Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Commun Signal ; 22(1): 81, 2024 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-38291404

RESUMO

BACKGROUND: Previous research has revealed that the 18 glycoside hydrolase gene family (GH18) member Chitinase 3-like 1 (Chi3l1) can regulate osteoclast differentiation and bone resorption. However, its downstream receptors and molecular mechanisms during osteoclastogenesis have yet to be elucidated. METHODS: Initially, we conducted a comprehensive investigation to evaluate the effects of recombinant Chi3l1 protein or Chi3l1 siRNA on osteoclast differentiation and the RANKL-induced MAPK/AKT signaling pathways. Moreover, we used immunofluorescence and immunoprecipitation assays to identify IL13Rα2 as the downstream receptor of Chi3l1. Subsequently, we investigated the impact of IL13Rα2 recombinant protein or IL13Rα2-siRNA on osteoclast differentiation and the associated signaling pathways. Finally, we performed in vivo experiments to examine the effect of recombinant IL13Rα2 protein in an LPS-induced mouse model of cranial osteolysis. RESULTS: Our findings highlight that the administration of recombinant Chi3l1 protein increased the formation of osteoclasts and bolstered the expression of several osteoclast-specific genes (TRAP, NFATC1, CTR, CTSK, V-ATPase d2, and Dc-STAMP). Additionally, Chi3l1 significantly promoted the RANKL-induced MAPK (ERK/P38/JNK) and AKT pathway activation, whereas Chi3l1 silencing inhibited this process. Next, using immunofluorescence and co-immunoprecipitation assays, we identified IL13Rα2 as the binding partner of Chi3l1 during osteoclastogenesis. IL13Rα2 recombinant protein or IL13Rα2-siRNA also inhibited osteoclast differentiation, and IL13Rα2-siRNA attenuated the RANKL-induced activation of the MAPK (ERK/P38/JNK) and AKT pathways, similar to the effects observed upon silencing of Chi3l1. Moreover, the promoting effect of recombinant Chi3l1 protein on osteoclastogenesis and the activation of the MAPK and AKT pathways was reversed by IL13Rα2 siRNA. Finally, recombinant LI13Rα2 protein significantly attenuated the LPS-induced cranial osteolysis and the number of osteoclasts in vivo. CONCLUSIONS: Our findings suggested that IL13Rα2 served as a crucial receptor for Chi3l1, enhancing RANKL-induced MAPK and AKT activation to promote osteoclast differentiation. These findings provide valuable insights into the molecular mechanisms of Chi3l1 in osteoclastogenesis, with potential therapeutic implications for osteoclast-related diseases. Video Abstract.


Assuntos
Reabsorção Óssea , Subunidade alfa2 de Receptor de Interleucina-13 , Osteólise , Animais , Camundongos , Reabsorção Óssea/tratamento farmacológico , Diferenciação Celular , Proteína 1 Semelhante à Quitinase-3/metabolismo , Subunidade alfa2 de Receptor de Interleucina-13/metabolismo , Subunidade alfa2 de Receptor de Interleucina-13/uso terapêutico , Lipopolissacarídeos/farmacologia , Fatores de Transcrição NFATC/metabolismo , Osteoclastos , Osteólise/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ligante RANK/metabolismo , Proteínas Recombinantes/farmacologia , RNA Interferente Pequeno/metabolismo
2.
Toxicol In Vitro ; 92: 105651, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37482138

RESUMO

Targeting cell surface receptors with immunotoxins provides a novel, unique and highly potent treatment against cancers. A high expression of interleukin-13 (IL13) receptor α2 (IL13Rα2) has been reported in different types of cancers including glioblastoma multiforme (GBM). In this paper, to target IL13Rα2 on GBM cells, a fusion protein was generated comprising human IL13 and staphylococcal enterotoxin B (SEB), termed IL13-linker-SEB. The fusion protein was cloned into pET28a(+) and expressed in Escherichia coli strain BL21 (DE3); U251 (IL13Rα2-positive) and T98G (IL13Rα2-negative) GBM cell lines were employed and the functional activity of IL13-linker-SEB was evaluated by cell ELISA, cytotoxicity (MTT and LDH), apoptosis (flow cytometry and caspase-3 activity), adhesion, scratch and RT-PCR tests. SEB and chemotherapeutic drugs were employed to be compared to IL13-linker-SEB function. The IL13-linker-SEB exhibited higher binding affinity and cytotoxicity compared to SEB on U251 cells, although both recombinant proteins had shown similar behavior regarding T98G cells. Furthermore, the highest induction of apoptosis was observed in U251 cells treated with IL13-linker-SEB which was confirmed by Bax/Bcl-2 ratio. The expression of MMP2, MMP9 and VEGFR2 in U251 cells experienced a significant reduction after treatment with IL13-linker-SEB compared to SEB and T98G treated cells. The data showed that IL13-linker-SEB can be considered as a novel potential agent for GBM treatment; however, further research is needed to investigate the efficacy.


Assuntos
Glioblastoma , Subunidade alfa2 de Receptor de Interleucina-13 , Humanos , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Interleucina-13/genética , Interleucina-13/farmacologia , Interleucina-13/metabolismo , Subunidade alfa2 de Receptor de Interleucina-13/genética , Subunidade alfa2 de Receptor de Interleucina-13/metabolismo , Subunidade alfa2 de Receptor de Interleucina-13/uso terapêutico , Proteínas Recombinantes
3.
J Immunother Cancer ; 11(2)2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36759014

RESUMO

INTRODUCTION: The immunosuppressive tumor microenvironment (TME) is a major barrier to the efficacy of chimeric antigen receptor T cells (CAR-T cells) in glioblastoma (GBM). Transgenic expression of IL15 is one attractive strategy to modulate the TME. However, at present, it is unclear if IL15 could be used to directly target myeloid-derived suppressor cells (MDSCs), a major cellular component of the GBM TME. Here, we explored if MDSC express IL15Rα and the feasibility of exploiting its expression as an immunotherapeutic target. METHODS: RNA-seq, RT-qPCR, and flow cytometry were used to determine IL15Rα expression in paired peripheral and tumor-infiltrating immune cells of GBM patients and two syngeneic murine GBM models. We generated murine T cells expressing IL13Rα2-CARs and secretory IL15 (CAR.IL15s) or IL13Rα2-CARs in which IL15 was fused to the CAR to serve as an IL15Rα-targeting moiety (CAR.IL15f), and characterized their effector function in vitro and in syngeneic IL13Rα2+glioma models. RESULTS: IL15Rα was preferentially expressed in myeloid, B, and dendritic cells in patients' and syngeneic GBMs. In vitro, CAR.IL15s and CAR.IL15f T cells depleted MDSC and decreased their secretion of immunosuppressive molecules with CAR.IL15f T cells being more efficacious. Similarly, CAR.IL15f T cells significantly improved the survival of mice in two GBM models. TME analysis showed that treatment with CAR.IL15f T cells resulted in higher frequencies of CD8+T cells, NK, and B cells, but a decrease in CD11b+cells in tumors compared with therapy with CAR T cells. CONCLUSIONS: We demonstrate that MDSC of the glioma TME express IL15Ra and that these cells can be targeted with secretory IL15 or an IL15Rα-targeting moiety incorporated into the CAR. Thus, IL15-modified CAR T cells act as a dual targeting agent against tumor cells and MDSC in GBM, warranting their future evaluation in early-phase clinical studies.


Assuntos
Glioblastoma , Glioma , Subunidade alfa2 de Receptor de Interleucina-13 , Células Supressoras Mieloides , Animais , Camundongos , Glioma/tratamento farmacológico , Subunidade alfa2 de Receptor de Interleucina-13/uso terapêutico , Interleucina-15 , Células Supressoras Mieloides/metabolismo , Microambiente Tumoral , Linfócitos T
4.
Cancer Rep (Hoboken) ; 6(2): e1701, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36806727

RESUMO

BACKGROUND: Several treatment strategies use upfront chemotherapy or androgen receptor axis-targeting therapies for metastatic prostate cancer. However, there are no useful biomarkers for selecting appropriate patients who urgently require these treatments. METHODS: Novel patient-derived xenograft (PDX) castration-sensitive and -resistant models were established and gene expression patterns were comprehensively compared. The function of a gene highly expressed in the castration-resistant models was evaluated by its overexpression in LNCaP prostate cancer cells. Protein expression in the tumors and serum of patients was examined by immunohistochemistry and ELISA, and correlations with castration resistance were analyzed. RESULTS: Expression of the α2 chain of interleukin-13 receptor (IL13Rα2) was higher in castration-resistant PDX tumors. LNCaP cells overexpressing IL13Rα2 acquired castration resistance in vitro and in vivo. In tissue samples, IL13Rα2 expression levels were significantly associated with castration-resistant progression (p < 0.05). In serum samples, IL13Rα2 levels could be measured in 5 of 28 (18%) castration-resistant prostate cancer patients. CONCLUSION: IL13Rα2 was highly expressed in castration-resistant prostate cancer PDX models and was associated with the castration resistance of prostate cancer cells. It might be a potential tissue and serum biomarker for predicting castration resistance in prostate cancer patients.


Assuntos
Subunidade alfa2 de Receptor de Interleucina-13 , Neoplasias de Próstata Resistentes à Castração , Masculino , Humanos , Neoplasias de Próstata Resistentes à Castração/patologia , Subunidade alfa2 de Receptor de Interleucina-13/uso terapêutico , Xenoenxertos , Orquiectomia , Biomarcadores
5.
Biochim Biophys Acta Rev Cancer ; 1877(5): 188802, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36152905

RESUMO

Interleukin 13 receptor alpha 2 (IL13Rα2) is increasingly recognized as a relevant player in cancer invasion and metastasis. Despite being initially considered a decoy receptor for dampening the levels of interleukin 13 (IL-13) in diverse inflammatory conditions, accumulating evidences in the last decades indicate the capacity of IL13Rα2 for mediating IL-13 signaling in cancer cells. The biological reasons behind the expression of this receptor with such extremely high affinity for IL-13 in cancer cells remain unclear. Elevated expression of IL13Rα2 is commonly associated with invasion, late stage and cancer metastasis that results in poor prognosis for glioblastoma, colorectal or breast cancer, among others. The discovery of new mediators and effectors of IL13Rα2 signaling has been critical for deciphering its underlying molecular mechanisms in cancer progression. Still, many questions about the effects of inflammation, the cancer type and the tumor degree in the expression of IL13Rα2 remain largely uncharacterized. Here, we review and discuss the current status of the IL13Rα2 biology in cancer, with particular emphasis in the role of inflammation-driven expression and the regulation of different signaling pathways. As IL13Rα2 implications in cancer continue to grow exponentially, we highlight new targeted therapies recently developed for glioblastoma, colorectal cancer and other IL13Rα2-positive tumors.


Assuntos
Glioblastoma , Subunidade alfa2 de Receptor de Interleucina-13 , Glioblastoma/patologia , Humanos , Inflamação , Interleucina-13/uso terapêutico , Subunidade alfa2 de Receptor de Interleucina-13/genética , Subunidade alfa2 de Receptor de Interleucina-13/metabolismo , Subunidade alfa2 de Receptor de Interleucina-13/uso terapêutico , Transdução de Sinais
7.
Drug Deliv ; 29(1): 1620-1630, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35612318

RESUMO

Despite surgical and therapeutic advances, glioblastoma multiforme (GBM) is among the most fatal primary brain tumor that is aggressive in nature. Patients with GBM have a median lifespan of just 15 months when treated with the current standard of therapy, which includes surgical resection and concomitant chemo-radiotherapy. In recent years, nanotechnology has shown considerable promise in treating a variety of illnesses, and certain nanomaterials have been proven to pass the blood-brain barrier (BBB) and stay in glioblastoma tissues. Recent preclinical research suggests that the diagnosis and treatment of brain tumor is significantly explored through the intervention of nanomaterials that has showed enhanced effect. In order to elicit an antitumor response, it is necessary to retain the therapeutic candidates within glioblastoma tissues and this job is effectively carried out by nanocarrier particularly functionalized nanocarriers. In the arena of neoplastic diseases including GBM have achieved great attention in recent decades. Furthermore, interleukin-13 receptor α chain variant 2 (IL13Rα2) is a highly expressed and studied target in GBM that is lacked by the surrounding environment. The absence of IL13Rα2 in surrounding normal tissues has made it a suitable target in glioblastoma therapy. In this review article, we highlighted the role of IL13Rα2 as a potential target in GBM along with design and fabrication of efficient targeting strategies for IL13Rα2 through surface functionalized nanocarriers.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Subunidade alfa2 de Receptor de Interleucina-13 , Barreira Hematoencefálica/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Glioblastoma/tratamento farmacológico , Humanos , Subunidade alfa2 de Receptor de Interleucina-13/metabolismo , Subunidade alfa2 de Receptor de Interleucina-13/uso terapêutico
8.
Pancreas ; 51(9): 1133-1139, 2022 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37078936

RESUMO

OBJECTIVES: Pancreatic ductal adenocarcinoma with strong expression of interleukin-13 receptor α2 (IL-13Rα2) was associated with poor prognosis and gemcitabine resistance in an orthotopic mouse model. We evaluated the influence of IL-13Rα2 expression in the endoscopic ultrasound-fine needle aspiration (EUS-FNA) specimen. METHODS: We included patients with pancreatic ductal adenocarcinoma, as diagnosed by EUS-FNA, who received gemcitabine-based chemotherapy (G-CTX). Tumor expression of IL-13Rα2 was assessed by immunohistochemistry and classified using a three scale (negative, weak, or strong) in a blinded fashion. The effect of G-CTX was assessed by tumor reduction rate by computed tomography after 3 months. RESULTS: A total of 95 patients were enrolled, and 63 and 32 cases were determined with strong and weak/negative expression of IL-13Rα2. The IL-13Rα2-strong group showed significantly poorer progression-free and overall survival rates than weak/negative group (P = 0.0191 and P = 0.0062, respectively). Strong expression of IL-13Rα2 was associated with progression factor after 3 months of the first G-CTX (odds ratio, 13.72; P = 0.0143). CONCLUSIONS: Pancreatic ductal adenocarcinoma with strong expression of IL-13Rα2 in EUS-FNA specimens showed poor prognosis and poor response to G-CTX.


Assuntos
Adenocarcinoma , Carcinoma Ductal Pancreático , Subunidade alfa2 de Receptor de Interleucina-13 , Neoplasias Pancreáticas , Animais , Camundongos , Neoplasias Pancreáticas/patologia , Gencitabina , Subunidade alfa2 de Receptor de Interleucina-13/metabolismo , Subunidade alfa2 de Receptor de Interleucina-13/uso terapêutico , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/patologia , Carcinoma Ductal Pancreático/patologia , Aspiração por Agulha Fina Guiada por Ultrassom Endoscópico/métodos , Neoplasias Pancreáticas
9.
Clin Cancer Res ; 27(22): 6209-6221, 2021 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-34544797

RESUMO

PURPOSE: The survival of women with brain metastases (BM) from breast cancer remains very poor, with over 80% dying within a year of their diagnosis. Here, we define the function of IL13Rα2 in outgrowth of breast cancer brain metastases (BCBM) in vitro and in vivo, and postulate IL13Rα2 as a suitable therapeutic target for BM. EXPERIMENTAL DESIGN: We performed IHC staining of IL13Rα2 in BCBM to define its prognostic value. Using inducible shRNAs in TNBC and HER2+ breast-brain metastatic models, we assessed IL13Rα2 function in vitro and in vivo. We performed RNAseq and functional studies to define the molecular mechanisms underlying IL13Rα2 function in BCBM. RESULTS: High IL13Rα2 expression in BCBM predicted worse survival after BM diagnoses. IL13Rα2 was essential for cancer-cell survival, promoting proliferation while repressing invasion. IL13Rα2 KD resulted in FAK downregulation, repression of cell cycle and proliferation mediators, and upregulation of Ephrin B1 signaling. Ephrin-B1 (i) promoted invasion of BC cells in vitro, (ii) marked micrometastasis and invasive fronts in BCBM, and (iii) predicted shorter disease-free survival and BM-free survival (BMFS) in breast primary tumors known to metastasize to the brain. In experimental metastases models, which bypass early tumor invasion, downregulation of IL13Rα2 before or after tumor seeding and brain intravasation decreased BMs, suggesting that IL13Rα2 and the promotion of a proliferative phenotype is critical to BM progression. CONCLUSIONS: Non-genomic phenotypic adaptations at metastatic sites are critical to BM progression and patients' prognosis. This study opens the road to use IL13Rα2 targeting as a therapeutic strategy for BM.


Assuntos
Neoplasias Encefálicas , Neoplasias da Mama , Subunidade alfa2 de Receptor de Interleucina-13 , Neoplasias Encefálicas/patologia , Neoplasias da Mama/patologia , Proliferação de Células/genética , Feminino , Humanos , Subunidade alfa2 de Receptor de Interleucina-13/genética , Subunidade alfa2 de Receptor de Interleucina-13/uso terapêutico , Prognóstico
10.
J Orthop Surg Res ; 14(1): 386, 2019 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-31775818

RESUMO

BACKGROUND: The incidence of degenerative disc disease caused by intervertebral disc injury is increasing annually, seriously affecting the quality of life of patients and increasing the disease burden on society. The mechanisms of intervertebral disc degeneration include changes in extracellular matrix (ECM) deposition and tissue fibrosis. sIL-13Rα2-Fc potently inhibits interleukin (IL)-13, as well as blocks related cell signaling pathways and inhibits fibrosis in certain tissues. However, it is unknown whether sIL-13Rα2-Fc inhibits fibrosis in injured intervertebral discs and slows the process of degeneration. We hypothesized that sIL-13Rα2-Fc delays the progression of intervertebral disc degeneration by inhibiting intervertebral disc fibrosis and improving ECM deposition. METHODS: A rat tail intervertebral disc degeneration model was established. Pathological changes in rat intervertebral disc tissue were observed by hematoxylin and eosin staining and Masson staining. Glycosaminoglycan (GAG), chondroitin sulfate (CS), keratan sulfate (KS), and hyaluronic acid (HA) contents were quantitatively analyzed by enzyme-linked immunosorbent assay. Type I and type II collagen expression levels were analyzed by reverse transcription-PCR and western blotting. RESULTS: Hematoxylin and eosin staining and Masson staining revealed annulus fibrosus rupture, disordered arrangement, decreased nucleus pulposus tissue, and decreased collagen fiber in the rat intervertebral disc tissue. Following treatment with sIL-13Rα2-Fc, pathological changes in the rat intervertebral disc were reduced. Rat intervertebral disc tissue showed decreased GAG, CS-KS, and (HA) contents, increased type I collagen levels, and decreased type II collagen levels in degenerated intervertebral discs. sIL-13Rα2-Fc intervention increased the contents of GAG, CS, KS, and HA; inhibited the expression of type I collagen; and promoted the expression of type II collagen. CONCLUSION: These results demonstrate that intervertebral disc degeneration is associated with tissue fibrosis. sIL-13Rα2-Fc can regulate type I and type II collagen expression levels by increasing GAG, CS, KS, and HA contents, thereby slowing the progression of intervertebral disc degeneration.


Assuntos
Subunidade alfa2 de Receptor de Interleucina-13/uso terapêutico , Degeneração do Disco Intervertebral/tratamento farmacológico , Animais , Avaliação Pré-Clínica de Medicamentos , Fragmentos Fc das Imunoglobulinas/uso terapêutico , Disco Intervertebral/efeitos dos fármacos , Disco Intervertebral/metabolismo , Ratos Sprague-Dawley , Cauda
11.
Br J Cancer ; 119(8): 940-949, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30318506

RESUMO

BACKGROUND: Interleukin 13 receptor α2 (IL13Rα2) is overexpressed in metastatic colorectal cancer. Here, we have developed novel strategies to block IL-13 binding to IL13Rα2 in order to reduce metastatic spread. METHODS: Synthetic IL13Rα2 D1 peptide (GSETWKTIITKN) was tested for the inhibition of IL-13 binding to IL13Rα2 using ELISA and different cellular assays. Peptide blocking effects on different cell signalling mediators were determined by western blot. An enantiomer version of the peptide (D-D1) was prepared to avoid proteolytic digestion. Nude mice were used for tumour growth and survival analysis after treatment with IL13Rα2 peptides. RESULTS: IL13Rα2 D1 peptide inhibited migration, invasion, and proliferation in metastatic colorectal and glioblastoma cancer cells treated with IL-13. Residues 82K, 83T, 85I and 86T were essential for blocking IL-13. IL13Rα2 peptide abolished ligand-mediated receptor internalisation and degradation, and substantially decreased IL-13 signalling capacity through IL13Rα2 to activate the FAK, PI3K/AKT and Src pathways as well as MT1-MMP expression. In addition, D1 significantly inhibited IL-13-mediated STAT6 activation through IL13Rα1. Nude mice treated with the enantiomer D-D1 peptide showed a remarkable survival increase. CONCLUSIONS: We propose that the D-D1 peptide from IL13Rα2 represents a promising therapeutic agent to inhibit metastatic progression in colorectal cancer and, likely, other solid tumours.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Colorretais/terapia , Glioblastoma/terapia , Subunidade alfa2 de Receptor de Interleucina-13/metabolismo , Subunidade alfa2 de Receptor de Interleucina-13/uso terapêutico , Interleucina-13/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/patologia , Quinase 1 de Adesão Focal/metabolismo , Glioblastoma/patologia , Células HT29 , Humanos , Subunidade alfa2 de Receptor de Interleucina-13/genética , Metaloproteinase 14 da Matriz/biossíntese , Camundongos , Camundongos Nus , Fosfatidilinositol 3-Quinases/metabolismo , Ligação Proteica/efeitos dos fármacos , Interferência de RNA , RNA Interferente Pequeno/genética , Fator de Transcrição STAT6/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Cancer Immunol Res ; 5(7): 571-581, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28550091

RESUMO

Glioblastoma (GBM) is the most aggressive primary brain tumor in adults and is virtually incurable with conventional therapies. Immunotherapy with T cells expressing GBM-specific chimeric antigen receptors (CAR) is an attractive approach to improve outcomes. Although CAR T cells targeting GBM antigens, such as IL13 receptor subunit α2 (IL13Rα2), HER2, and EGFR variant III (EGFRvIII), have had antitumor activity in preclinical models, early-phase clinical testing has demonstrated limited antiglioma activity. Transgenic expression of IL15 is an appealing strategy to enhance CAR T-cell effector function. We tested this approach in our IL13Rα2-positive glioma model in which limited IL13Rα2-CAR T-cell persistence results in recurrence of antigen-positive gliomas. T cells were genetically modified with retroviral vectors encoding IL13Rα2-CARs or IL15 (IL13Rα2-CAR.IL15 T cells). IL13Rα2-CAR.IL15 T cells recognized glioma cells in an antigen-dependent fashion, had greater proliferative capacity, and produced more cytokines after repeated stimulations in comparison with IL13Rα2-CAR T cells. No autonomous IL13Rα2-CAR.IL15 T-cell proliferation was observed; however, IL15 expression increased IL13Rα2-CAR T-cell viability in the absence of exogenous cytokines or antigen. In vivo, IL13Rα2-CAR.IL15 T cells persisted longer and had greater antiglioma activity than IL13Rα2-CAR T cells, resulting in a survival advantage. Gliomas recurring after 40 days after T-cell injection had downregulated IL13Rα2 expression, indicating that antigen loss variants occur in the setting of improved T-cell persistence. Thus, CAR T cells for GBM should not only be genetically modified to improve their proliferation and persistence, but also to target multiple antigens.Summary: Glioblastoma responds imperfectly to immunotherapy. Transgenic expression of IL15 in T cells expressing CARs improved their proliferative capacity, persistence, and cytokine production. The emergence of antigen loss variants highlights the need to target multiple tumor antigens. Cancer Immunol Res; 5(7); 571-81. ©2017 AACR.


Assuntos
Glioblastoma/imunologia , Imunoterapia Adotiva , Subunidade alfa2 de Receptor de Interleucina-13/imunologia , Interleucina-15/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Animais , Antígenos de Neoplasias/imunologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Glioblastoma/genética , Glioblastoma/terapia , Humanos , Subunidade alfa2 de Receptor de Interleucina-13/genética , Subunidade alfa2 de Receptor de Interleucina-13/uso terapêutico , Interleucina-15/genética , Interleucina-15/uso terapêutico , Ativação Linfocitária/imunologia , Camundongos , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Clin Cancer Res ; 21(18): 4062-72, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26059190

RESUMO

PURPOSE: A first-in-human pilot safety and feasibility trial evaluating chimeric antigen receptor (CAR)-engineered, autologous primary human CD8(+) cytotoxic T lymphocytes (CTL) targeting IL13Rα2 for the treatment of recurrent glioblastoma (GBM). EXPERIMENTAL DESIGN: Three patients with recurrent GBM were treated with IL13(E13Y)-zetakine CD8(+) CTL targeting IL13Rα2. Patients received up to 12 local infusions at a maximum dose of 10(8) CAR-engineered T cells via a catheter/reservoir system. RESULTS: We demonstrate the feasibility of manufacturing sufficient numbers of autologous CTL clones expressing an IL13(E13Y)-zetakine CAR for redirected HLA-independent IL13Rα2-specific effector function for a cohort of patients diagnosed with GBM. Intracranial delivery of the IL13-zetakine(+) CTL clones into the resection cavity of 3 patients with recurrent disease was well-tolerated, with manageable temporary brain inflammation. Following infusion of IL13-zetakine(+) CTLs, evidence for transient anti-glioma responses was observed in 2 of the patients. Analysis of tumor tissue from 1 patient before and after T-cell therapy suggested reduced overall IL13Rα2 expression within the tumor following treatment. MRI analysis of another patient indicated an increase in tumor necrotic volume at the site of IL13-zetakine(+) T-cell administration. CONCLUSIONS: These findings provide promising first-in-human clinical experience for intracranial administration of IL13Rα2-specific CAR T cells for the treatment of GBM, establishing a foundation on which future refinements of adoptive CAR T-cell therapies can be applied.


Assuntos
Neoplasias Encefálicas/terapia , Linfócitos T CD8-Positivos/imunologia , Glioblastoma/terapia , Imunoterapia Adotiva/métodos , Subunidade alfa2 de Receptor de Interleucina-13/uso terapêutico , Receptores de Antígenos de Linfócitos T/uso terapêutico , Adulto , Idoso , Encéfalo/patologia , Neoplasias Encefálicas/imunologia , Linfócitos T CD8-Positivos/citologia , Estudos de Viabilidade , Feminino , Glioblastoma/imunologia , Glioma/imunologia , Glioma/terapia , Antígenos HLA/química , Humanos , Inflamação , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Segurança do Paciente , Projetos Piloto , Receptores de Antígenos de Linfócitos T/química , Receptores de Antígenos de Linfócitos T/imunologia , Recidiva , Resultado do Tratamento , Adulto Jovem
14.
Neuro Oncol ; 16(10): 1304-12, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24723564

RESUMO

Glioblastoma multiforme (GBM) remains one of the most lethal primary brain tumors despite surgical and therapeutic advancements. Targeted therapies of neoplastic diseases, including GBM, have received a great deal of interest in recent years. A highly studied target of GBM is interleukin-13 receptor α chain variant 2 (IL13Rα2). Targeted therapies against IL13Rα2 in GBM include fusion chimera proteins of IL-13 and bacterial toxins, nanoparticles, and oncolytic viruses. In addition, immunotherapies have been developed using monoclonal antibodies and cell-based strategies such as IL13Rα2-pulsed dendritic cells and IL13Rα2-targeted chimeric antigen receptor-modified T cells. Advanced therapeutic development has led to the completion of phase I clinical trials for chimeric antigen receptor-modified T cells and phase III clinical trials for IL-13-conjugated bacterial toxin, with promising outcomes. Selective expression of IL13Rα2 on tumor cells, while absent in the surrounding normal brain tissue, has motivated continued study of IL13Rα2 as an important candidate for targeted glioma therapy. Here, we review the preclinical and clinical studies targeting IL13Rα2 in GBM and discuss new advances and promising applications.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Subunidade alfa2 de Receptor de Interleucina-13/uso terapêutico , Animais , Neoplasias Encefálicas/imunologia , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Exotoxinas/uso terapêutico , Glioblastoma/imunologia , Humanos , Interleucina-13/uso terapêutico , Subunidade alfa2 de Receptor de Interleucina-13/imunologia , Lipossomos/uso terapêutico , Camundongos , Terapia de Alvo Molecular/métodos , Proteínas Recombinantes de Fusão , Resultado do Tratamento
15.
J Immunol ; 187(10): 4935-46, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22013118

RESUMO

Optimum efficacy of therapeutic cancer vaccines may require combinations that generate effective antitumor immune responses, as well as overcome immune evasion and tolerance mechanisms mediated by progressing tumor. Previous studies showed that IL-13Rα2, a unique tumor-associated Ag, is a promising target for cancer immunotherapy. A targeted cytotoxin composed of IL-13 and mutated Pseudomonas exotoxin induced specific killing of IL-13Rα2(+) tumor cells. When combined with IL-13Rα2 DNA cancer vaccine, surprisingly, it mediated synergistic antitumor effects on tumor growth and metastasis in established murine breast carcinoma and sarcoma tumor models. The mechanism of synergistic activity involved direct killing of tumor cells and cell-mediated immune responses, as well as elimination of myeloid-derived suppressor cells and, consequently, regulatory T cells. These novel results provide a strong rationale for combining immunotoxins with cancer vaccines for the treatment of patients with advanced cancer.


Assuntos
ADP Ribose Transferases/uso terapêutico , Toxinas Bacterianas/uso terapêutico , Vacinas Anticâncer/uso terapêutico , Exotoxinas/uso terapêutico , Marcação de Genes/métodos , Imunoterapia/métodos , Subunidade alfa2 de Receptor de Interleucina-13/uso terapêutico , Neoplasias Mamárias Experimentais/terapia , Sarcoma Experimental/terapia , Vacinas de DNA/uso terapêutico , Fatores de Virulência/uso terapêutico , ADP Ribose Transferases/genética , ADP Ribose Transferases/imunologia , Animais , Toxinas Bacterianas/genética , Toxinas Bacterianas/imunologia , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Sinergismo Farmacológico , Quimioterapia Combinada , Exotoxinas/genética , Exotoxinas/imunologia , Feminino , Subunidade alfa2 de Receptor de Interleucina-13/biossíntese , Subunidade alfa2 de Receptor de Interleucina-13/genética , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Sarcoma Experimental/genética , Sarcoma Experimental/imunologia , Vacinas de DNA/imunologia , Fatores de Virulência/genética , Fatores de Virulência/imunologia
16.
Mol Ther ; 16(9): 1556-64, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18665158

RESUMO

The majority of glioblastoma multiforme (GBM) tumors (80%) overexpress interleukin-13 receptor alpha2 (IL-13Ralpha2), but there is no expression of IL-13Ralpha2 in normal brain. Vaccine strains of measles virus have significant antitumor activity against gliomas. We tested the hypothesis that measles virus entry could be retargeted via the IL-13Ralpha2. MV-GFP-H(AA)-IL-13 was generated from the Edmonston-NSe vaccine strain, by displaying human IL-13 at the C-terminus of the H protein, and introducing CD46 and signaling lymphocyte activation molecule (SLAM)-ablating mutations in H. The IL-13 retargeted virus showed significant cytopathic effect (CPE) against IL-13Ralpha2 overexpressing glioma lines, and lack of CPE/viral replication in normal human astrocytes and normal human fibroblasts not expressing IL-13Ralpha2. In vivo treatment of orthotopically implanted GBM12 xenografts demonstrated significant prolongation of survival in mice treated with the retargeted strain (P < 0.0001), and comparable activity between the IL-13R retargeted strain and MV-GFP (P = 0.6377). In contrast to MV-GFP-treated mice, administration of the retargeted strain in the central nervous system of measles replication-permissive Ifnar(ko) CD46 Ge mice resulted in lack of neurotoxicity. Strains of measles virus retargeted against the glioma-specific IL-13Ralpha2 receptor have comparable therapeutic efficacy, and improved specificity as compared with the unmodified measles virus strain MV-GFP in vitro and in vivo.


Assuntos
Neoplasias Encefálicas/terapia , Terapia Genética , Glioma/terapia , Subunidade alfa2 de Receptor de Interleucina-13/uso terapêutico , Interleucina-13/metabolismo , Vírus do Sarampo/fisiologia , Terapia Viral Oncolítica , Animais , Western Blotting , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/imunologia , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/virologia , Chlorocebus aethiops , Citometria de Fluxo , Glioma/genética , Glioma/imunologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/imunologia , Humanos , Subunidade alfa2 de Receptor de Interleucina-13/genética , Sarampo/genética , Sarampo/imunologia , Sarampo/terapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas do Nucleocapsídeo/metabolismo , Células Tumorais Cultivadas , Células Vero , Ensaios Antitumorais Modelo de Xenoenxerto
17.
J Exp Clin Cancer Res ; 26(3): 347-52, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17987794

RESUMO

Dalton's lymphoma (DL) is a T-cell lymphoma of spontaneous origin, characterized by highly invasive and malignant nature, killing the host in a very short period of life span. DL-bearing host is reflected by very high titer of IL-13 in serum. Therefore, we hypothesized that over expression of IL-13 may greatly affect the growth of DL-cells in a tumor-bearing host. In this study, to assess the involvement of IL-13 in DL-cell progression, we have blocked the IL-13 activity/signalling by the systemic delivery of non-signaling decoy receptor IL-13 Ralpha2, and IL-13 level vs DL-cell proliferation were measured. We observed that systemic delivery of IL-13 Ralpha2 inhibits the DL-cell progression in much extent and enhances the survival and longevity of DL-bearing mice. Further, this study re-inforce the therapeutic advantage of IL-13 Ralpha2 in a T-cell lymphoma tumor system.


Assuntos
Subunidade alfa2 de Receptor de Interleucina-13/uso terapêutico , Interleucina-13/antagonistas & inibidores , Linfoma de Células T/tratamento farmacológico , Animais , Proliferação de Células , Feminino , Interleucina-13/sangue , Interleucina-13/metabolismo , Subunidade alfa2 de Receptor de Interleucina-13/administração & dosagem , Linfoma de Células T/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Transdução de Sinais , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...