Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Cell Immunol ; 348: 104041, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31983398

RESUMO

Chimeric antigen receptor (CAR)-modified T cell therapy evokes only modest antitumor responses in solid tumors. Meso-CAR-T cells are CAR-T cells targeted mesothelin, which are over-expressed in tumor tissues of breast cancer patients. To improve the therapeutic effects, we combined it with rAd.sT, a transforming growth factor ß signaling-targeted oncolytic adenovirus, to therapy breast cancer. In subcutaneous MDA-MB-231 xenograft of NSG mice, both rAd.sT and meso-CAR-T inhibited tumor growth, however combination therapy produced stronger inhibitory effects. Interestingly, rAd.sT reduced tumor burden at initial stage following vector treatments, while meso-CAR-T cells decreased tumor burden at a later stage. Moreover, meso-CAR-T could target tumor microenvironments, and combination therapy could enhance cytokines production, such as interleukin (IL)-6 and IL-12 in tumor microenvironment. In conclusion, combination of rAd.sT with meso-CAR-T produced much more impressive antitumor responses to breast cancer and its metastasis, which could be developed as a promising therapeutic strategy.


Assuntos
Neoplasias da Mama , Terapia Combinada/métodos , Imunoterapia Adotiva/métodos , Terapia Viral Oncolítica/métodos , Adenoviridae , Animais , Antineoplásicos/farmacologia , Feminino , Proteínas Ligadas por GPI/antagonistas & inibidores , Humanos , Mesotelina , Camundongos , Vírus Oncolíticos , Receptores de Antígenos Quiméricos , Fator de Crescimento Transformador beta/antagonistas & inibidores , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Int J Mol Sci ; 18(4)2017 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-28417919

RESUMO

In advanced prostate cancer, small ubiquitin-like modifier (SUMO)-specific cysteine protease 1 (SENP1) is up-regulated. However, the role of SENP1 in regulating deSUMOylation of TGF-ß/SMADs signaling is unknown. In this study, we developed a lentiviral vector, PLKO.1-shSENP1, to silence SENP1 in prostate cancer cells with high metastatic characteristics (PC3M). Likewise, we also created an adenovirus vector, Ad5/F11p-SENP1 to over-express SENP1 in prostate cancer cells with low metastatic potential (LNCaP). We showed that silencing of SENP1 promoted cellular apoptosis, and inhibited proliferation and migration of PC3M cells. Moreover, SENP1 silencing increased the SMAD4 expression at protein level, up-regulated E-cadherin and down-regulated Vimentin expression, indicating the inhibition of epithelial mesenchymal transition (EMT). Furthermore, SMAD4 interference abolished SENP1-mediated up-regulation of E-cadherin, suggesting that SENP1 regulated E-cadherin expression via SMAD4. SENP1 over-expression in LNCaP cells reduced SMAD4 protein, and promoted EMT via decreasing E-cadherin and increasing Vimentin. Moreover, down-regulation of SMAD4 and E-cadherin were blocked, after transfection with two SUMOylation sites mutated SMAD4, suggesting that SENP1 might reduce SMAD4 levels to regulate E-cadherin expression via deSUMOylation of SMAD4. In conclusion, SENP1 deSUMOylated SMAD4 to promote EMT via up-regulating E-cadherin in prostate cancer cells. Therefore, SENP1 is a potential target for treatment of advanced prostate cancer.


Assuntos
Endopeptidases/metabolismo , Transição Epitelial-Mesenquimal , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Proteína Smad4/metabolismo , Androgênios , Apoptose/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Cisteína Endopeptidases , Transição Epitelial-Mesenquimal/genética , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Humanos , Masculino , Neoplasias da Próstata/genética , RNA Interferente Pequeno/genética , Transdução de Sinais , Sumoilação , Fator de Crescimento Transformador beta/metabolismo
3.
Mol Ther ; 22(8): 1504-1517, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24791939

RESUMO

We are interested in developing oncolytic adenoviruses for the treatment of prostate cancer (PCa) bone metastases. A key limitation of Adenovirus 5 (Ad5) is that upon systemic administration, it produces major liver and systemic toxicities. To address this issue, a chimaeric Ad5/48 adenovirus mHAd.sTßRFc was created. Seven hypervariable regions of Ad5 hexon present in Ad5-based Ad.sTßRFc expressing soluble transforming growth factor beta receptor II-Fc fusion protein (sTGßRIIFc), were replaced by those of Ad48. mHAd.sTßRFc, like Ad.sTßRFc, was replication competent in the human PCa cells, and produced high levels of sTGßRIIFc expression. Compared to Ad.sTßRFc, the systemic delivery of mHAd.sTßRFc in nude mice resulted in much reduced systemic toxicity, and reduced liver sequestration. Ad.sTßRFc produced significant liver necrosis, and increases in alanine transaminase, aspartate transaminase, lactate dehydrogenase, tumor necrosis factor-α, and interleukin-6 levels, while mHAd.sTßRFc produced much reduced responses of these markers. Intravenous delivery of Ad.sTßRFc or mHAd.sTßRFc (5 × 10(10) viral particles/mouse) in nude mice bearing PC-3-luc PCa bone metastases produced inhibition of bone metastases. Moreover, a larger dose of the mHAd.sTßRFc (4 × 10(11) viral particles /mouse) was also effective in inhibiting bone metastases. Thus, mHAd.sTßRFc could be developed for the treatment of PCa bone metastases.


Assuntos
Neoplasias Ósseas/terapia , Proteínas do Capsídeo/genética , Vetores Genéticos/efeitos adversos , Vírus Oncolíticos/genética , Neoplasias da Próstata/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Animais , Neoplasias Ósseas/secundário , Linhagem Celular Tumoral , Dependovirus/classificação , Dependovirus/genética , Vetores Genéticos/administração & dosagem , Vetores Genéticos/uso terapêutico , Humanos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Terapia Viral Oncolítica , Vírus Oncolíticos/classificação , Neoplasias da Próstata/terapia , Proteínas Serina-Treonina Quinases/genética , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
4.
Mol Ther ; 19(9): 1609-18, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21712815

RESUMO

In recent years, oncolytic adenoviruses have shown some promise as a novel class of antitumor agents. However, their utility in targeting bone metastases is relatively less studied. We have examined whether the systemic therapy of oncolytic adenoviruses expressing the soluble form of transforming growth factor-ß (TGFß) receptor II fused with human immunoglobulin G1 can be developed for the treatment of established breast cancer bone metastases. MDA-MB-231-luc2 human breast cancer cells were injected in the left heart ventricle of nude mice to establish bone metastasis. Mice with hind limb tumors were administered (on days 8 and 11) oncolytic adenoviruses-Ad.sTßRFc or mhTERTAd.sTßRFc. Skeletal tumor growth was monitored weekly by bioluminescence imaging (BLI) and radiography. At the termination time on day 28, hind limb bones were analyzed for tumor burden, synchrotron micro-computed tomography, and osteoclast activation. Intravenous delivery of Ad.sTßRFc and mhTERTAd.sTßRFc induced significant inhibition of tumor growth, reduction of tumor burden, osteoclast activation, and increased animals' survival. Oncolytic adenoviruses were safer than dl309, a wild-type virus. A slight elevation of liver enzyme activity was observed after Ad.sTßRFc administration; this subsided with time. Based on these studies, we believe that Ad.sTßRFc and mhTERTAd.sTßRFc can be developed as a safe and effective approach for the treatment of established bone metastasis.


Assuntos
Adenoviridae/genética , Neoplasias Ósseas/secundário , Neoplasias da Mama/terapia , Terapia Viral Oncolítica/métodos , Proteínas Serina-Treonina Quinases/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Fosfatase Ácida/sangue , Animais , Neoplasias Ósseas/patologia , Neoplasias Ósseas/terapia , Feminino , Terapia Genética/métodos , Células HEK293 , Humanos , Injeções Intravenosas , Isoenzimas/sangue , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Vírus Oncolíticos/genética , Osteoclastos/patologia , Radiografia , Receptor do Fator de Crescimento Transformador beta Tipo II , Síncrotrons/instrumentação , Fosfatase Ácida Resistente a Tartarato , Carga Tumoral , Replicação Viral , Redução de Peso , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
5.
RSC Adv ; 10(61): 37327-37345, 2020 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-35521263

RESUMO

The demand for lightweight, high specific strength alloys has drastically increased in the last two decades. Magnesium and aluminum alloys are very suitable candidate materials. Research on alloys with an Mg2Si phase started about three decades ago. The current scenario is that magnesium and aluminum alloys containing an Mg2Si phase are very popular in the scientific community and extensively used in the automotive and aerospace industries. Mg2Si is a very stable phase and exhibits excellent mechanical, thermal, electrochemical and tribological properties. This paper presents a brief review of Mg-Si binary alloys, and Mg-Si-Al and Al-Si-Mg alloys. Grain refinement methods and mechanical properties have been reported on lightweight alloys containing Mg and Si. The available results show that silicon reacts with magnesium and forms an intermetallic compound with the stoichiometric formula Mg2Si. There is in situ formation of an Mg2Si phase in Mg-Si, Mg-Si-Al and Al-Mg-Si alloys by the diffusion or precipitation process. The morphology and size of an in situ developed Mg2Si phase depend on the synthesis route and base metal or matrix. In the liquid metallurgy process the precipitation sequence depends on the cooling rate. The morphology of the Mg2Si phase depends on the precipitation sequence and the mechanical properties depend on the morphology and size of the Mg2Si phase within the alloy matrix.

6.
Hum Gene Ther ; 31(15-16): 863-880, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32394753

RESUMO

We report here the development of oncolytic adenoviruses (Ads) that have reduced toxicity, enhanced tumor tropism, produce strong antitumor response, and can overcome resistance to immune checkpoint inhibitor therapy in breast cancer. We have shown that LyP-1 receptor (p32) is highly expressed on the surface of breast cancer cells and tumors from cancer patients, and that increased stromal expression of transforming growth factor ß-1 (TGFß-1) is associated with triple-negative breast cancer. Therefore, we constructed oncolytic Ads, AdLyp.sT and mHAdLyp.sT, in which the p32-binding LyP-1 peptide was genetically inserted into the adenoviral fiber protein. Both AdLyp.sT and mHAdLyp.sT express sTGFßRIIFc, a TGFß decoy that can inhibit TGFß pathways. mHAdLyp.sT is an Ad5/48 chimeric hexon virus in which hypervariable regions (HVRs 1-7) of Ad5 are replaced with the corresponding Ad48 HVRs. AdLyp.sT and mHAdLyp.sT exhibited better binding, replication, and produced higher sTGFßRIIFc protein levels in breast cancer cell lines compared with Ad.sT or mHAd.sT control viruses without LyP-1 peptide modification. Systemic delivery of mHAdLyp.sT in mice resulted in reduced hepatic/systemic toxicity compared with Ad.sT and AdLyp.sT. Intravenous delivery of AdLyp.sT and mHAdLyp.sT elicited a strong antitumor response in a human MDA-MB-231 bone metastasis model in mice, as indicated by bioluminescence imaging, radiographic tumor burden, serum TRACP 5b and calcium, and body weight analyses. Furthermore, intratumoral delivery of AdLyp.sT in 4T1 model in immunocompetent mice inhibited tumor growth and metastases, and augmented anti-PD-1 and anti-CTLA-4 therapy. Based on these studies, we believe that AdLyp.sT and mHAdLyp.sT can be developed as potential targeted immunotherapy agents for the treatment of breast cancer.


Assuntos
Adenoviridae/genética , Neoplasias Ósseas/terapia , Neoplasias da Mama/terapia , Inibidores de Checkpoint Imunológico/farmacologia , Terapia Viral Oncolítica/métodos , Proteína Tirosina Fosfatase não Receptora Tipo 22/metabolismo , Fator de Crescimento Transformador beta/antagonistas & inibidores , Animais , Neoplasias Ósseas/genética , Neoplasias Ósseas/secundário , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Terapia Combinada , Feminino , Vetores Genéticos/administração & dosagem , Humanos , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Proteína Tirosina Fosfatase não Receptora Tipo 22/genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Cancer Gene Ther ; 27(12): 923-933, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32307442

RESUMO

LIGHT, also known as tumor-necrosis factor (TNF) superfamily member 14 (TNFSF14), is predominantly expressed on activated immune cells and some tumor cells. LIGHT is a pivotal regulator both for recruiting and activating immune cells in the tumor lesions. In this study, we armed human telomerase reverse transcriptase (TERT) promoter controlled oncolytic adenovirus with LIGHT to generate rAd.Light. rAd.Light effectively transduced both human and mouse breast tumor cell lines in vitro, and expressed LIGHT protein on the surface of tumor cells. Both rAd.Null, and rAd.Light could replicate in human breast cancer cells, and produced cytotoxicity to human and mouse mammary tumor cells. rAd.Light induced apoptosis resulting in tumor cell death. Using a subcutaneous model of 4T1 cells in BALB/c mice, rAd.Light was delivered intratumorally to evaluate the anti-tumor responses. Both rAd.Light and rAd.Null significantly inhibited the tumor growth, but rAd.Light produced much stronger anti-tumor effects. Histopathological analysis showed the infiltration of T lymphocytes in the tumor tissues. rAd.Light also induced stronger cellular apoptosis than rAd.Null in the tumors. Interestingly, on day 15, compared to rAd.Null, there was a significant reduction of Tregs following rAd.Light treatment. rAd.Light significantly increased Th1 cytokine interleukin (IL)-2 expression, and reduced Th2 cytokines expression, such as transforming growth factor ß (TGF-ß) and IL-10 in the tumors. These results suggest rAd.Light induced activation of anti-tumor immune responses. In conclusion, rAd.Light produced anti-tumor effect in a subcutaneous model of breast cancer via inducing tumor apoptosis and evoking strong anti-tumor immune responses. Therefore, rAd.Light has great promise to be developed as an effective therapeutic approach for the treatment of breast cancer.


Assuntos
Neoplasias Mamárias Animais/genética , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Animais , Linhagem Celular Tumoral , Proliferação de Células , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Endogâmicos BALB C
8.
Hum Gene Ther ; 30(2): 197-210, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30032645

RESUMO

The majority of advanced breast cancer patients develop distal metastasis, including lung and bone metastasis. However, effective therapeutic strategies to prevent metastasis are still lacking. Decorin is a natural inhibitor of transforming growth factor ß, which plays a pivotal role in tumor metastasis. An oncolytic adenovirus expressing decorin, rAd.DCN, has been developed previously. In an immune-competent breast tumor (4T1) model, intratumoral (i.t.) as well as intravenous (i.v.) delivery of rAd.DCN inhibited growth of orthotopic tumors and spontaneous lung metastasis. It was shown that i.t. delivery of rAd.DCN produced higher levels of transgene expression and evoked stronger oncolysis of the tumors compared to i.v. delivery. However, i.v. delivery resulted in higher amount of virus accumulation in the lungs and produced stronger responses to prevent tumor lung metastasis. Oncolytic adenovirus-mediated decorin expression in the tumors downregulated the decorin target genes and decreased epithelial mesenchymal transition markers. Decorin expression in lung tissues also increased Th1 cytokine expression, such as interleukin (IL)-2, IL-12, and tumor necrosis factor α, and decreased Th2 cytokines, such as transforming growth factor ß and IL-6. Moreover, rAd.DCN treatment induced strong systemic inflammatory responses and upregulated CD8+ T lymphocytes. In conclusion, rAd.DCN inhibits tumor growth and lung metastasis of breast cancer via regulating wnt/ß-catenin, vascular endothelial growth factor (VEGF), and Met pathways, and modulating the antitumor inflammatory and immune responses. Considering that i.v. delivery was much more effective in preventing lung metastasis, systemic delivery of rAd.DCN might be a promising strategy to treat breast cancer lung metastasis.


Assuntos
Adenoviridae , Neoplasias da Mama , Decorina , Neoplasias Pulmonares , Terapia Viral Oncolítica , Vírus Oncolíticos , Adenoviridae/genética , Adenoviridae/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Decorina/biossíntese , Decorina/genética , Feminino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/terapia , Camundongos , Metástase Neoplásica , Vírus Oncolíticos/genética , Vírus Oncolíticos/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Hum Gene Ther ; 30(9): 1117-1132, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31126191

RESUMO

In an effort to develop a new therapy for cancer and to improve antiprogrammed death inhibitor-1 (anti-PD-1) and anticytotoxic T lymphocyte-associated protein (anti-CTLA-4) responses, we have created a telomerase reverse transcriptase promoter-regulated oncolytic adenovirus rAd.sT containing a soluble transforming growth factor receptor II fused with human IgG Fc fragment (sTGFßRIIFc) gene. Infection of breast and renal tumor cells with rAd.sT produced sTGFßRIIFc protein with dose-dependent cytotoxicity. In immunocompetent mouse 4T1 breast tumor model, intratumoral delivery of rAd.sT inhibited both tumor growth and lung metastases. rAd.sT downregulated the expression of several transforming growth factor ß (TGFß) target genes involved in tumor growth and metastases, inhibited Th2 cytokine expression, and induced Th1 cytokines and chemokines, and granzyme B and perforin expression. rAd.sT treatment also increased the percentage of CD8+ T lymphocytes, promoted the generation of CD4+ T memory cells, reduced regulatory T lymphocytes (Tregs), and reduced bone marrow-derived suppressor cells. Importantly, rAd.sT treatment increased the percentage of CD4+ T lymphocytes, and promoted differentiation and maturation of antigen-presenting dendritic cells in the spleen. In the immunocompetent mouse Renca renal tumor model, similar therapeutic effects and immune activation results were observed. In the 4T1 mammary tumor model, rAd.sT improved the inhibition of tumor growth and lung and liver metastases by anti-PD-1 and anti-CTLA-4 antibodies. Analysis of the human breast and kidney tumors showed that a significant number of tumor tissues expressed high levels of TGFß and TGFß-inducible genes. Therefore, rAd.sT could be a potential enhancer of anti-PD-1 and anti-CTLA-4 therapy for treating breast and kidney cancers.


Assuntos
Adenoviridae/genética , Vetores Genéticos/genética , Imunidade , Terapia Viral Oncolítica , Vírus Oncolíticos/genética , Fator de Crescimento Transformador beta/genética , Animais , Antineoplásicos Imunológicos/farmacologia , Antígeno CTLA-4/antagonistas & inibidores , Linhagem Celular Tumoral , Terapia Combinada , Citocinas/metabolismo , Modelos Animais de Doenças , Técnicas de Transferência de Genes , Humanos , Imunomodulação , Camundongos , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/terapia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Transdução de Sinais , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Transdução Genética , Fator de Crescimento Transformador beta/metabolismo , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Int J Radiat Oncol Biol Phys ; 101(4): 945-956, 2018 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-29976507

RESUMO

PURPOSE: To evaluate the therapeutic effects of decorin (DCN)-modified mesenchymal stem cells (MSCs) on radiation-induced lung injuries (RILIs) and to clarify the underlying mechanisms. METHODS AND MATERIALS: Umbilical cord-derived mesenchymal stem cells (MSCs) were modified with Ad(E1-).DCN to generate DCN-expressing MSCs (DCN-modified MSCs [MSCs.DCN]). In an experimental mouse model of RILI, MSCs.DCN and MSCs.Null [MSCs modified with Ad(E1-).Null] were intravenously engrafted at 6 hours or 28 days after irradiation. The therapeutic effects on lung inflammation and fibrosis were evaluated by histopathologic analysis at 28 days and 3 months after irradiation. Inflammatory cytokines and chemokines were analyzed in both sera and lung tissues, and subtypes of T lymphocytes including regulatory T cells (Tregs) were analyzed in the peripheral blood and spleen. RESULTS: Both MSC treatments could alleviate histopathologic injuries by reducing lymphocyte infiltration, decreasing apoptosis, increasing proliferation of epithelial cells, and inhibiting fibrosis in the later phase. However, treatment with MSCs.DCN resulted in much more impressive therapeutic effects. Moreover, we discovered that MSC treatment reduced the expression of chemokines and inflammatory cytokines and increased the expression of anti-inflammatory cytokines in both the peripheral blood and local pulmonary tissues. An important finding was that MSCs.DCN were much more effective in inducing interferon-γ expression, inhibiting collagen type III α1 expression in pulmonary tissues, and decreasing the proportion of Tregs. Furthermore, our data suggested that treatment during the acute phase (6 hours) after irradiation evoked much stronger responses both in attenuating inflammation and in inhibiting fibrosis than in the later phase (28 days). CONCLUSIONS: MSCs.DCN could attenuate acute inflammation after irradiation and significantly inhibit later fibrosis. Likewise, DCN enhanced the functions of MSCs by targeting profibrotic factors and Tregs.


Assuntos
Quimiocinas/metabolismo , Decorina/metabolismo , Decorina/farmacologia , Pulmão/efeitos da radiação , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/efeitos dos fármacos , Fibrose Pulmonar/prevenção & controle , Lesões Experimentais por Radiação/terapia , Cordão Umbilical/citologia , Adenoviridae , Animais , Apoptose , Proliferação de Células , Colágeno/análise , Colágeno Tipo I/metabolismo , Colágeno Tipo III/metabolismo , Citocinas/metabolismo , Progressão da Doença , Regulação para Baixo , Raios gama , Vetores Genéticos , Humanos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Interferon gama/metabolismo , Pulmão/química , Pulmão/metabolismo , Pulmão/patologia , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Pneumonia/prevenção & controle , Lesões Experimentais por Radiação/metabolismo , Linfócitos T Reguladores/citologia , Fatores de Tempo , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Fator de Necrose Tumoral alfa/administração & dosagem , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
11.
Methods Mol Med ; 130: 135-44, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17401169

RESUMO

This chapter describes several methods for recognizing apoptosis in tumor cells following infection with a replication-deficient adenovirus expressing the tumor suppressor gene p53. We include cytotoxicity assays and assays of apoptosis, including DNA-nucleosomal DNA fragmentation (DNA laddering), TUNEL, DAPI staining, analysis of the sub-G1 (subdiploid) population, and degradation of poly(ADP-ribose) polymerase (as assayed by Western blot). Although this is not a comprehensive list of protocols to evaluate apoptosis, we believe that these will cover the majority of conditions of apoptosis that may arise. The chapter also describes the characteristics of each technique, including the advantages and disadvantages of each method.


Assuntos
Adenoviridae/genética , Apoptose/fisiologia , Genes p53 , Linhagem Celular Tumoral , Fragmentação do DNA , DNA Viral , Humanos , Marcação In Situ das Extremidades Cortadas , Poli(ADP-Ribose) Polimerases/metabolismo , Células Tumorais Cultivadas
12.
Mol Cancer Ther ; 5(2): 367-73, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16505111

RESUMO

In recent years, adenoviruses that selectively replicate in tumor cells have been developed. However, there is a tremendous need to improve their anticancer efficacy. We wish to investigate whether a strategy that combines the oncolytic effects of an adenoviral vector with simultaneous expression of soluble form of transforming growth factor-beta type II receptor (sTGFbetaRII) offers a therapeutic advantage. We chose to target TGF-betas because they play a pivotal role in late-stage tumorigenesis by enhancing tumor invasion and metastasis. A sTGFbetaRII cDNA was cloned in conditionally replicating adenoviral vector rAd-sTRII and in a replication-deficient adenovirus Ad-sTRII. Infection of MDA-MB-231 breast cancer cells with rAd-sTRII or Ad-sTRII followed by Western blot analysis indicated the expression of diffused glycosylated forms of sTGFbetaRII that were also secreted into the extracellular medium. The secreted proteins were shown to bind with TGF-beta and antagonize TGF-beta-induced p38 mitogen-activated protein kinase activity. However, marked differences in the replication potential of rAd-sTRII and Ad-sTRII were observed in breast tumor cells. Infection of MDA-MB-231 cells with rAd-sTRII resulted in cytotoxicity and significant increase in the adenoviral titers that were comparable with a wild-type adenovirus dl309. However, Ad-sTRII was much less toxic to the tumor cells, and the viral titers of Ad-sTRII remained relatively unchanged. These results suggest that the infection of breast tumor cells with conditionally replicating adenoviral vector rAd-sTRII produced sTGFbetaRII that can abrogate TGF-beta signaling while maintaining the replication potential of the virus, indicating that rAd-sTRII could be a potential anticancer agent.


Assuntos
Adenoviridae/fisiologia , Neoplasias da Mama/terapia , Terapia Viral Oncolítica , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Fator de Crescimento Transformador beta/antagonistas & inibidores , Adenoviridae/genética , Humanos , Fosforilação , Proteínas Serina-Treonina Quinases , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/genética , Células Tumorais Cultivadas , Replicação Viral , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
13.
Hum Gene Ther ; 28(8): 667-680, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28530155

RESUMO

In advanced and metastatic stages of colorectal cancer (CRC), reduced sensitivity to conventional strategies is still a major obstacle to successful treatments. Decorin is an important regulator in the development and progression of various cancers. To examine if CRC patients have altered decorin levels, expression of decorin and its target genes, Met and vascular endothelial growth factor A (VEGFA), were analyzed in their tumors. Compared to normal tissues, decorin expression was reduced in CRC patients' tumors, while there were increased Met and VEGFA levels. To develop a novel therapy for CRC, rAd.DCN.GM, an oncolytic adenovirus encoding decorin and granulocyte macrophage colony stimulating factor (GM-CSF), has been created. Several therapeutic strategies expressing GM-CSF have been employed in clinical trials for treating metastatic colorectal cancer. In this study, infection of CRC cells with rAd.DCN.GM expressed decorin and GM-CSF, and produced cytotoxicity. In murine CT26 xenografts, rAd.DCN.GM and control adenoviruses were administrated intratumorally on days 7 and 10, and tumor volumes were monitored over time. The study showed that rAd.DCN.GM inhibited the tumor growth and lung metastases significantly. rAd.DCN.GM induced apoptosis, inhibited proliferation, and downregulated angiogenesis and epithelial mesenchymal transition markers in the tumors. On day 12 and day 29, the immune-activation in the peripheral blood, tumors, and spleens were analyzed. rAd.DCN.GM increased CD8+ T lymphocytes in the blood, upregulated perforin and granzyme B in the tumors, inhibited transforming growth factor beta expression, and promoted dendritic-cell production in the spleen. In conclusion, rAd.DCN.GM inhibited the tumor growth and metastasis of CT26 tumors, downregulated multiple pro-tumorigenic pathways, and activated antitumor immune responses.


Assuntos
Adenoviridae/genética , Neoplasias Colorretais/genética , Neoplasias Colorretais/imunologia , Decorina/genética , Terapia Genética , Vetores Genéticos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Terapia Viral Oncolítica , Vírus Oncolíticos/genética , Animais , Carcinogênese/genética , Carcinogênese/metabolismo , Linhagem Celular Tumoral , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/terapia , Citocinas/metabolismo , Efeito Citopatogênico Viral , Decorina/metabolismo , Modelos Animais de Doenças , Transição Epitelial-Mesenquimal , Expressão Gênica , Ordem dos Genes , Vetores Genéticos/administração & dosagem , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Imunomodulação , Camundongos , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Transdução de Sinais , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Transdução Genética , Transgenes , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Hum Gene Ther ; 17(11): 1152-60, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17032151

RESUMO

We have developed an approach to cancer gene therapy in which the oncolytic effects of an adenoviral vector have been combined with selective expression of a soluble form of transforming growth factor (TGF)-beta receptor II fused with Fc (sTGFbetaRIIFc). We chose to use adenoviral dl01/07 mutant because it can replicate in all cancer cells regardless of their genetic defects. An oncolytic adenovirus expressing sTGFbetaRIIFc (Ad.sT- betaRFc) was constructed by homologous recombination. Infection of MDA-MB-231 and MCF-7 human breast cancer cells with Ad.sTbetaRFc produced sTGFbetaRIIFc, which was released into the media. The conditioned media containing sTGFbetaRIIFc could bind with TGF-beta 1 and inhibited TGF-beta-dependent transcription in target cells. Infection of MDA-MB-231, MCF-7, and 76NE human breast cancer cells with Ad.sTbetaRFc resulted in high levels of viral replication, comparable to that of a wild-type dl309 virus. Although some viral replication was observed in actively dividing normal human lung fibroblasts, there was no replication in nonproliferating normal cells. Direct injection of Ad.sTbetaRFc into MDA-MB-231 human breast xenograft tumors grown in nude mice resulted in a significant inhibition of tumor growth, causing tumor regression in more than 85% of the animals. These results indicate that it is possible to construct an oncolytic virus expressing sTGFbetaRIIFc in which both viral replication and transgene expression remain intact, and the recombinant adenovirus is oncolytic in a human tumor xenograft model. On the basis of these results we believe that it may be feasible to develop a cancer gene therapy approach using Ad.sTbetaRFc as an antitumor agent.


Assuntos
Adenoviridae/genética , Neoplasias da Mama/terapia , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/uso terapêutico , Vírus Oncolíticos/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/uso terapêutico , Animais , Apoptose , Linhagem Celular Tumoral , Feminino , Humanos , Fragmentos Fc das Imunoglobulinas/metabolismo , Camundongos , Camundongos Nus , Proteínas Serina-Treonina Quinases , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Replicação Viral/genética , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Oncogene ; 21(8): 1187-95, 2002 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-11850838

RESUMO

Squamous cell carcinomas (SCCs) of the head and neck are characterized by high tendency to invade locally and metastasize to lymph nodes. SCC cells express several matrix metalloproteinases (MMPs) and they often harbor mutations in p53 tumor suppressor gene. Collagenase-3 (MMP-13) is specifically expressed by tumor cells of SCCs and it apparently plays an important role in their invasion and metastasis. We used adenoviral gene delivery to examine the effect of wild-type p53 on MMP-13 expression in four head and neck SCC cell lines with mutated p53. Adenoviral delivery of p53 resulted in potent inhibition in production of proMMP-13 (by 71 to 92%) and collagenase-1 (MMP-1) (by 27 to 93%) by all cell lines in 24 h, whereas production of gelatinase-A (MMP-2) and gelatinase-B (MMP-9) was not altered. Adenoviral expression of p53 also suppressed invasion of SCC cells through Matrigel by 35%. Expression of cyclin-dependent kinase inhibitor p21(Waf1/Cip1) was induced 24 h after p53 gene delivery in all SCC cell lines, except one, which lacked detectable p21(Waf1/Cip1) expression. Number of viable cells was not altered and no apoptotic cells were seen 24 h after p53 delivery. These results show, that wild-type p53 potently inhibits expression of MMP-13 and MMP-1 by SCC cells independently of its pro-apoptotic effect. Together these results indicate, that p53 exerts a bi-phasic tumor suppressor effect on SCC cells: inhibition of cell invasion followed by induction of programmed cell death.


Assuntos
Adenoviridae/genética , Carcinoma de Células Escamosas/enzimologia , Colagenases/biossíntese , Regulação Neoplásica da Expressão Gênica , Transgenes/genética , Proteína Supressora de Tumor p53/metabolismo , Apoptose , Western Blotting , Carcinoma de Células Escamosas/genética , Proteínas de Ciclo Celular/metabolismo , Sobrevivência Celular , Colágeno/metabolismo , Colagenases/genética , Inibidor de Quinase Dependente de Ciclina p21 , Inibidor de Quinase Dependente de Ciclina p27 , Ciclinas/metabolismo , Combinação de Medicamentos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes p53/genética , Humanos , Laminina/metabolismo , Metaloproteinase 1 da Matriz/biossíntese , Metaloproteinase 1 da Matriz/genética , Metaloproteinase 13 da Matriz , Invasividade Neoplásica , Proteoglicanas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Fator de Crescimento Transformador alfa/farmacologia , Fator de Crescimento Transformador beta/farmacologia , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor/metabolismo , Regulação para Cima
16.
Cancer Biol Ther ; 4(5): 512-7, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15908802

RESUMO

In recent years there has been a dramatic increase in developing gene therapy approaches for the treatment of cancer. The two events that have permitted the formulation of concept of cancer gene therapy are the new understanding of the molecular mechanisms underlying oncogenesis, and the development of the DNA-delivery vehicles or vectors. Many approaches to cancer gene therapy have been proposed, and several viral and non-viral vectors have been utilized. The purpose of this review article is to describe the various strategies of cancer gene therapy (transfer of tumor suppressor genes, suicide genes-enzyme/pro-drug approach, inhibition of dominant oncogenes, immunomodulation approaches, expression of molecules that affect angiogenesis, tumor invasion and metastasis, chemosensitization and radiosensitization approaches, and chemoprotection of stem cells). The chapter also reviews the commonly used vectors (retroviral vectors, adenoviral vectors, adeno-associated viral vectors, pox viruses, herpes simplex viruses, HIV- vectors, non-viral vectors and targetable vectors) for cancer gene therapy. Some of the important issues in cancer gene therapy, and the potential future directions are also being discussed.


Assuntos
Terapia Genética/métodos , Vetores Genéticos , Neoplasias/terapia , Previsões , Técnicas de Transferência de Genes/tendências , Terapia Genética/tendências , Humanos , Neoplasias/genética
17.
Cancer Biol Ther ; 4(3): 261-6, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15846079

RESUMO

Transforming growth factor (TGF) betas are multifunctional polypeptides that regulate several cellular functions, including cell growth and differentiation, extra cellular matrix production, motility and immunosuppression. The growth-inhibiting properties of TGFbeta have gained much attention into its role as a tumor suppressor. There is, however, now increasing evidence that TGFbeta switches roles, from tumor suppressor to tumor promoter, as the tumor progresses. Given the integral role of TGFbeta in the tumor progression, it follows that TGFbeta signaling offers an attractive target for cancer therapy. Several strategies including the use of antisense oligonucleotides for TGFbeta, TGFbeta antibodies, dominant negative TGFbeta receptor II, and small drug-molecules to inhibit TGFbeta receptor I kinase have shown great promise in the preclinical studies. These new findings, coupled with progressing clinical trials indicate that inhibition of TGFbeta signaling may, indeed, be a viable option to cancer therapy. This review summarizes the TGFbeta signaling, the dual role of TGFbeta--as a tumor suppressor and tumor promoter, and various strategies targeted against TGFbeta signaling for cancer therapy. The next few years promise to better our understanding of approaching cancer therapy with an eye to the inhibition of TGFbeta signaling.


Assuntos
Neoplasias/terapia , Fator de Crescimento Transformador beta/antagonistas & inibidores , Proteínas Supressoras de Tumor/antagonistas & inibidores , Animais , Humanos , Camundongos , Neoplasias/tratamento farmacológico , Transdução de Sinais , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
18.
Clin Cancer Res ; 10(20): 6969-76, 2004 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-15501976

RESUMO

Design and development of new approaches for targeted radiotherapy of cancer and improvement of therapeutic index by more local radiation therapy are very important issues. Adenovirus-mediated delivery of the sodium iodide symporter (NIS) gene to cancer cells is a powerful technique to concentrate lethal radiation in tumor cells and eradicate tumors with increased therapeutic index. A replication-defective adenoviral vector expressing the rat NIS gene (Ad-rNIS) was used for in vitro gene delivery and into human prostate cancer xenografts to study antitumor effect. Robust function of the rat symporter was detected in DU145, T47D, and HCT-15 human cancer cell lines transduced with Ad-rNIS. All three cancer cell lines successfully transferred functionally active rat symporter to the plasma membrane, resulting in very high levels of iodine-125 accumulation. Three-dimensional multicellular tumor spheroids derived from DU145 human prostate cancer cells were transduced with Ad-rNIS and incubated with (131)I for 24 hours. After treatment, spheroids rapidly decreased in size and disappeared within 10 days. In vivo data revealed an inhibition of tumor growth in athymic nude mice after intratumoral Ad-rNIS injection followed by (131)I administration. Eighty-eight percent of experimental mice survived >30 days, whereas control groups had only 18% survival >30 days. This is the first report that demonstrates the rat NIS gene can effectively induce growth arrest of human tumor xenografts after in vivo adenoviral gene delivery and (131)I administration. The data confirm our hypothesis that the rat NIS gene is an attractive suicide gene candidate for cancer treatment.


Assuntos
Terapia Genética , Vetores Genéticos , Radioisótopos do Iodo/farmacocinética , Radioisótopos do Iodo/uso terapêutico , Adenocarcinoma/patologia , Adenoviridae , Animais , Membrana Celular , Proliferação de Células , Neoplasias Colorretais/patologia , Técnicas de Transferência de Genes , Humanos , Masculino , Camundongos , Camundongos Nus , Neoplasias da Próstata/patologia , Ratos , Esferoides Celulares , Simportadores , Células Tumorais Cultivadas
19.
Hum Gene Ther ; 31(17-18): 907, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32945724
20.
Hum Gene Ther ; 31(19-20): 1026, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33074025
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa