Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Oncol ; 12: 772403, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35463378

RESUMO

Purpose: Prostate biopsy histopathology and immunohistochemistry are important in the differential diagnosis of the disease and can be used to assess the degree of prostate cancer differentiation. Today, prostate biopsy is increasing the demand for experienced uropathologists, which puts a lot of pressure on pathologists. In addition, the grades of different observations had an indicating effect on the treatment of the patients with cancer, but the grades were highly changeable, and excessive treatment and insufficient treatment often occurred. To alleviate these problems, an artificial intelligence system with clinically acceptable prostate cancer detection and Gleason grade accuracy was developed. Methods: Deep learning algorithms have been proved to outperform other algorithms in the analysis of large data and show great potential with respect to the analysis of pathological sections. Inspired by the classical semantic segmentation network, we propose a pyramid semantic parsing network (PSPNet) for automatic prostate Gleason grading. To boost the segmentation performance, we get an auxiliary prediction output, which is mainly the optimization of auxiliary objective function in the process of network training. The network not only includes effective global prior representations but also achieves good results in tissue micro-array (TMA) image segmentation. Results: Our method is validated using 321 biopsies from the Vancouver Prostate Centre and ranks the first on the MICCAI 2019 prostate segmentation and classification benchmark and the Vancouver Prostate Centre data. To prove the reliability of the proposed method, we also conduct an experiment to test the consistency with the diagnosis of pathologists. It demonstrates that the well-designed method in our study can achieve good results. The experiment also focused on the distinction between high-risk cancer (Gleason pattern 4, 5) and low-risk cancer (Gleason pattern 3). Our proposed method also achieves the best performance with respect to various evaluation metrics for distinguishing benign from malignant. Availability: The Python source code of the proposed method is publicly available at https://github.com/hubutui/Gleason. All implementation details are presented in this paper. Conclusion: These works prove that the Gleason grading results obtained from our method are effective and accurate.

2.
Mol Ther Methods Clin Dev ; 25: 136, 2022 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-35402631

RESUMO

[This corrects the article DOI: 10.1016/j.omtm.2021.08.007.].

3.
Cancer Immunol Immunother ; 71(6): 1479-1495, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34716463

RESUMO

An important mechanism of oncolytic virotherapy in ameliorating cancer immunotherapy is by inducing significant changes in the immune landscape in the tumor microenvironment (TME). Despite this notion and the potential therapeutic implications, a comprehensive analysis of the immune changes in carcinomas induced by virotherapy has not yet been elucidated. We conducted single-cell RNA sequencing analysis on carcinomas treated with an HSV-2-based oncolytic virus to characterize the immunogenic changes in the TME. We specifically analyzed and compared the immune cell composition between viral treated and untreated tumors. We also applied CellChat to analyze the complex interactions among the infiltrated immune cells. Our data revealed significant infiltration of B cells in addition to other important immune cells, including CD4+, CD8+, and NK cells following virotherapy. Further analysis identified distinct subset compositions of the infiltrated immune cells and their activation status upon virotherapy. The intensive interactions among the infiltrated immune cells as revealed by CellChat analysis may further shape the immune landscape in favor of generating antitumor immunity. Our findings will facilitate the design of new strategies in incorporating immunotherapy into virotherapy for clinical translation. Moreover, the significant infiltration of B cells makes it suitable for combining virotherapy with immune checkpoint inhibitors.


Assuntos
Carcinoma , Terapia Viral Oncolítica , Vírus Oncolíticos , Humanos , Imunoterapia , Vírus Oncolíticos/genética , Análise de Sequência de RNA , Microambiente Tumoral
4.
Mol Ther Oncolytics ; 23: 330-341, 2021 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-34786476

RESUMO

The oncolytic effect of virotherapy derives from the intrinsic capability of the applied virus in selectively infecting and killing tumor cells. Although oncolytic viruses of various constructions have been shown to efficiently infect and kill tumor cells in vitro, the efficiency of these viruses to exert the same effect on tumor cells within tumor tissues in vivo has not been extensively investigated. Here we report our studies using single-cell RNA sequencing to comprehensively analyze the gene expression profile of tumor tissues following herpes simplex virus 2-based oncolytic virotherapy. Our data revealed the extent and cell types within the tumor microenvironment that could be infected by the virus. Moreover, we observed changes in the expression of cellular genes, including antiviral genes, in response to viral infection. One notable gene found to be upregulated significantly in oncolytic virus-infected tumor cells was Gadd45g, which is desirable for optimal virus replication. These results not only help reveal the precise infection status of the oncolytic virus in vivo but also provide insight that may lead to the development of new strategies to further enhance the therapeutic efficacy of oncolytic virotherapy.

5.
Mol Ther Methods Clin Dev ; 23: 78-86, 2021 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-34631928

RESUMO

The current methods for detecting circulating tumor cells (CTCs) suffer from several drawbacks. We report a novel method that is based on a chimeric virus probe and can detect CTCs with extremely high specificity and sensitivity. Moreover, it exclusively detects live CTCs, and its detection efficacy is not impacted by the variation of epithelial cell adhesion molecule (EpCAM) expression. The chimeric virus probe is composed of a capsid from human papillomavirus that provides the detection with high specificity and an SV40-based genome that can amplify extensively inside CTCs and, hence, endows the detection with high sensitivity. Furthermore, different marker genes can be incorporated into the probe to provide detection with versatility. These unique capabilities will likely improve the validity and utility of this CTC detection in several clinical applications, which is one of the drawbacks suffered by many of the current CTC detection methods.

6.
J Immunother Cancer ; 9(7)2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34230110

RESUMO

BACKGROUND: Although oncolytic virotherapy has shown substantial promises as a new treatment modality for many malignancies, further improvement on its therapeutic efficacy will likely bring more clinical benefits. One plausible way of enhancing the therapeutic effect of virotherapy is to enable it with the ability to concurrently engage the infiltrating immune cells to provide additional antitumor mechanisms. Here, we report the construction and evaluation of two novel chimeric molecules (bispecific chimeric engager proteins, BiCEP and trispecific chimeric engager protein, TriCEP) that can engage both natural killer (NK) and T cells with tumor cells for enhanced antitumor activities. METHODS: BiCEP was constructed by linking orthopoxvirus major histocompatibility complex class I-like protein, which can selectively bind to NKG2D with a high affinity to a mutant form of epidermal growth factor (EGF) that can strongly bind to EGF receptor. TriCEP is similarly constructed except that it also contains a modified form of interleukin-2 that can only function as a tethered form. As NKG2D is expressed on both NK and CD8+ T cells, both of which can thus be engaged by BiCEP and TriCEP. RESULTS: Both BiCEP and TriCEP showed the ability to engage NK and T cells to kill tumor cells in vitro. Coadministration of BiCEP and TriCEP with an oncolytic herpes simplex virus enhanced the overall antitumor effect. Furthermore, single-cell RNA sequencing analysis revealed that TriCEP not only engaged NK and T cells to kill tumor cells, it also promotes the infiltration and activation of these important immune cells. CONCLUSIONS: These novel chimeric molecules exploit the ability of the oncolytic virotherapy in altering the tumor microenvironment with increased infiltration of important immune cells such as NK and T cells for cancer immunotherapy. The ability of BiCEP and TriCEP to engage both NK and T cells makes them an ideal choice for arming an oncolytic virotherapy.


Assuntos
Anticorpos Biespecíficos/metabolismo , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Simplexvirus/efeitos dos fármacos , Humanos , Simplexvirus/genética
7.
JBMR Plus ; 5(5): e10484, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33977202

RESUMO

Osteoporosis causes fragile bone, and bone microstructural quality is a critical determinant of bone strength and fracture risk. This study pursues technical validation of novel CT-based methods for assessment of peripheral bone microstructure together with a human pilot study examining relationships between bone microstructure and vertebral fractures in smokers. To examine the accuracy and reproducibility of the methods, repeat ultra-high-resolution (UHR) CT and micro-CT scans of cadaveric ankle specimens were acquired. Thirty smokers from the University of Iowa COPDGene cohort were recruited at their 5-year follow-up visits. Chest CT scans, collected under the parent study, were used to assess vertebral fractures. UHR CT scans of distal tibia were acquired for this pilot study to obtain peripheral cortical and trabecular bone (Cb and Tb) measures. UHR CT-derived Tb measures, including volumetric bone mineral density (BMD), network area, transverse trabecular density, and mean plate width, showed high correlation (r > 0.901) with their micro-CT-derived values over small regions of interest (ROIs). Both Cb and Tb measures showed high reproducibility-intra-class correlation (ICC) was greater than 0.99 for all Tb measures except erosion index and greater than 0.97 for all Cb measures. Female sex was associated with lower transverse Tb density (p < 0.1), higher Tb spacing (p < 0.05), and lower cortical thickness (p < 0.001). Participants with vertebral fractures had significantly degenerated values (p < 0.05) for all Tb measures except thickness. There were no statistically significant differences for Cb measures between non-fracture and fracture groups. Vertebral fracture-group differences of Tb measures remained significant after adjustment with chronic obstructive pulmonary disease (COPD) status. Although current smokers at baseline had more fractures-81.8% versus 63.2% for former smokers-the difference was not statistically significant. This pilot cross-sectional human study demonstrates CT-based peripheral bone microstructural differences among smokers with and without vertebral fractures. © 2021 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research. © 2021 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.

8.
Mol Ther Oncolytics ; 19: 33-46, 2020 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-33024817

RESUMO

One of the major hurdles for cancer immunotherapy is the host's innate antiviral defense mechanisms. They include innate immune cells, such as natural killer (NK) cells and macrophages, which can be recruited within hours to the site of injection to clear the introduced oncolytic viruses. Here, we report a strategy to redirect these infiltrating innate immune cells to attack tumor cells instead by arming herpes simplex virus (HSV)-derived oncolytic viruses with secreted chimeric molecules that can engage these innate immune cells with tumor cells to kill the latter. These chimeric molecules have, at their N terminus, a custom-binding moiety for a tumor-associated antigen (TAA) and at their C terminus, protein L (PL) that binds to immunoglobulins (Igs). The binding of PL to Igs exposes the Fc to the Fc receptors on the surface of the innate immune cells, trigging them to attack the engaged tumor cells. In vitro and in vivo evaluation in a murine tumor model with limited permissiveness to oncolytic HSVs showed that arming the viruses with these chimeric molecules significantly boosts the killing effect and therapeutic activity. Moreover, our data also showed that the combined killing effect from the engaged innate immune cells and the oncolytic virus resulted in a more efficient stimulation of neoantigen-specific antitumor immunity than the virotherapy alone. Our data suggest that arming an oncolytic virus with this strategy represents a unique and pragmatic way of potentiating the oncolytic and immunotherapeutic effect of virotherapy.

9.
Oncotarget ; 9(77): 34543-34553, 2018 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-30349648

RESUMO

Current oncolytic virotherapy is primarily administered by intratumoral injection. However, systemic delivery is desirable for treating patients, particularly for those who have developed metastatic diseases. Several components are impeding the systemic delivery efficiency of oncolytic viruses. Chief among them is the rapid clearance of viral particles by the host's mononuclear phagocyte system (MPS). We explored the possibility of genetically engrafting CD47, a "don't eat me" signal molecule, to the membrane envelop of an oncolytic herpes simplex virus (HSV) to enable it to escape from the MPS for systemic delivery. Our results show that this modification indeed allows the virus to be more efficiently delivered to local tumors by the systemic route. Moreover, this modification also prolongs the virus persistence in local tumors after it arrives there. Consequently, systemic delivery of the modified virus produced a measurable antitumor effect against a murine tumor model that is otherwise resistant to the parental virus delivered by the same route. Our data thus suggest that engrafting enveloped oncolytic viruses such as those derived from HSV with CD47 molecule represents a conceivable strategy to enhance the efficiency of systemic delivery.

10.
Oncotarget ; 9(30): 21348-21358, 2018 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-29765544

RESUMO

Herpes simplex virus (HSV) is one of the many viruses that have been modified or adapted for oncolytic purposes. There are two serotypes of HSV, HSV-1 and HSV-2. The majority of oncolytic HSVs, including T-VEC which has recently been approved by the US Food and Drug Administration (FDA) for clinical use in treating late stage melanoma patients, are derived from HSV-1. Recently, we and others have developed several HSV-2 based oncolytic viruses. During our in vitro characterization of oncolytic viruses developed from both serotypes (Baco-1 from HSV-1 and FusOn-H2 from HSV-2), we noticed there is a subpopulation of cancer cells in which both viruses could infect but only FusOn-H2 could spread from cell to cell on monolayers. This observation prompted us to investigate the virus receptor expression profiles in these and other tumor cells. Our data show the following: 1) This subpopulation of tumor cells only express nectin-2, not the other two major receptors (HVEM or nectin-1). 2) Baco-1 grows to a higher titer than FusOn-H2 in this subpopulation of tumor cells, but the latter kills these tumor cells more efficiently than the former. 3) FusOn-H2 is effective at treating tumors formed from these tumor cells while Baco-1 is completely ineffective. Our results suggest that this subpopulation of tumor cells may be intrinsically resistant to the therapeutic effect of a HSV-1 based oncolytic virus but they remain sensitive to a HSV-2 based virotherapy.

11.
Endocr Relat Cancer ; 25(4): 453-469, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29431615

RESUMO

Despite altered metabolism being an accepted hallmark of cancer, it is still not completely understood which signaling pathways regulate these processes. Given the central role of androgen receptor (AR) signaling in prostate cancer, we hypothesized that AR could promote prostate cancer cell growth in part through increasing glucose uptake via the expression of distinct glucose transporters. Here, we determined that AR directly increased the expression of SLC2A12, the gene that encodes the glucose transporter GLUT12. In support of these findings, gene signatures of AR activity correlated with SLC2A12 expression in multiple clinical cohorts. Functionally, GLUT12 was required for maximal androgen-mediated glucose uptake and cell growth in LNCaP and VCaP cells. Knockdown of GLUT12 also decreased the growth of C4-2, 22Rv1 and AR-negative PC-3 cells. This latter observation corresponded with a significant reduction in glucose uptake, indicating that additional signaling mechanisms could augment GLUT12 function in an AR-independent manner. Interestingly, GLUT12 trafficking to the plasma membrane was modulated by calcium/calmodulin-dependent protein kinase kinase 2 (CaMKK2)-5'-AMP-activated protein kinase (AMPK) signaling, a pathway we previously demonstrated to be a downstream effector of AR. Inhibition of CaMKK2-AMPK signaling decreased GLUT12 translocation to the plasma membrane by inhibiting the phosphorylation of TBC1D4, a known regulator of glucose transport. Further, AR increased TBC1D4 expression. Correspondingly, expression of TBC1D4 correlated with AR activity in prostate cancer patient samples. Taken together, these data demonstrate that prostate cancer cells can increase the functional levels of GLUT12 through multiple mechanisms to promote glucose uptake and subsequent cell growth.


Assuntos
Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/metabolismo , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Próstata/metabolismo , Neoplasias da Próstata/metabolismo , Receptores Androgênicos/metabolismo , Transdução de Sinais/fisiologia , Androgênios/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proteínas Facilitadoras de Transporte de Glucose/genética , Humanos , Masculino , Metribolona/farmacologia , Fosforilação/efeitos dos fármacos , Próstata/efeitos dos fármacos , Próstata/patologia , Neoplasias da Próstata/patologia , RNA Interferente Pequeno , Transdução de Sinais/efeitos dos fármacos
12.
Sci Rep ; 7(1): 16836, 2017 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-29203891

RESUMO

RNA-guided endonucleases (RGENs) have invigorated the field of site-specific nucleases. The success of Streptococcus pyogenes Cas9 (SpCas9) has led to the discovery of several other CRISPR-associated RGENs. As more RGENs become available, it will be necessary to refine their activity before they can be translated into the clinic. With this in mind, we sought to demonstrate how deep mutational scanning (DMS) could provide details about important functional regions in SpCas9 and speed engineering efforts. Consequently, we developed a nuclease screening platform which could distinguish active Cas9 mutants. We screened a library of 1.9 × 107 with over 8500 possible non-synonymous mutations and inferred the effects of each mutation using DMS. We demonstrate that the RuvC and HNH domains are the least tolerant regions to mutation. In contrast, the Rec2 and PI domains tolerate mutation better than other regions. The mutation information defined in this work provides a foundation for further SpCas9 engineering. Together, our results demonstrate how DMS can be a powerful tool to uncover features important to RGEN function. Application of this approach to emerging RGENs should enhance their engineering and optimization for therapeutic and other applications.


Assuntos
Sistemas CRISPR-Cas/genética , Streptococcus pyogenes/genética , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Linhagem Celular Tumoral , DNA Bacteriano/química , DNA Bacteriano/metabolismo , Edição de Genes , Biblioteca Gênica , Humanos , Hidrolases/química , Hidrolases/genética , Mutação , Plasmídeos/genética , Plasmídeos/metabolismo
13.
BMC Cancer ; 15: 964, 2015 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-26674012

RESUMO

BACKGROUND: Many commonly used xenograft tumor models do not spontaneously metastasize to distant organs following subcutaneous or orthotopic implantation, limiting their usefulness in preclinical studies. It is generally believed that natural killer cells are the key component of the innate immune system in determining tumor metastatic potential in xenograft models. However, recent studies suggest that macrophages may play an important role, as resident macrophages can eliminate the invading tumor cells if they do not express adequate levels of the CD47 molecule. METHODS: We investigated the effect of overexpressing murine CD47 (mCD47) in PC-3 cells, a commonly used human prostate cancer line, on the metastatic potential in three mouse strains with different genetic background and varying degrees of immunodeficiency. We implanted the tumor cells either subcutaneously or orthotopically and then examined their local and distant metastases. RESULTS: Our results show that mCD47-expressing PC-3 cells subcutaneously implanted in NSG and CB17. Scid mice metastasized to the sentinel lymph node, lung and liver significantly more efficiently than the control cells. When implanted orthotopically to NOD. Scid mice, these cells spontaneously metastasized to lung and liver. CONCLUSIONS: Our data demonstrate that mCD47 can facilitate human tumor cell metastasis in murine models, and that these mCD47-expressing tumor cells may be useful for in vivo studies where spontaneous metastases are desirable.


Assuntos
Antígeno CD47/biossíntese , Modelos Animais de Doenças , Neoplasias da Próstata/patologia , Animais , Antígeno CD47/imunologia , Linhagem Celular Tumoral , Separação Celular , Citometria de Fluxo , Xenoenxertos , Humanos , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Invasividade Neoplásica/imunologia , Transplante de Neoplasias , Neoplasias da Próstata/imunologia , Transfecção
14.
Theranostics ; 5(10): 1058-67, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26199645

RESUMO

Herein, we report the development of a new "smart" radioactive probe (i.e., 1) which can undergo furin-controlled condensation and self-assembly of radioactive nanoparticles (i.e., 1-NPs) in tumor cells and its application for enhanced microPET imaging of tumors in nude mice co-injected with its cold analog (i.e., 1-Cold). Furin-controlled condensation of 1-Cold and self-assembly of its nanoparticles (i.e., 1-Cold-NPs) in vitro were validated and characterized with HPLC, mass spectra, SEM, and TEM analyses. Cell uptake studies showed that both 1 and 1-Cold have good cell permeability. TEM images of 1-Cold-treated MDA-MB-468 cells directly uncovered that the intracellular 1-Cold-NPs were at/near the location of furin (i.e., Golgi bodies). MTT results indicated that 50 µM 1-Cold did not impose cytotoxicity to MDA-MB-468 cells up to 12 hours. MicroPET imaging of MDA-MB-468 tumor-bearing mice indicated that mice co-injected with 1 and 1-Cold showed higher uptake and longer attenuation of the radioactivity in tumors than those mice only injected with same dosage of 1. Tumor uptake ratios of 1 between these two groups of mice reached the maximum of 8.2 folds at 240 min post injection. Biodistribution study indicated that the uptake ratios of 1 in kidneys between these two groups continuously increased and reached 81.9 folds at 240 min post injection, suggesting the formation of radioactive NPs (i.e., 1-NPs) in MDA-MB-468 tumors of mice co-injected with 1 and 1-Cold. And the nanoparticles were slowly digested and secreted from the tumors, accumulating in the kidneys. Our ''smart'' probe (i.e., 1), together with the strategy of co-injection, might help researchers trace the biomarkers of interest within a longer time window.


Assuntos
Radioisótopos de Flúor/química , Furina/química , Neoplasias/diagnóstico por imagem , Animais , Linhagem Celular Tumoral , Diagnóstico por Imagem , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanopartículas/química , Peptídeos/química , Tomografia por Emissão de Pósitrons/instrumentação
15.
Bladder Cancer ; 1(1): 81-90, 2015 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-30561438

RESUMO

PURPOSE: Despite recent improvements, resistance to traditional immunotherapy or chemotherapy is still common in patients with bladder cancer. We constructed an oncolytic virus from herpes simplex virus type II (HSV-2), which selectively targets tumor cells with an activated Ras signaling pathway. We evaluated the antitumor effect of this oncolytic HSV-2 (FusOn-H2) against bladder cancer, and compared with that of a first generation oncolytic virus derived from HSV-1 (Baco-1). MATERIALS AND METHODS: We established bladder tumor at the orthotopic site in C3H/He mice using the MBT-2 cells. Baco-1 or FusOn-H2 was instilled into the bladder through the urethra respectively. Tumor volume and weight were recorded by the end of the experiment. Animal spleens were also collected to determine if any anti-tumor immunity was elicited during virotherapy in this syngeneic bladder cancer model. RESULTS: Two instillations of the oncolytic HSVs into bladder of tumor-bearing mice almost completely eradicated the tumor in majority of tumor bearing mice. The results of tumor-specific cytotoxic T lymphocyte activity assay showed that tumor destruction by oncolytic viruses in vivo, especially by the FusOn-H2, induced potent anti-tumor immune responses. CONCLUSION: Oncolytic virus derived from HSV-2 has potent anti-tumor activity against bladder cancer. Oncolytic effect of this virus in vivo induces tumor specific cellular immunity that further enhances the overall anti-tumor activity. Translating this novel virotherapy into the clinic could present an alternative intravesical therapy strategy for patients with bladder cancer.

16.
Oncotarget ; 6(2): 902-14, 2015 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-25460506

RESUMO

Adoptive T-cell therapy has shown promises for cancer treatment. However, for treating solid tumors, there is a need for improving the ability of the adoptively transferred T cells to home to tumor sites. We explored the possibility of using an oncolytic virus derived from HSV-2, which can actively pull T effector cells to the site of infection, as a local attractant for migration of adoptively transferred T cells. Our data show that intratumoral administration of this virus can indeed attract active migration of the adoptively transferred T cells to the treated tumor. Moreover, once attracted to the tumor site by the virus, T cells persisted in there significantly longer than in mock-treated tumor. Chemokine profiling identified significant elevation of CXCL9 and CXCL10, as well as several other chemokines belonging to the inflammatory chemokine family in the virus-treated tumors. These chemokines initially guided the T-cell migration to and then maintained their persistence in the tumor site, leading to a significantly enhanced therapeutic effect. Our data suggests that this virotherapy may be combined with adoptive T-cell therapy to potentiate its therapeutic effect against solid tumors that are otherwise difficult to manage with the treatment alone.


Assuntos
Movimento Celular/imunologia , Herpesvirus Humano 2/imunologia , Neoplasias Experimentais/imunologia , Vírus Oncolíticos/imunologia , Linfócitos T/imunologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Quimiocinas/imunologia , Quimiocinas/metabolismo , Chlorocebus aethiops , Terapia Combinada , Herpesvirus Humano 2/fisiologia , Humanos , Imunoterapia Adotiva/métodos , Subunidade gama Comum de Receptores de Interleucina/deficiência , Subunidade gama Comum de Receptores de Interleucina/genética , Subunidade gama Comum de Receptores de Interleucina/imunologia , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Camundongos Transgênicos , Neoplasias Experimentais/terapia , Neoplasias Experimentais/virologia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/fisiologia , Receptores de Quimiocinas/imunologia , Receptores de Quimiocinas/metabolismo , Linfócitos T/transplante , Carga Tumoral/imunologia , Células Vero
17.
PLoS One ; 9(4): e95790, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24755922

RESUMO

Recent work has shown that the combinatorial use of multiple TALE activators can selectively activate certain cellular genes in inaccessible chromatin regions. In this study, we aimed to interrogate the activation potential of TALEs upon transcriptionally silenced immune genes in the context of non-immune cells. We designed a unique strategy, in which a single TALE fused to the TATA-box binding protein (TBP-TALE) is coupled with multiple VP64-TALE activators. We found that our strategy is significantly more potent than multiple TALE activators alone in activating expression of IL-2 and GM-CSF in diverse cell origins in which both genes are otherwise completely silenced. Chromatin analysis revealed that the gene activation was due in part to displacement of a distinctly positioned nucleosome. These studies provide a novel epigenetic mechanism for artificial gene induction and have important implications for targeted cancer immunotherapy, DNA vaccine development, as well as rational design of TALE activators.


Assuntos
Citocinas/genética , Regulação da Expressão Gênica , Regiões Promotoras Genéticas , Proteína de Ligação a TATA-Box/metabolismo , Transativadores/metabolismo , Transcrição Gênica , Ativação Transcricional , Sítios de Ligação , Linhagem Celular , Ordem dos Genes , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Humanos , Interleucina-2/genética , Especificidade de Órgãos , Ligação Proteica , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteína de Ligação a TATA-Box/genética , Transativadores/genética
18.
Int J Cancer ; 133(10): 2483-92, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23661285

RESUMO

Converting T cells into tumor cell killers by grafting them with a chimeric antigen receptor (CAR) has shown promise as a cancer immunotherapeutic. However, the inability of these cells to actively migrate and extravasate into tumor parenchyma has limited their effectiveness in vivo. Here we report the construction of a CAR containing an echistatin as its targeting moiety (eCAR). As echistatin has high binding affinity to αvß3 integrin that is highly expressed on the surface of endothelial cells of tumor neovasculature, T cells engrafted with eCAR (T-eCAR) can efficiently lyse human umbilical vein endothelial cells and tumor cells that express αvß3 integrin when tested in vitro. Systemic administration of T-eCAR led to extensive bleeding in tumor tissues with no evidence of damage to blood vessels in normal tissues. Destruction of tumor blood vessels by T-eCAR significantly inhibited the growth of established bulky tumors. Moreover, when T-eCAR was codelivered with nanoparticles in a strategically designed temporal order, it dramatically increased nanoparticle deposition in tumor tissues, pointing to the possibility that it may be used together with nanocarriers to increase their capability to selectively deliver antineoplastic drugs to tumor tissues.


Assuntos
Melanoma Experimental/irrigação sanguínea , Melanoma Experimental/terapia , Nanopartículas/administração & dosagem , Linfócitos T/fisiologia , Sequência de Aminoácidos , Animais , Antígenos de Neoplasias/biossíntese , Antígenos de Neoplasias/genética , Vasos Sanguíneos/patologia , Linhagem Celular , Linhagem Celular Tumoral , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Integrina alfaVbeta3/metabolismo , Masculino , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Proteínas Mutantes Quiméricas/biossíntese , Proteínas Mutantes Quiméricas/genética , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Receptores de Antígenos/metabolismo , Linfócitos T/imunologia
19.
Mol Ther ; 20(10): 1871-81, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22692498

RESUMO

Interferon (IFN) antiviral defense mechanism plays a critical role in controlling virus infection. It thus represents a formidable hurdle for virotherapy. Despite the reported ability of herpes simplex virus (HSV) to counteract this defense, the duration and extent of HSV infection in vivo is still largely dictated by host's IFN activity status. Because the HSV genes that have been reported to block IFN activity mainly act intracellularly, we hypothesized that their inhibitory effect could be enhanced by exploiting a gene whose product acts extracellularly. The B18R gene from vaccinia virus encodes a secreted decoy receptor with a broad antagonizing effect against type I IFNs. We therefore cloned B18R into an HSV-1-based oncolytic virus to generate Synco-B18R. In the presence of increased IFN levels in vitro, Synco-B18R largely retained its oncolytic effect, whereas the tumor-killing ability of the parental virus, Synco-2D, was severely compromised. When injected intratumorally in vivo, Synco-B18R showed significantly greater oncolytic activity than Synco-2D. Our results suggest that incorporation of the vaccinia virus B18R gene can safely potentiate the antitumor effect of an oncolytic HSV, and that similar strategies may be useful with other types of oncolytic viruses.


Assuntos
Herpesvirus Humano 1/genética , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Vaccinia virus/genética , Proteínas Virais/genética , Animais , Antivirais/farmacologia , Linhagem Celular Tumoral , Chlorocebus aethiops , Clonagem Molecular , Feminino , Herpesvirus Humano 1/fisiologia , Humanos , Interferons/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Vírus Oncolíticos/fisiologia , Plasmídeos , Recombinação Genética , Células Vero , Replicação Viral
20.
Curr Pharm Biotechnol ; 13(9): 1773-85, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21740353

RESUMO

The recent FDA approval of Sipuleucel-T for the treatment of prostate cancer represents an important milestone of cancer immunotherapy, which, for the first time, validates the concept of bringing true clinical benefit to cancer patients by stimulating patients' own anti-tumor immunity. Among the different experimental cancer immunotherapies, oncolytic virotherapy may represent a low-cost yet potent and personalized cancer vaccine for the treatment of solid tumors. This review describes the constructions of several human herpes simplex virus (HSV)-derived oncolytic viruses as candidate cancer vaccines, which induce specific and potent anti-tumor immunity in pre-clinical models, and thus resulting in stronger overall anti-tumor efficacy as compared to oncolytic effect alone. This article also describes the approaches to enhance the antitumor immunity of oncolytic HSVs, and in particular, the key role played by integrating membrane-fusion activity into these viruses. Additionally, this article reviews the potential effect of certain chemotherapeutic agents (e.g. cyclophosphamide) in boosting antitumor immunity induced by oncolytic HSV, and the mechanisms behind it. In summary, all the preclinical and clinical data have suggested that HSV-based oncolytic virotherapies could likely be developed as a new generation of cancer vaccines for the treatment of solid tumors.


Assuntos
Vacinas Anticâncer/uso terapêutico , Neoplasias/terapia , Neoplasias/virologia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/fisiologia , Medicina de Precisão/métodos , Simplexvirus/fisiologia , Animais , Ensaios Clínicos como Assunto , Avaliação Pré-Clínica de Medicamentos , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Vírus Oncolíticos/genética , Vírus Oncolíticos/metabolismo , Simplexvirus/genética , Simplexvirus/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA