive cancer cells, and the antibody affinities are in the nanomolar range. Immunohistochemistry analysis demonstrated that these antibodies can recognize GPC3 protein on many types of solid tumors. In summary, the human anti-human GPC3 monoclonal antibodies described here are leading candidates for further preclinical studies of cancer therapy, further, the CAMouse platform is a robust tool for human therapeutic antibody discovery.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Glipicanos/antagonistas & inhibidores , Neoplasias/metabolismo , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Femenino , Glipicanos/inmunología , Glipicanos/metabolismo , Hepatitis/metabolismo , Humanos , Intestino Delgado/metabolismo , Pulmón/metabolismo , Masculino , Ratones Transgénicos , Placenta/metabolismo , Embarazo
11.
J Cancer Res Clin Oncol ; 147(2): 499-505, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33225417

RESUMEN

PURPOSE: There is not much progress in the treatment for lung squamous cell carcinoma LSCC in the past few years. Rapamycin Rapa, an inhibitor of mammalian target of rapamycin mTOR, has exhibited antitumor efficacy in a variety of malignant tumors. It has recently been reported that Rapamycin can induce autophagy signaling pathway in lung cancer and Glypican-3GPC3 can promote the growth of hepatocellular carcinoma by stimulating canonical Wnt signaling pathway. The aim of this study is to investigate the mechanisms of rapamycin's antitumor efficacy in relation to GPC3/Wnt/ß-catenin pathway and autophagy in LSCC. METHODS: SK-MES-1 cells, a LSCC cell line, were treated with various concentrations of rapamycin with or without Glypican-3 GPC3-targeting siRNA. SK-MES-1 cell proliferation was determined by MTT assay. Protein expression levels of GPC3, ß-catenin, Beclin-1 were checked via western blotting. We established the xenograft mice model to investigate the suppression effect of rapamycin on LSCC. In addition, we further testified the metabolism protein of autophagy process using the xenograft tumor tissue. RESULTS: Rapamycin could inhibit the SK-MES-1 cell proliferation in a concentration-dependent manner both in vitro and in vivo by decreasing the GPC3 expression and downregulating the glypican-3/Wnt/ß-catenin signaling pathway. In addition, we found that GPC3 silencing can activate the glypican-3/Wnt/ß-catenin pathway and autophagy, which contribute to the suppression of tumor growth both in vitro and in vivo. CONCLUSION: Rapamycin suppresses the growth of lung cancer through down-regulating glypican-3/Wnt/ß-catenin signaling, which mediates with activation of autophagy. This study suggests GPC3 is a new promising target for rapamycin in the treatment of lung cancer.


Asunto(s)
Autofagia/efectos de los fármacos , Carcinoma de Células Escamosas/tratamiento farmacológico , Glipicanos/antagonistas & inhibidores , Neoplasias Pulmonares/tratamiento farmacológico , Sirolimus/farmacología , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo , Femenino , Glipicanos/fisiología , Humanos , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos BALB C , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/fisiología , Vía de Señalización Wnt/fisiología , beta Catenina/fisiología
12.
Arch Dermatol Res ; 313(4): 263-273, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-32638071

RESUMEN

Glypican-3 (GPC3) is considered as a cell surface heparan sulfate proteoglycan. It is overexpressed in skin cancer and promotes tumor progression and pathogenicity. Therefore, we aimed to find out the therapeutic effects of immuno-suppressing GPC3 in skin cancer experimentally induced in mice as well as to underline molecular mechanisms especially inflammatory and apoptotic pathways. Skin cancer was experimentally induced in mice by repeated rubbing of mice skin with 7,12-dimethylbenz (a) anthracene. Mice were injected with anti-GPC3. Skin samples were isolated to investigate the gene and protein expression of GPC3, Wnt-1, NFκB, TNF-α, IGF-1, p38 MAPK and caspase-3 using PCR, Western blot and ELISA. Moreover, skin sections were stained with hematoxylin and eosin. Treating skin cancer mice with anti-GPC3 significantly blocked GPC3, which is accompanied by amelioration of skin cancer-induced increase in the numbers of tumors and scratching behavior. Moreover, anti-GPC3 attenuated skin cancer-induced increase in the expression of Wnt-1, NFκB, TNF-α, IGF-1, p38 MAPK and caspase-3. In parallel, anti-GPC3 reduced degeneration of melanocyte cells and reduced phagocytic cells epidermal hyperplasia and dysplasia in skin sections stained with hematoxylin and eosin stain. In conclusion, anti-GPC3 produced anti-tumor effects against skin cancer, which can be explained by reduction in both inflammatory and apoptotic pathways. Targeting GPC3 is a promising therapeutic approach for skin cancer.


Asunto(s)
Antineoplásicos/farmacología , Carcinogénesis/efectos de los fármacos , Glipicanos/antagonistas & inhibidores , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , 9,10-Dimetil-1,2-benzantraceno/administración & dosificación , 9,10-Dimetil-1,2-benzantraceno/toxicidad , Animales , Antineoplásicos/uso terapéutico , Carcinogénesis/inducido químicamente , Carcinógenos/administración & dosificación , Carcinógenos/toxicidad , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Ratones , Neoplasias Experimentales/etiología , Piel/efectos de los fármacos , Piel/patología , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/patología
13.
BMC Cancer ; 20(1): 1214, 2020 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-33302918

RESUMEN

BACKGROUND: Glypican-1 is a heparan sulfate proteoglycan that is overexpressed in prostate cancer (PCa), and a variety of solid tumors. Importantly, expression is restricted in normal tissue, making it an ideal tumor targeting antigen. Since there is clinical and preclinical evidence of the efficacy of Bispecific T cell Engager (BiTE) therapy in PCa, we sought to produce and test the efficacy of a GPC-1 targeted BiTE construct based on the Miltuximab® sequence. Miltuximab® is a clinical stage anti-GPC-1 antibody that has proven safe in first in human trials. METHODS: The single chain variable fragment (scFv) of Miltuximab® and the CD3 binding sequence of Blinatumomab were combined in a standard BiTE format. Binding of the construct to immobilised recombinant CD3 and GPC-1 antigens was assessed by ELISA and BiaCore, and binding to cell surface-expressed antigens was measured by flow cytometry. The ability of MIL-38-CD3 to activate T cells was assessed using in vitro co-culture assays with tumour cell lines of varying GPC-1 expression by measurement of CD69 and CD25 expression, before cytolytic activity was assessed in a similar co-culture. The release of inflammatory cytokines from T cells was measured by ELISA and expression of PD-1 on the T cell surface was measured by flow cytometry. RESULTS: Binding activity of MIL-38-CD3 to both cell surface-expressed and immobilised recombinant GPC-1 and CD3 was retained. MIL-38-CD3 was able to mediate the activation of peripheral blood T cells from healthy individuals, resulting in the release of inflammatory cytokines TNF and IFN-g. Activation was reliant on GPC-1 expression as MIL-38-CD3 mediated only low level T cell activation in the presence of C3 cells (constitutively low GPC-1 expression). Activated T cells were redirected to lyse PCa cell lines PC3 and DU-145 (GPC-1 moderate or high expression, respectively) but could not kill GPC-1 negative Raji cells. The expression of PD-1 was up-regulated on the surface of MIL-38-CD3 activated T cells, suggesting potential for synergy with checkpoint inhibition. CONCLUSIONS: This study reports preclinical findings into the efficacy of targeting GPC-1 in PCa with BiTE construct MIL-38-CD3. We show the specificity and efficacy of the construct, supporting its further preclinical development.


Asunto(s)
Adenocarcinoma/patología , Anticuerpos Biespecíficos/farmacología , Glipicanos/inmunología , Proteínas de Neoplasias/inmunología , Neoplasias de la Próstata/patología , Anticuerpos de Cadena Única/farmacología , Especificidad del Receptor de Antígeno de Linfocitos T , Linfocitos T Citotóxicos/inmunología , Adenocarcinoma/inmunología , Anticuerpos Biespecíficos/inmunología , Antígenos CD/análisis , Antígenos de Diferenciación de Linfocitos T/análisis , Complejo CD3/inmunología , Línea Celular Tumoral , Técnicas de Cocultivo , Citocinas/metabolismo , Citotoxicidad Inmunológica , Glipicanos/antagonistas & inhibidores , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Subunidad alfa del Receptor de Interleucina-2/análisis , Lectinas Tipo C/análisis , Activación de Linfocitos , Masculino , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias de la Próstata/inmunología , Proteínas Recombinantes/inmunología , Anticuerpos de Cadena Única/inmunología , Linfocitos T Citotóxicos/metabolismo
14.
Cancer Control ; 27(3): 1073274820945975, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32799550

RESUMEN

Hepatocellular carcinoma (HCC), one of the most common lethal diseases in the world, has a 5-year survival rate of only 7%. Hepatocellular carcinoma has no symptoms in the early stage but obvious symptoms in the late stage, leading to delayed diagnosis and reduced treatment efficacy. In recent years, as the scope of HCC research has increased in depth, the clinical development and application of molecular targeted drugs and immunotherapy drugs have brought new breakthroughs in HCC treatment. Targeted therapy drugs for HCC have high specificity, allowing them to selectively kill tumor cells and minimize damage to normal tissues. At present, these targeted drugs are mainly classified into 3 categories: small molecule targeted drugs, HCC antigen-specific targeted drugs, and immune checkpoint targeted drugs. This article reviews the latest research progress on the targeted drugs for HCC.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Terapia Molecular Dirigida/métodos , Glipicanos/antagonistas & inhibidores , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inhibidores de la Metaloproteinasa de la Matriz/uso terapéutico , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , alfa-Fetoproteínas/antagonistas & inhibidores
15.
Biomolecules ; 10(6)2020 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-32575752

RESUMEN

Hepatocellular carcinoma (HCC) accounts for most liver cancers and represents one of the deadliest cancers in the world. Despite the global demand for liver cancer treatments, there remain few options available. The U.S. Food and Drug Administration (FDA) recently approved Lumoxiti, a CD22-targeting immunotoxin, as a treatment for patients with hairy cell leukemia. This approval helps to demonstrate the potential role that immunotoxins can play in the cancer therapeutics pipeline. However, concerns have been raised about the use of immunotoxins, including their high immunogenicity and short half-life, in particular for treating solid tumors such as liver cancer. This review provides an overview of recent efforts to develop a glypican-3 (GPC3) targeting immunotoxin for treating HCC, including strategies to deimmunize immunotoxins by removing B- or T-cell epitopes on the bacterial toxin and to improve the serum half-life of immunotoxins by incorporating an albumin binding domain.


Asunto(s)
Toxinas Bacterianas/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Glipicanos/antagonistas & inhibidores , Inmunotoxinas/farmacología , Neoplasias Hepáticas/tratamiento farmacológico , Animales , Toxinas Bacterianas/inmunología , Humanos , Inmunotoxinas/inmunología
16.
Hepatology ; 71(5): 1696-1711, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31520528

RESUMEN

BACKGROUND AND AIMS: Treatment of hepatocellular carcinomas using our glypican-3 (GPC3)-targeting human nanobody (HN3) immunotoxins causes potent tumor regression by blocking protein synthesis and down-regulating the Wnt signaling pathway. However, immunogenicity and a short serum half-life may limit the ability of immunotoxins to transition to the clinic. APPROACH AND RESULTS: To address these concerns, we engineered HN3-based immunotoxins to contain various deimmunized Pseudomonas exotoxin (PE) domains. This included HN3-T20, which was modified to remove T-cell epitopes and contains a PE domain II truncation. We compared them to our previously reported B-cell deimmunized immunotoxin (HN3-mPE24) and our original HN3-immunotoxin with a wild-type PE domain (HN3-PE38). All of our immunotoxins displayed high affinity to human GPC3, with HN3-T20 having a KD value of 7.4 nM. HN3-T20 retained 73% enzymatic activity when compared with the wild-type immunotoxin in an adenosine diphosphate-ribosylation assay. Interestingly, a real-time cell growth inhibition assay demonstrated that a single dose of HN3-T20 at 62.5 ng/mL (1.6 nM) was capable of inhibiting nearly all cell proliferation during the 10-day experiment. To enhance HN3-T20's serum retention, we tested the effect of adding a streptococcal albumin-binding domain (ABD) and a llama single-domain antibody fragment specific for mouse and human serum albumin. For the detection of immunotoxin in mouse serum, we developed a highly sensitive enzyme-linked immunosorbent assay and found that HN3-ABD-T20 had a 45-fold higher serum half-life than HN3-T20 (326 minutes vs. 7.3 minutes); consequently, addition of an ABD resulted in HN3-ABD-T20-mediated tumor regression at 1 mg/kg. CONCLUSION: These data indicate that ABD-containing deimmunized HN3-T20 immunotoxins are high-potency therapeutics ready to be evaluated in clinical trials for the treatment of liver cancer.


Asunto(s)
ADP Ribosa Transferasas/uso terapéutico , Toxinas Bacterianas/uso terapéutico , Carcinoma Hepatocelular/terapia , Exotoxinas/uso terapéutico , Glipicanos/antagonistas & inhibidores , Inmunotoxinas/uso terapéutico , Neoplasias Hepáticas/terapia , Anticuerpos de Dominio Único/uso terapéutico , Factores de Virulencia/uso terapéutico , ADP Ribosa Transferasas/química , ADP Ribosa Transferasas/farmacología , Animales , Toxinas Bacterianas/química , Toxinas Bacterianas/farmacología , Línea Celular Tumoral , Exotoxinas/química , Exotoxinas/farmacología , Humanos , Inmunotoxinas/química , Inmunotoxinas/farmacología , Ratones , Ratones Desnudos , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/farmacología , Factores de Virulencia/química , Factores de Virulencia/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Exotoxina A de Pseudomonas aeruginosa
17.
Biomater Sci ; 7(12): 5258-5269, 2019 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-31603456

RESUMEN

Low binding affinity and lack of therapy functions limit tumor targeting peptide applications in the biomedical field. Herein, we successfully modified a previous phage display derived Glypican-3 (GPC3) binding peptide (GBP) on the surface of a Fe3O4 Core/Au shell nanocomplex (FANP) to improve GBP binding affinity and enhance FANP tumor photothermal therapy (PTT) efficacy. As a result, GBP-FANP showed improved avidity to GPC-3 (Apparent Kd = 396.3 ± 70.8 nM) compared to that of GPB (Apparent Kd = 735.2 ± 53.6 nM). After intravenous administration, GBP-FANP was found specifically accumulated in GPC-3 positive HepG2 tumors and peaked at 24 h post-injection as observed by magnetic resonance imaging (MRI)/photoacoustic (PA)/fluorescent imaging. Moreover, HepG2 tumors that received GBP-FANP treatment were significantly inhibited with laser irradiation (630 nm, 1 W cm-2, 10 min). In conclusion, our present strategy provides a way of improving peptide ligand avidity with nanotechnology for cancer theranostics applications.


Asunto(s)
Compuestos Ferrosos/química , Glipicanos/metabolismo , Neoplasias Hepáticas/terapia , Péptidos/administración & dosificación , Fototerapia/métodos , Administración Intravenosa , Animales , Línea Celular Tumoral , Glipicanos/antagonistas & inhibidores , Oro/química , Células Hep G2 , Humanos , Hipertermia Inducida/métodos , Neoplasias Hepáticas/diagnóstico por imagen , Neoplasias Hepáticas/metabolismo , Imagen por Resonancia Magnética , Nanopartículas del Metal/química , Ratones , Péptidos/química , Péptidos/farmacología , Nanomedicina Teranóstica , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Colloids Surf B Biointerfaces ; 184: 110498, 2019 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-31536939

RESUMEN

Hepatocellular carcinoma (HCC) is the fifth most common cancer in the world with increasing incidence. Chemotherapy is required for HCC patients after receiving surgical resection. Serious off-target induced side effects and systemic toxicity limit the clinical utility of drugs. Targeting therapeutic nanomedicine is an innovative strategy for enhancing drug delivery efficiency and reducing side effects. Here, we successfully formulated nanocarriers to encapsulate sorafenib, an FDA approved drug for treatment of HCC. Sorafenib is encapsulated with an entrapment efficiency >80% over 20 days. The effective aqueous solubility is improved over 1900-fold. The release ratio in vitro is characterized by a half-life of T1/2 = 22.7 h. The peak target-to-background ratio for nanocarrier uptake by tumor occurs at 24 h post-injection, and is significantly greater for the target peptide versus controls. Ex vivo biodistribution confirms the in vivo results. Tumor regression is significantly greater for the target peptide versus controls after 21 days of therapy. No acute toxicity is found by blood chemistry or necropsy. In summary, a peptide specific for GPC3 has been identified, and used to modify the surface of a nanocarrier that encapsulates sorafenib with high entrapment efficiency. Regression of HCC xenograft tumors showed promise for targeted drug delivery.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Portadores de Fármacos/química , Glipicanos/química , Neoplasias Hepáticas/tratamiento farmacológico , Nanopartículas/química , Péptidos/uso terapéutico , Sorafenib/uso terapéutico , Animales , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Composición de Medicamentos/métodos , Femenino , Glipicanos/antagonistas & inhibidores , Glipicanos/metabolismo , Humanos , Neoplasias Hepáticas/patología , Ratones Desnudos , Terapia Molecular Dirigida/métodos , Péptidos/química , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/uso terapéutico , Sorafenib/química , Sorafenib/farmacocinética , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
19.
Sci Rep ; 9(1): 11478, 2019 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-31391540

RESUMEN

Recent studies suggest that glypican-1 (GPC-1) is a biomarker for prostate cancer, but there are few studies elucidating the role of GPC-1 in prostate cancer progression. We observed high expression of GPC-1 in more aggressive prostate cancer cell lines such as PC-3 and DU-145. While inhibition of GPC-1 expression in PC-3 cells decreased cell growth and migration in vitro, it surprisingly increased cell proliferation and migration in DU-145 cells, suggesting that the role of GPC-1 is cell type-dependent. Further, GPC-1 inhibition increased PC-3 tumor size in NCr nude mice xenografts. We hypothesized that the discrepancy between the in vitro and in vivo data is mediated by stromal cells in the tumor microenvironment. Thus, we tested the effect of tumor conditioned media (TCM) on gene expression in human mesenchymal stem cells and fibroblasts. Treatment of stromal cells with TCM from PC-3 cells transfected with GPC-1 shRNA increased the expression of migration markers, endocrine/paracrine biomolecules, and extracellular matrix components. Additionally, the decreased cell growth in GPC-1 knockdown PC-3 cells was rescued by coculturing with stromal cells. These data demonstrate the paradoxical role that GPC-1 plays in prostate cancer cell growth by interacting with stromal cells and through ECM remodeling and endocrine/paracrine signaling.


Asunto(s)
Antineoplásicos/farmacología , Matriz Extracelular/patología , Glipicanos/metabolismo , Neoplasias de la Próstata/patología , Células del Estroma/patología , Animales , Antineoplásicos/uso terapéutico , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Técnicas de Cocultivo , Medios de Cultivo Condicionados , Matriz Extracelular/efectos de los fármacos , Fibroblastos , Técnicas de Silenciamiento del Gen , Glipicanos/antagonistas & inhibidores , Glipicanos/genética , Humanos , Masculino , Células Madre Mesenquimatosas , Ratones , Comunicación Paracrina/efectos de los fármacos , Próstata/citología , Próstata/patología , Neoplasias de la Próstata/tratamiento farmacológico , Células del Estroma/efectos de los fármacos , Carga Tumoral/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
20.
ACS Appl Mater Interfaces ; 11(26): 23591-23604, 2019 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-31179679

RESUMEN

The specific-targeting approach could promote the specificity of diagnosis and the accuracy of cancer treatment. The choice of a specific-targeting receptor is the key step in this approach. Glypican-3 (GPC3) is an oncofetal proteoglycan anchored on the cell membrane. It is overexpressed even in the early stage of hepatocellular carcinoma (HCC), whereas it shows almost no expression in the healthy adult liver. Therefore, GPC3 may be applied as a specific-targeting receptor for HCC theranostics. In this study, a GPC3 specific-targeting theranostics nanodevice, GPC3 targeting peptide (named G12)-modified liposomes co-loaded with sorafenib (SF) and IR780 iodide (IR780), was developed (GSI-Lip), which aims to realize early diagnosis and precise chemo-photothermal therapy of HCC. SF was the first-line chemotherapy drug for the treatment of HCC. IR780 was used for photothermal therapy and near-infrared fluorescence imaging. The evaluation of early diagnosis verified that early-stage tumors (3.45 ± 0.98 mm3, 2 days after 5 × 105 H22 cells' inoculation in mice) could be clearly detected using GSI-Lip, which was significantly more sensitive than folic acid-modified liposomes ( p < 0.01, 32.90 ± 10.01 mm3, 4 days after 1 × 106 H22 cells' inoculation in mice). The study of the endocytic pathway indicated that specific G12/GPC3 recognition may induce caveolae-mediated endocytosis of GSI-Lip. Notably, the accumulation of GSI-Lip in tumors was significantly increased compared with that observed with folic acid-modified liposomes ( p < 0.01). Specific-targeting endowed the precise antitumor effect of GSI-Lip. GSI-Lip showed a higher antitumor efficacy in comparison with folic acid-modified liposomes (inhibition rate: 90.52% vs 84.22%, respectively; p < 0.01). During a period of 21 days, the synergistic chemo-photothermal therapy (GSI-Lip + laser) exhibited a better antitumor effect versus GSI-Lip without laser (inhibition rate: 94.93% vs 90.52%, respectively; p < 0.01). Overall, GPC3-targeted GSI-Lip promoted the sensitivity and specificity of HCC early diagnosis and achieved synergistic efficacy of chemo-photothermal theranostics, which has potential clinical applications. Furthermore, the present study revealed that a more specific-targeting ligand could further improve the efficacy of theranostics against HCC.


Asunto(s)
Carcinoma Hepatocelular/diagnóstico , Glipicanos/genética , Neoplasias Hepáticas/diagnóstico , Fototerapia , Animales , Carcinoma Hepatocelular/terapia , Detección Precoz del Cáncer , Glipicanos/antagonistas & inhibidores , Humanos , Liposomas/química , Liposomas/farmacología , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas/terapia , Ratones , Nanomedicina Teranóstica/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...