Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
2.
Ann Ig ; 32(6): 648-663, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33175076

RESUMO

BACKGROUND: Cancer is considered as one of the leading causes of death today. The wrong lifestyles have led to an increase in the incidence rate of this deadly disease. There are many complications associated with common treatments of this disease. Immunotherapy is one of the new approaches taken recently. The purpose of this systematic review was to evaluate the studies on Staphylococcus aureus enterotoxins as a treatment of cancer worldwide. STUDY DESIGN: We conducted a systematic review of articles published in PubMed, Cochrane, Scopus and Google scholar databases from 1995 to 2016 to evaluate the effects of Staphylococci enterotoxins on cancer. METHODS: Eligible studies were evaluated qualitatively based on a checklist prepared by two independent reviewers, and they were subsequently matched. RESULTS: Our review identified 97 records through searching PubMed and Cochrane database and 1306 records through searching Google scholar and Scopus. Forty six studies were excluded from PubMed and Cochrane database and 1281 studies were excluded from Google scholar and Scopus after screening abstracts and titles. Therefore, our systematic review was based on 51 publications on PubMed and Cochrane, and 25 publications on Google scholar and Scopus, which met our criteria. Staphylococcal enterotoxin A was the most commonly used toxin in these studies. The side effects of using this toxin in immunotherapy have been reported to be low and all studies have identified this toxin as a suitable option for immunotherapy. CONCLUSIONS: The data obtained from these studies showed that due to the low rates of complications, Staphylococcal enterotoxins have the potential to induce immune system against various cancers as super-antigens. Therefore, they can be considered as a suitable candidate for immunotherapy of cancer.


Assuntos
Enterotoxinas/uso terapêutico , Imunoterapia/métodos , Neoplasias/terapia , Adjuvantes Imunológicos/efeitos adversos , Adjuvantes Imunológicos/uso terapêutico , Animais , Ensaios Clínicos como Assunto , Ensaios de Seleção de Medicamentos Antitumorais , Enterotoxinas/efeitos adversos , Humanos , Camundongos , Neoplasias Experimentais/terapia , Coelhos , Projetos de Pesquisa , Resultado do Tratamento
3.
Eur J Pharmacol ; 887: 173596, 2020 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-32979353

RESUMO

Bifidobacterium is a nonpathogenic strain of anaerobic bacteria that selectively localizes and proliferates in tumors. It has emerged as a specific carrier of anticancer proteins against malignant tumors. Claudins are tetraspanin transmembrane proteins that form tight junctions. Claudin-4 is overexpressed in certain epithelial malignant cancers. The C-terminal fragment of the Clostridium perfringens enterotoxin (C-CPE), an exotoxin without the cytotoxic domain, strongly binds to claudin-4. The C-CPE fusion toxin (C-CPE-PE23), which targets claudin-4, strongly suppresses tumor growth; however, C-CPE fusion toxins exhibit hepatic toxicity. In this study, we successfully generated a strain of Bifidobacterium longum that secreted C-CPE-PE23 (B. longum-C-CPE-PE23) and was specific to and cross reactive with human and mouse claudin-4. We evaluated the therapeutic potential of this strain against triple-negative breast cancer using a mouse model. C-CPE-PE23 decreased cell viability in a dose-dependent manner in human and mouse breast cancer cell lines. After intravenous injection, Bifidobacterium was specifically distributed in the tumors of mice bearing breast cancer tumors. Moreover, B. longum-C-CPE-PE23 significantly suppressed tumor growth in mice with breast cancer without serious side effects, such as weight loss or hepatic and renal damage. We suggest that B. longum-C-CPE-PE23 is a good candidate for breast cancer treatment. Bifidobacterium could also be used as a drug delivery system for hepatotoxic agents.


Assuntos
Antineoplásicos/administração & dosagem , Antineoplásicos/uso terapêutico , Bifidobacterium/metabolismo , Claudinas/metabolismo , Sistemas de Liberação de Medicamentos , Neoplasias de Mama Triplo Negativas/terapia , Animais , Linhagem Celular Tumoral , Claudina-4/metabolismo , DNA Recombinante , Relação Dose-Resposta a Droga , Enterotoxinas/administração & dosagem , Enterotoxinas/uso terapêutico , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Plasmídeos/genética
4.
Prep Biochem Biotechnol ; 50(1): 47-55, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31478797

RESUMO

Clostridium perfringens enterotoxin (CPE) has anti-prostate cancer effects and the prostate stem cell antigen (PSCA) has been used as a plasmid-based vaccine. So we expressed both of them in the PC3 cells to evaluate their effects on cell cycling and apoptosis. The PC3 cells were transfected either by the pBudCE4.1-CPE-PSCA or empty plasmid. The expression of the cpe and PSCA genes in transfected PC3 was evaluated. The apoptosis genes (Fas, P53, Bak, and Bax) as well as cell cycling genes (cyclin D1 and E) expression was evaluated by qPCR. Successful expression of cpe and PSCA in PC3 cells was confirmed. The flow cytometry results showed the cellular death rates of 62.6% and 21.8% for PC3 cells transformed with recombinant and empty plasmids respectively. Bak, Fas, Bax and P53 genes were significantly upregulated in PC3 cells transformed with pBudCE4.1-CPE-PSCA, while cyclin D1 and E were downregulated when compared with the pBudCE4.1-transfected PC3 and normal cells (p < .05). The results showed the lethal consequences of cpe and PSCA genes expression on PC3 transfected cells. Expression of the cpe and PSCA genes affects the PC3 cell death so it could be a suitable candidate for further researches in prostate cancer vaccine development.


Assuntos
Antígenos de Neoplasias/genética , Apoptose , Ciclo Celular , Enterotoxinas/genética , Vetores Genéticos/genética , Proteínas de Neoplasias/genética , Neoplasias da Próstata/terapia , Antígenos de Neoplasias/uso terapêutico , Enterotoxinas/uso terapêutico , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/uso terapêutico , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Terapia Genética/métodos , Vetores Genéticos/uso terapêutico , Humanos , Masculino , Proteínas de Neoplasias/uso terapêutico , Células PC-3 , Neoplasias da Próstata/genética , Transfecção/métodos
5.
Mol Oncol ; 14(2): 261-276, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31825142

RESUMO

Clostridium perfringens enterotoxin (CPE) can be used to eliminate carcinoma cells that overexpress on their cell surface CPE receptors - a subset of claudins (e.g., Cldn3 and Cldn4). However, CPE cannot target tumors expressing solely CPE-insensitive claudins (such as Cldn1 and Cldn5). To overcome this limitation, structure-guided modifications were used to generate CPE variants that can strongly bind to Cldn1, Cldn2 and/or Cldn5, while maintaining the ability to bind Cldn3 and Cldn4. This enabled (a) targeting of the most frequent endocrine malignancy, namely, Cldn1-overexpressing thyroid cancer, and (b) improved targeting of the most common cancer type worldwide, non-small-cell lung cancer (NSCLC), which is characterized by high expression of several claudins, including Cldn1 and Cldn5. Different CPE variants, including the novel mutant CPE-Mut3 (S231R/S313H), were applied on thyroid cancer (K1 cells) and NSCLC (PC-9 cells) models. In vitro, CPE-Mut3, but not CPEwt, showed Cldn1-dependent binding and cytotoxicity toward K1 cells. For PC-9 cells, CPE-Mut3 improved claudin-dependent cytotoxic targeting, when compared to CPEwt. In vivo, intratumoral injection of CPE-Mut3 in xenograft models bearing K1 or PC-9 tumors induced necrosis and reduced the growth of both tumor types. Thus, directed modification of CPE enables eradication of tumor entities that cannot be targeted by CPEwt, for instance, Cldn1-overexpressing thyroid cancer by using the novel CPE-Mut3.


Assuntos
Antineoplásicos/farmacologia , Claudinas/metabolismo , Clostridium perfringens/metabolismo , Enterotoxinas/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias da Glândula Tireoide/tratamento farmacológico , Animais , Antineoplásicos/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/terapia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Claudina-1/química , Claudina-1/genética , Claudina-1/metabolismo , Claudina-3/química , Claudina-3/genética , Claudina-3/metabolismo , Claudina-4/química , Claudina-4/genética , Claudina-4/metabolismo , Claudina-5/química , Claudina-5/genética , Claudina-5/metabolismo , Claudinas/química , Claudinas/genética , Enterotoxinas/química , Enterotoxinas/uso terapêutico , Feminino , Humanos , Imuno-Histoquímica , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/terapia , Camundongos , Mutagênese Sítio-Dirigida , Mutação , Necrose/induzido quimicamente , Ligação Proteica , Proteínas Recombinantes , Neoplasias da Glândula Tireoide/metabolismo , Neoplasias da Glândula Tireoide/terapia , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Biomed Pharmacother ; 110: 190-196, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30471512

RESUMO

BACKGROUND: TGFαL3-SEB chimeric protein is a synthetic protein, which is produced by combining the third loop (L3) of transforming growth factor-α (TGF-α) with staphylococcal enterotoxin type B. To the best of our knowledge, anti-cancer activity of this chimeric protein against colon cancer that overexpresses epidermal growth factor receptor (EGFR) has not yet been studied. Thus, in the present study, the anti-tumor effects of TGFαL3-SEB chimeric protein on HT-29 colon cancer cells were evaluated. MATERIALS AND METHODS: The TGFαL3-SEB chimeric protein was previously designed and cloned in Escherichia coli (E. coli) [1,2]. The level of expression and the purity of this novel protein were examined for further analysis. For this purpose, the cells were treated with different concentrations (25, 50 and 75 µg/ml) of TGFαL3-SEB and then the proliferation was detected using the MTT assay. The apoptosis-inducing potential of TGFαL3-SEB in HT-29 and HEK-293 cells was evaluated by flow cytometry using Annexin V/PI double staining method; in addition, bax/bcl2 mRNA ratio, caspase-3 and caspase-9 activity were also assessed. RESULTS: In the present study, TGFαL3-SEB chimeric protein was produced in E. coli. After effective purification, its growth inhibitory effect was evaluated. Our results indicated that the incubation of HT-29 colon cancer cell with 25, 50 and 75 µg/ml of TGFαL3-SEB for 24 h leads to significant reduction of proliferation in a dose-dependent manner (P < 0.05). Further analysis indicated that exposure of EGFR expressing HT-29 cells to TGFαL3-SEB leads to significant increase of the caspase-3 and caspase-9 activity in a concentration-dependent manner (P < 0.05). Bax/bcl-2 ratio also confirmed that TGFαL3-SEB has the pro-apoptotic effect. Flow cytometry analysis of TGFαL3-SEB treated cells showed that in addition to apoptotic cells, necrotic cells were also increased significantly at the concentration of 25, 50 and 75 µg/ml (P < 0.05). CONCLUSION: In conclusion, our results demonstrated that TGFαL3-SEB chimeric protein induced cell death through both mechanisms of apoptosis and necrosis in HT-29 colon cancer cells. This paper has highlighted that TGFαL3-SEB has the potential to target EGFR expressing cancer cell.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias do Colo , Enterotoxinas/uso terapêutico , Inibidores do Crescimento/uso terapêutico , Fator de Crescimento Transformador alfa/uso terapêutico , Apoptose/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/patologia , Relação Dose-Resposta a Droga , Enterotoxinas/farmacologia , Escherichia coli/efeitos dos fármacos , Escherichia coli/fisiologia , Inibidores do Crescimento/farmacologia , Células HEK293 , Células HT29 , Humanos , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes de Fusão/uso terapêutico , Fator de Crescimento Transformador alfa/farmacologia
7.
Sci Rep ; 8(1): 14963, 2018 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-30297847

RESUMO

A recombinant produced C-terminus of the C. perfringens enterotoxin (C-CPE) was conjugated to gold nanoparticles (AuNPs) to produce a C-CPE-AuNP complex (C-CPE-AuNP). By binding to claudins, the C- CPE should allow to target the AuNPs onto the claudin expressing tumor cells for a subsequent cell killing by application of the gold nanoparticle-mediated laser perforation (GNOME-LP) technique. Using qPCR and immunocytochemistry, we identified the human Caco-2, MCF-7 and OE-33 as well as the canine TiHoDMglCarc1305 as tumor cells expressing claudin-3, -4 and -7. Transepithelial electrical resistance (TEER) measurements of Caco-2 cell monolayer showed that the recombinant C-CPE bound to the claudins. GNOME-LP at a laser fluence of 60 mJ/cm2 and a scanning speed of 0.5 cm/s specifically eliminated more than 75% of claudin expressing human and canine cells treated with C-CPE-AuNP. The same laser fluence did not affect the cells when non-functionalized AuNPs were used. Furthermore, most of the claudin non-expressing cells treated with C-CPE-AuNP were not killed by GNOME-LP. Additionally, application of C-CPE-AuNP to spheroids formed by MCF-7 and OE-33 cells grown in Matrigel reduced spheroid area. The results demonstrate that specific ablation of claudin expressing tumor cells is efficiently increased by activated C-CPE functionalized AuNPs using optical methods.


Assuntos
Enterotoxinas/uso terapêutico , Ouro/uso terapêutico , Nanopartículas Metálicas/uso terapêutico , Neoplasias/terapia , Linhagem Celular Tumoral , Claudinas/metabolismo , Clostridium perfringens/química , Enterotoxinas/química , Ouro/química , Humanos , Terapia a Laser/métodos , Nanopartículas Metálicas/química , Neoplasias/metabolismo
8.
BMC Cancer ; 17(1): 129, 2017 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-28193196

RESUMO

BACKGROUND: Bacterial toxins have evolved to an effective therapeutic option for cancer therapy. The Clostridium perfringens enterotoxin (CPE) is a pore-forming toxin with selective cytotoxicity. The transmembrane tight junction proteins claudin-3 and -4 are known high affinity CPE receptors. Their expression is highly upregulated in human cancers, including breast, ovarian and colon carcinoma. CPE binding to claudins triggers membrane pore complex formation, which leads to rapid cell death. Previous studies demonstrated the anti-tumoral effect of treatment with recombinant CPE-protein. Our approach aimed at evaluation of a selective and targeted cancer gene therapy of claudin-3- and/or claudin-4- expressing colon carcinoma in vitro and in vivo by using translation optimized CPE expressing vector. METHODS: In this study the recombinant CPE and a translation optimized CPE expressing vector (optCPE) was used for targeted gene therapy of claudin-3 and/or -4 overexpressing colon cancer cell lines. All experiments were performed in the human SW480, SW620, HCT116, CaCo-2 and HT-29 colon cancer and the isogenic Sk-Mel5 and Sk-Mel5 Cldn-3-YFP melanoma cell lines. Claudin expression analysis was done at protein and mRNA level, which was confirmed by immunohistochemistry. The CPE induced cytotoxicity was analyzed by the MTT cytotoxicity assay. In addition patient derived colon carcinoma xenografts (PDX) were characterized and used for the intratumoral in vivo gene transfer of the optCPE expressing vector in PDX bearing nude mice. RESULTS: Claudin-3 and -4 overexpressing colon carcinoma lines showed high sensitivity towards both recCPE application and optCPE gene transfer. The positive correlation between CPE cytotoxicity and level of claudin expression was demonstrated. Transfection of optCPE led to targeted, rapid cytotoxic effects such as membrane disruption and necrosis in claudin overexpressing cells. The intratumoral optCPE in vivo gene transfer led to tumor growth inhibition in colon carcinoma PDX bearing mice in association with massive necrosis due to the intratumoral optCPE expression. CONCLUSIONS: This novel approach demonstrates that optCPE gene transfer represents a promising and efficient therapeutic option for a targeted suicide gene therapy of claudin-3 and/or claudin-4 overexpressing colon carcinomas, leading to rapid and effective tumor cell killing in vitro and in vivo.


Assuntos
Claudina-3/genética , Claudina-4/genética , Neoplasias do Colo/terapia , Enterotoxinas/uso terapêutico , Genes Transgênicos Suicidas , Terapia Genética , Animais , Efeito Espectador , Clostridium perfringens , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Feminino , Humanos , Camundongos , Camundongos Nus , Ensaios Antitumorais Modelo de Xenoenxerto
9.
J Orthop Res ; 35(6): 1215-1225, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-27431811

RESUMO

Distraction osteogenesis (DO) technique could be used to manage large-size bone defect successfully, but DO process usually requires long duration of bone consolidation. Innovative approaches for augmenting bone consolidation are of great need. Staphylococcal enterotoxin C2 (SEC2) has been found to suppress osteoclastogenesis of mesenchymal stem cells in vitro. In this study, we investigated the effect of SEC2 on proliferation and osteogenic differentiation of rat bone marrow derived mesenchymal stem cells (rBMSCs). Further, we locally administrated SEC2 (10 ng/ml) or PBS into the distraction gap in Sprague-Dawley male rat DO model every 3 days till termination at 3 and 6 weeks. The regenerates were subjected to X-rays, micro-computed tomography, mechanical testing, histology, and immunohischemistry examinations to assess new bone quality. SEC2 had no effect on cell viability. The calcium deposition was remarkably increased and osteogenic marker genes were significantly up-regulated in rBMSCs treated with SEC2. In rat DO model, SEC2 group had higher bone volume/total tissue volume in the regenerates. At 6 weeks, mechanical properties were significantly higher in SEC2-treated tibiae comparing to the control group. Histological analysis confirmed that the new bone had improved quality in SEC2 treated group, where the osteocalcin and osterix expression in the regenerates was up-regulated, indicating faster bone formation. The current study demonstrated that SEC2 local injection promotes osteogenesis and enhanced bone consolidation in DO. The findings support application of SEC2 as a potential novel strategy to expedite bone consolidation in patients undergoing DO treatment. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:1215-1225, 2017.


Assuntos
Células da Medula Óssea/efeitos dos fármacos , Enterotoxinas/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteogênese por Distração , Osteogênese/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Módulo de Elasticidade , Enterotoxinas/uso terapêutico , Interleucinas/sangue , Masculino , Cultura Primária de Células , Ratos Sprague-Dawley , Microtomografia por Raio-X
10.
Pflugers Arch ; 469(1): 45-53, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27629072

RESUMO

Given that most malignant tumors are derived from epithelium, developing a strategy for treatment of epithelium-derived cancers (i.e., carcinomas) is a pivotal issue in cancer therapy. Carcinomas, including ovarian, breast, prostate, and pancreatic cancers, are known to overexpress various claudins (CLDNs); in particular, CLDN-3 and -4 are frequently overexpressed in malignant case. The generation of CLDN binders is a key for expanding CLDN-targeted cancer therapy but has been delayed due to the small size of CLDN extracellular domains (approximately 50 amino acids for the first domain and 15 amino acids for the second) and their high homology among species. Interestingly, however, the receptors for Clostridium perfringens enterotoxin (CPE), a foodborne toxin in humans, happen to be identical to CLDN-3 and -4. Thus, the first CLDN binder, CPE, has provided us CLDN-targeted cancer therapy from a concept into a potential reality. In this review, we describe roles of CPE technology in cancer therapy and discuss future directions in the CLDN-targeting concept-to-therapy process.


Assuntos
Claudinas/metabolismo , Enterotoxinas/farmacologia , Enterotoxinas/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Animais , Epitélio/efeitos dos fármacos , Epitélio/metabolismo , Humanos
11.
Allergy ; 72(6): 908-917, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27861999

RESUMO

BACKGROUND: The generation of the tolerogenic dendritic cells (DC) is not fully understood yet. Forkhead box protein-3 (Foxp3) is an important molecule in the immune tolerance. This study tests a hypothesis that DCs express Foxp3, which can be upregulated by Staphylococcal enterotoxin B (SEB). METHODS: The expression of Foxp3 by DCs was evaluated by real-time RT-PCR, Western blotting, flow cytometry, and chromatin immunoprecipitation assay. RESULTS: We observed that mice treated with SEB at 0.25-0.5 µg/mouse showed high frequencies of transforming growth factor (TGF)-ß-producing CD4+ T cells and TGF-ß-producing DCs in the intestine, while the IL-4+ CD4+ T cells and TIM4+ DCs were dominated in the intestine in mice treated with SEB at 1-10 µg/mouse. Treating DCs with SEB in the culture induced high levels of Foxp3 at the TGF-ß promoter locus. The function of Foxp3 was blocked by STAT6 (signal transducer and activator transcription-6); the latter was induced by exposing DCs to SEB in the culture at doses of 100-400 ng/ml. Treating allergic mice with specific immunotherapy (SIT) together with SEB significantly promoted the therapeutic effects on the allergic responses than treating with SIT alone. CONCLUSION: Dendritic cells have the capacity to express Foxp3, which can be upregulated by exposure to SEB.


Assuntos
Células Dendríticas/imunologia , Fatores de Transcrição Forkhead/metabolismo , Hipersensibilidade/terapia , Tolerância Imunológica , Animais , Células Dendríticas/metabolismo , Enterotoxinas/farmacologia , Enterotoxinas/uso terapêutico , Fatores de Transcrição Forkhead/biossíntese , Imunoterapia , Interleucina-4/metabolismo , Camundongos , Fator de Transcrição STAT6/imunologia , Fator de Crescimento Transformador beta/metabolismo , Regulação para Cima
12.
Zhongguo Gu Shang ; 30(6): 538-544, 2017 Jun 25.
Artigo em Chinês | MEDLINE | ID: mdl-29424175

RESUMO

OBJECTIVE: To explore highly agglutinative staphylococcin (HAS) pomoting on tendon-bone healing after reconstruction of anterior cruciate ligament(ACL) in rabbits. METHODS: Animal model of ACL reconstruction in 24 mature New Zealand white rabbits(3 months, 2.56 kg on average, either gender) were established using autologous digital long extensor tendon and randomly classified into 2 groups(HAS and control group), 12 rabbits for each group. HAS group was separately injected 0.1ml highly agglutinative staphylococcin immediately into tendon-bone interface during the operation and 2 days after operation. Control group was injected with the same dose of physiological saline for 3 days. All animals were sacrificed at 4, 8, and 12 weeks after operation for histological examinations. The specimens were stained with hematoxylin-eosin, picric acid-sirius red, VEGF immunohistochemistry stain, and toluidine blue to histologically analysis the total pathology changes of the tendon-bone healing tissue, the tendon bone interface morphology classification, hyperplasia and arrangement of collagen fiber, vascularization and new bone formation, respectively. RESULTS: The Yamakado morphological interface results showed that the tissue healing at tendon-bone interface of the HAS group was better than that of the control group. The histological examination revealed that on the 4th week after operation, the tendon-bone interface of HAS group was filled with fibrous connective tissue. The proliferated fibroblasts, chondroblasts and the angiogenesis were rich. On the 8th week after operation, the healing tissue at the bone-tendon interface had developed into dense connective tissue, the neo-vessels were very rich, the collagen fibers were formed abundantly, some Sharpey's fibers spanning parts of the tendon-bone interface. On the 12th week after operation, the transition zones were full of Sharpey's fibers;the neo-vessels were not as much as the 8th weeks, but new bone formation was further increased and immature fibrocartilage appeared. For quantitative histological analysis at 4, 8 and 12 weeks after operation, the proportion of neo-vessel area and the area of now bone formation of the HAS group were all significantly higher than those of the control group(P<0.05). CONCLUSIONS: Under the synergy of staphylococcal enterotoxin C and other active ingredients, Highly Agglutinative Staphylococcin can significantly improve the tendon-bone healing after ACL reconstruction in rabbit knees, which is expected to be a new method to improve the clinical results of ACL reconstruction.


Assuntos
Reconstrução do Ligamento Cruzado Anterior , Bacteriocinas/uso terapêutico , Cicatrização/efeitos dos fármacos , Animais , Ligamento Cruzado Anterior , Osso e Ossos/efeitos dos fármacos , Enterotoxinas/uso terapêutico , Feminino , Masculino , Coelhos , Distribuição Aleatória , Tendões/efeitos dos fármacos
13.
J Infect Dis ; 214(1): 122-9, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-26908749

RESUMO

BACKGROUND: Chronic inflammation and composition of the colon microbiota have been associated with colorectal cancer in humans. The human commensal enterotoxigenic Bacteroides fragilis (ETBF) is linked to both inflammatory bowel disease and colorectal cancer and, in our murine model, causes interleukin 17A (IL-17A)-dependent colon tumors. In these studies, we hypothesized that persistent colonization by ETBF is required for tumorigenesis. METHODS: We established a method for clearing ETBF in mice, using the antibiotic cefoxitin. Multiple intestinal neoplasia mice were colonized with ETBF for the experiment duration or were cleared of infection after 5 or 14 days. Gross tumors and/or microadenomas were then evaluated. In parallel, IL-17A expression was evaluated in wild-type littermates. RESULTS: Cefoxitin treatment resulted in complete and durable clearance of ETBF colonization. We observed a stepwise increase in median colon tumor numbers as the duration of ETBF colonization increased before cefoxitin treatment. ETBF eradication also significantly decreased mucosal IL-17A expression. CONCLUSIONS: The timing of ETBF clearance profoundly influences colon adenoma formation, defining a period during which the colon is susceptible to IL-17A-dependent tumorigenesis in this murine model. This model system can be used to study the microbiota-dependent and molecular mechanisms contributing to IL-17A-dependent colon tumor initiation.


Assuntos
Carcinogênese/efeitos dos fármacos , Cefoxitina/efeitos adversos , Transformação Celular Neoplásica/efeitos dos fármacos , Neoplasias do Colo/complicações , Neoplasias do Colo/tratamento farmacológico , Enterotoxinas/efeitos adversos , Enterotoxinas/uso terapêutico , Animais , Bacteroides fragilis/química , Colo/microbiologia , Neoplasias do Colo/microbiologia , Humanos , Camundongos
14.
Clin Cancer Res ; 22(13): 3172-81, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-26851187

RESUMO

PURPOSE: To prospectively determine the efficacy of naptumomab estafenatox (Nap) + IFNα versus IFN in metastatic renal cell carcinoma (RCC). EXPERIMENTAL DESIGN: In a randomized, open-label, multicenter, phase II/III study, 513 patients with RCC received Nap (15 µg/kg i. v. in three cycles of four once-daily injections) + IFN (9 MU s.c. three times weekly), or the same regimen of IFN monotherapy. The primary endpoint was overall survival (OS). RESULTS: This phase II/III study did not meet its primary endpoint. Median OS/PFS for Nap + IFN patients was 17.1/5.8 months versus 17.5/5.8 months for the patients receiving IFN alone (P = 0.56; HR, 1.08/P = 0.41; HR, 0.92). Post hoc exploratory subgroup and trend analysis revealed that the baseline plasma concentrations of anti-SEA/E-120 (anti-Nap antibodies) for drug exposure and IL6 for immune status could be used as predictive biomarkers. A subgroup of patients (SG; n = 130) having concentrations below median of anti-SEA/E-120 and IL6 benefitted greatly from the addition of Nap. In SG, median OS/PFS for the patients treated with Nap + IFN was 63.3/13.7 months versus 31.1/5.8 months for the patients receiving IFN alone (P = 0.02; HR, 0.59/P = 0.02; HR, 0.62). Addition of Nap to IFN showed predicted and transient immune related AEs and the treatment had an acceptable safety profile. CONCLUSIONS: The study did not meet its primary endpoint. Nap + IFN has an acceptable safety profile, and results from post hoc subgroup analyses showed that the treatment might improve OS/PFS in a baseline biomarker-defined RCC patient subgroup. The results warrant further studies with Nap in this subgroup. Clin Cancer Res; 22(13); 3172-81. ©2016 AACR.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma de Células Renais/tratamento farmacológico , Enterotoxinas/uso terapêutico , Imunoconjugados/uso terapêutico , Neoplasias Renais/tratamento farmacológico , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos/sangue , Anticorpos Monoclonais/efeitos adversos , Antineoplásicos/efeitos adversos , Biomarcadores/sangue , Intervalo Livre de Doença , Enterotoxinas/efeitos adversos , Enterotoxinas/imunologia , Feminino , Humanos , Imunoconjugados/efeitos adversos , Interferon-alfa/efeitos adversos , Interferon-alfa/uso terapêutico , Interleucina-6/sangue , Masculino , Pessoa de Meia-Idade
15.
Methods Mol Biol ; 1317: 69-85, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26072402

RESUMO

Suicide gene therapy has been shown to be very efficient in tumor eradication. Numerous suicide genes were tested in vitro and in vivo demonstrating their therapeutic potential in clinical trials. Apart from this, still growing efforts are made to generate more targeted and more effective suicide gene systems for cancer gene therapy. In this regard bacterial toxins are an alternative, which add to the broad spectrum of different suicide strategies. In this context, the claudin-targeted bacterial Clostridium perfringens enterotoxin (CPE) is an attractive new type of suicide oncoleaking gene, which as pore-forming protein exerts specific and rapid toxicity towards claudin-3- and -4-overexpressing cancers. In this chapter we describe the generation and use of CPE-expressing vectors for the effective tumor cell killing as novel suicide gene approach particularly for treatment of therapy refractory tumors.


Assuntos
Clostridium perfringens/metabolismo , Enterotoxinas/genética , Enterotoxinas/uso terapêutico , Genes Transgênicos Suicidas , Terapia Genética/métodos , Western Blotting , Efeito Espectador , Linhagem Celular Tumoral , Claudinas/química , Claudinas/metabolismo , Ensaio de Imunoadsorção Enzimática , Vetores Genéticos/metabolismo , Humanos , Imuno-Histoquímica , L-Lactato Desidrogenase/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Transfecção
16.
Toxicol Mech Methods ; 25(7): 574-80, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26099602

RESUMO

A network of inflammation factors is related to pulmonary fibrosis induced by paraquat (PQ) poisoning. At high doses, the superantigen staphylococcal enterotoxin C1 (SEC1) can induce immunological unresponsiveness and inhibit release of inflammation factors. In this study, site-directed mutagenesis was performed at the H118 and H122 amino acid residues of SEC1 to reduce SEC1 toxicity. The SEC1 mutant showed significantly decreased pyrogenic toxicity, but retained clonal anergy at high dosages in vitro. Pretreatment with the SEC1 mutant prior to PQ poisoning in mice reduced symptom duration and severity, prolonged survival time, and decreased the splenocyte response to ConA induction. The SEC1 mutant also down-regulated several important cytokines related to fibrosis in the plasma after PQ poisoning. SEC1 decreased the expression of genes related to pulmonary fibrosis, including α-SMA, COL1a1, COL3 and TGF-ß1, in PQ poisoned mice. Histomorphological observation indicated alleviation of pathological changes in the lungs after SEC1 pretreatment compared to mice in the PQ group. In conclusion, the SEC1 mutant reduced pulmonary interstitial fibrosis induced by PQ poisoning.


Assuntos
Antígenos de Bactérias/uso terapêutico , Enterotoxinas/uso terapêutico , Herbicidas/toxicidade , Paraquat/toxicidade , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/prevenção & controle , Animais , Antígenos de Bactérias/genética , Antígenos de Bactérias/toxicidade , Proliferação de Células , Citocinas/metabolismo , Regulação para Baixo/efeitos dos fármacos , Enterotoxinas/genética , Enterotoxinas/toxicidade , Escherichia coli/genética , Feminino , Expressão Gênica/efeitos dos fármacos , Herbicidas/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos BALB C , Mutagênese Sítio-Dirigida , Paraquat/antagonistas & inibidores , Fibrose Pulmonar/genética , Coelhos , Baço/citologia , Baço/efeitos dos fármacos , Superantígenos , Análise de Sobrevida
17.
Toxins (Basel) ; 7(4): 1116-25, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25835384

RESUMO

Clostridium perfringens enterotoxin (CPE) is a three-domain polypeptide, which binds to Claudin-3 and Claudin-4 with high affinity. Because these receptors are highly differentially expressed in many human tumors, claudin-3 and claudin-4 may provide an efficient molecular tool to specifically identify and target biologically aggressive human cancer cells for CPE-specific binding and cytolysis. In this review we will discuss these surface proteins as targets for the detection and treatment of chemotherapy-resistant gynecologic malignancies overexpressing claudin-3 and -4 using CPE-based theranostic agents. We will also discuss the use of fluorescent c-CPE peptide in the operative setting for real time detection of micro-metastatic tumors during surgery and review the potential role of CPE in other medical applications.


Assuntos
Enterotoxinas/uso terapêutico , Neoplasias dos Genitais Femininos/diagnóstico , Neoplasias dos Genitais Femininos/tratamento farmacológico , Animais , Sítios de Ligação , Claudina-3/metabolismo , Claudina-4/metabolismo , Feminino , Neoplasias dos Genitais Femininos/metabolismo , Humanos , Estrutura Terciária de Proteína
18.
Cancer Med ; 4(3): 457-71, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25641882

RESUMO

Latent membrane protein 2A (LMP2A) is expressed on almost all Epstein-Barr virus (EBV)-associated tumors and is a potential target for immunotherapeutic intervention and vaccination. However, LMP2A is not efficiently processed and presented on major histocompatibility antigens class I molecules to generate potent cytotoxic T-lymphocytes (CTL) responses capable of killing these tumors. The B subunit of Escherichia coli enterotoxin (EtxB), causes rapid internalization and processing of membrane-bound LMP2A on EBV-infected B cells, and facilitates loading of processed-LMP2A peptides onto MHC class I. This re-directed trafficking/delivery of LMP2A to the MHC class I machinery enhances recognition and killing by LMP2A-specific CTL in vitro. To test the potential of EtxB to enhance immune targeting of LMP2A expressed in solid tumors, we generated a murine tumor model (Renca-LMP2A), in which LMP2A is expressed as a transgenic neoantigen on a renal carcinoma (Renca) cell line and forms solid tumors when injected subcutaneously into BALB/c mice. The data show that in BALB/c mice which have only low levels of peripheral K(d)-LMP2A-specific CD8(+) T cells, merely a transient inhibition of tumor growth is achieved compared with naïve mice; suggesting that there is suboptimal LMP2A-specifc CTL recognition and poorly targeted tumor killing. However, importantly, treatment of these mice with EtxB led to a significant delay in the onset of tumor growth and significantly lower tumor volumes compared with similar mice that did not receive EtxB. Moreover, this remarkable effect of EtxB was achieved despite progressive reduction in tumor expression of LMP2A and MHC class I molecules. These data clearly demonstrate the potential efficacy of EtxB as a novel therapeutic agent that could render EBV-associated tumors susceptible to immune control.


Assuntos
Antineoplásicos/uso terapêutico , Toxinas Bacterianas/uso terapêutico , Enterotoxinas/uso terapêutico , Proteínas de Escherichia coli/uso terapêutico , Neoplasias/tratamento farmacológico , Proteínas da Matriz Viral/metabolismo , Animais , Linhagem Celular Tumoral , Feminino , Imunização , Camundongos Endogâmicos BALB C , Neoplasias/imunologia , Neoplasias/metabolismo , Linfócitos T Citotóxicos/imunologia , Vaccinia virus/genética , Vaccinia virus/imunologia , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/imunologia
19.
Oncotarget ; 6(6): 4428-39, 2015 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-25669986

RESUMO

Naptumomab estafenatox/ABR-217620/ANYARA (Nap) has been evaluated in clinical phase 1 and 2/3 studies. RCC patients in the phase 2/3 trial were randomized 1:1 in an open label study to receive Nap+IFN-α or IFN-α. In this study, we analyzed the UK patients for their immunological response in relation to prolonged overall survival (OS). We found that Nap-specific T cells were reduced after 3 treatment days in patients' peripheral blood. Levels of both Nap-specific CD4+ and CD8+ T cells were significantly higher 8 days after the first treatment. Patients with such pattern of reduction and expansion of Nap-binding T cells also showed increased levels of IL-2 and IFN-γ in plasma 3 hours after the first Nap treatment. In addition, Nap caused an increase of IL-6, IL-10 and TNF-α. The patients in the UK subset showed a tendency of OS benefit after Nap treatment. Most Nap treated patients with long OS had low baseline IL-6 and normal levels of anti-SEA/E-120 antibodies. Furthermore, patients with pronounced Nap induced IL-2 and T cell expansion had long OS. In conclusion, patients with low baseline IL-6 and normal anti-SEA/E-120 may respond well to Nap by T cell activation and expansion paving the way for anti-tumour effects.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Carcinoma de Células Renais/tratamento farmacológico , Enterotoxinas/uso terapêutico , Imunoconjugados/uso terapêutico , Neoplasias Renais/tratamento farmacológico , Subpopulações de Linfócitos T/efeitos dos fármacos , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma de Células Renais/imunologia , Carcinoma de Células Renais/mortalidade , Feminino , Citometria de Fluxo , Humanos , Interferon-alfa/uso terapêutico , Interleucina-6/sangue , Estimativa de Kaplan-Meier , Neoplasias Renais/imunologia , Neoplasias Renais/mortalidade , Masculino , Pessoa de Meia-Idade , Linfócitos T/efeitos dos fármacos , Adulto Jovem
20.
Respirology ; 19(7): 1025-33, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25123375

RESUMO

BACKGROUND AND OBJECTIVE: Mesothelioma is an incurable cancer with a rising global incidence. Intrapleural delivery of a commercially available compound made up of proteins produced by Staphylococcus aureus has been used clinically to induce pleurodesis. We investigate if this bacterial compound has anti-tumoural activities against pleural malignancies, in addition to its pleurodesing effect. METHODS: The effects of the treatment on mesothelioma cells were evaluated in vitro and further tested in two validated murine models. RESULTS: This S. aureus bio-product mixture effectively kills mesothelioma cells and induces the release of interleukin (IL)-8, monocyte chemotactic protein (MCP)-1 and vascular endothelial growth factor from primary human mesothelial cells but not malignant pleural mesothelioma cells in vitro. Intratumoural delivery of the treatment in BALB/c mice induced tumour necrosis and local activation of T cells. Tumour growth was significantly inhibited in the treatment group during and after the treatment period (size of tumour 58.8 ± 10.3 mm(2) vs 118.3 ± 6.7 mm(2) from saline controls at day 23, n = 9-12 per group), P < 0.001. Tumour growth resumed on cessation of treatment, confirming the inhibition was treatment related. Treatment benefits were further validated in an orthotopic peritoneal model of mesothelioma and the compound significantly reduced the mesothelioma load (P < 0.05 vs saline controls). Mice in the treatment group had a significant increase in the percentage of activated CD4(+) and CD8(+) T cells in tumour-draining lymph nodes. No histological side-effects were observed with the treatment. CONCLUSIONS: This proof-of-principle study demonstrates promising antitumoural activity of a commercially available compound of S. aureus bio-products against mesothelioma.


Assuntos
Antígenos de Bactérias/uso terapêutico , Enterotoxinas/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Mesotelioma/tratamento farmacológico , Mesotelioma/patologia , Staphylococcus aureus/imunologia , Animais , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Modelos Animais de Doenças , Humanos , Mesotelioma Maligno , Camundongos , Camundongos Endogâmicos BALB C
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...