Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 14(2): e0211117, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30726287

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is characterized by a fibrotic stroma with a poor lymphocyte infiltrate, in part driven by cancer-associated fibroblasts (CAFs). CAFs, which express fibroblast activation protein (FAP), contribute to immune escape via exclusion of anti-tumor CD8+ T cells from cancer cells, upregulation of immune checkpoint ligand expression, immunosuppressive cytokine production, and polarization of tumor infiltrating inflammatory cells. FAP is a post-proline peptidase selectively expressed during tissue remodeling and repair, such as with wound healing, and in the tumor microenvironment by cancer-associated fibroblasts. We targeted FAP function using a novel small molecule inhibitor, UAMC-1110, and mice with germline knockout of FAP and concomitant knock-in of E. coli beta-galactosidase. We depleted CAFs by adoptive transfer of anti-ßgal T cells into the FAP knockout animals. Established syngeneic pancreatic tumors in immune competent mice were targeted with these 3 strategies, followed by focal radiotherapy to the tumor. FAP loss was associated with improved antigen-specific tumor T cell infiltrate and enhanced collagen deposition. However, FAP targeting alone or with tumor-directed radiation did not improve survival even when combined with anti-PD1 therapy. Targeting of CAFs alone or in combination with radiation did not improve survival. We conclude that targeting FAP and CAFs in combination with radiation is capable of enhancing anti-tumor T cell infiltrate and function, but does not result in sufficient tumor clearance to extend survival.


Assuntos
Anticorpos/metabolismo , Carcinoma Ductal Pancreático/terapia , Gelatinases/antagonistas & inibidores , Proteínas de Membrana/antagonistas & inibidores , Neoplasias Pancreáticas/terapia , Bibliotecas de Moléculas Pequenas/administração & dosagem , Linfócitos T/transplante , Transferência Adotiva , Animais , Fibroblastos Associados a Câncer/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Linhagem Celular Tumoral , Quimiorradioterapia , Terapia Combinada , Endopeptidases , Gelatinases/genética , Técnicas de Introdução de Genes , Técnicas de Inativação de Genes , Humanos , Proteínas de Membrana/genética , Camundongos , Neoplasias Pancreáticas/metabolismo , Serina Endopeptidases/genética , Bibliotecas de Moléculas Pequenas/farmacologia , Linfócitos T/imunologia , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto , beta-Galactosidase/imunologia
3.
Hepatol Res ; 47(7): 702-714, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27501850

RESUMO

AIM: Small, solitary hepatocellular carcinoma is curable with stereotactic radiation or other methods of tumor ablation, however, regional and systemic tumor recurrence occurs in over 70% of patients. Here we describe the ability of immunoradiotherapy to induce an antitumor immune response and delay the growth of tumors in immunocompetent mice. METHODS: A syngeneic hepatocellular carcinoma cell line (Hep-55.1c) was injected directly into the livers of C57BL/6 mice using ultrasound guidance, then tumors were treated with stereotactic radiation using a Small Animal Radiation Research Platform with computed tomography guidance. RESULTS: Delivery of three doses of 250 µg anti-programmed cell death protein-1 (αPD-1) antibody concurrently with 30 Gy stereotactic body radiation therapy in three fractions reduced the growth rate of tumors and improved survival (P < 0.05). Combined treatment was associated with increased CD8+ cytotoxic T cells in the tumor; depletion of CD8 T cells eliminated the efficacy of combined treatment. Combined treatment also induced expression of programmed cell death-1 ligand expression on tumor-infiltrating macrophages, and the tumors grew rapidly after αPD-1 treatment was discontinued. CONCLUSIONS: Tumor response to stereotactic radiation can be augmented by concurrent treatment with αPD-1. The efficacy of this combination therapy was transient, however, and treatment induced markers of adaptive immune resistance. These data are promising, but also indicate that mechanisms of immune resistance will need to be durably overcome for this combination to generate lasting immunity to protect against tumor recurrence.

4.
Oncotarget ; 7(48): 78653-78666, 2016 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-27602953

RESUMO

Radiation therapy provides a means to kill large numbers of cancer cells in a controlled location resulting in the release of tumor-specific antigens and endogenous adjuvants. However, by activating pathways involved in apoptotic cell recognition and phagocytosis, irradiated cancer cells engender suppressive phenotypes in macrophages. We demonstrate that the macrophage-specific phagocytic receptor, Mertk is upregulated in macrophages in the tumor following radiation therapy. Ligation of Mertk on macrophages results in anti-inflammatory cytokine responses via NF-kB p50 upregulation, which in turn limits tumor control following radiation therapy. We demonstrate that in immunogenic tumors, loss of Mertk is sufficient to permit tumor cure following radiation therapy. However, in poorly immunogenic tumors, TGFß inhibition is also required to result in tumor cure following radiation therapy. These data demonstrate that Mertk is a highly specific target whose absence permits tumor control in combination with radiation therapy.


Assuntos
Macrófagos/efeitos da radiação , Recidiva Local de Neoplasia , Neoplasias Experimentais/radioterapia , c-Mer Tirosina Quinase/metabolismo , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Compostos Azabicíclicos/farmacologia , Linhagem Celular Tumoral , Técnicas de Cocultura , Citocinas/metabolismo , Macrófagos/enzimologia , Macrófagos/imunologia , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Neoplasias Experimentais/enzimologia , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Células RAW 264.7 , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Transdução de Sinais/efeitos da radiação , Fatores de Tempo , Fator de Crescimento Transformador beta/metabolismo , c-Mer Tirosina Quinase/antagonistas & inibidores , c-Mer Tirosina Quinase/deficiência , c-Mer Tirosina Quinase/genética
5.
J Immunother Cancer ; 4: 45, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27532020

RESUMO

BACKGROUND: Preclinical studies have shown synergy between radiation therapy and immunotherapy. However, in almost all preclinical models, radiation is delivered in single doses or short courses of high doses (hypofractionated radiation). By contrast in most clinical settings, radiation is delivered as standard small daily fractions of 1.8-2 Gy to achieve total doses of 50-54 Gy (fractionated radiation). We do not yet know the optimal dose and scheduling of radiation for combination with chemotherapy and immunotherapy. METHODS: To address this, we analyzed the effect of neoadjuvant standard fractionated and hypofractionated chemoradiation on immune cells in patients with locally advanced and borderline resectable pancreatic adenocarcinoma. RESULTS: We found that standard fractionated chemoradiation resulted in a significant and extended loss of lymphocytes that was not explained by a lack of homeostatic cytokines or response to cytokines. By contrast, treatment with hypofractionated radiation therapy avoided the loss of lymphocytes associated with conventional fractionation. CONCLUSION: Hypofractionated neoadjuvant chemoradiation is associated with reduced systemic loss of T cells. TRIAL REGISTRATION: ClinicalTrials.gov NCT01342224, April 21, 2011; NCT01903083, July 2, 2013.

6.
PLoS One ; 11(6): e0157164, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27281029

RESUMO

The anecdotal reports of promising results seen with immunotherapy and radiation in advanced malignancies have prompted several trials combining immunotherapy and radiation. However, the ideal timing of immunotherapy with radiation has not been clarified. Tumor bearing mice were treated with 20Gy radiation delivered only to the tumor combined with either anti-CTLA4 antibody or anti-OX40 agonist antibody. Immunotherapy was delivered at a single timepoint around radiation. Surprisingly, the optimal timing of these therapies varied. Anti-CTLA4 was most effective when given prior to radiation therapy, in part due to regulatory T cell depletion. Administration of anti-OX40 agonist antibody was optimal when delivered one day following radiation during the post-radiation window of increased antigen presentation. Combination treatment of anti-CTLA4, radiation, and anti-OX40 using the ideal timing in a transplanted spontaneous mammary tumor model demonstrated tumor cures. These data demonstrate that the combination of immunotherapy and radiation results in improved therapeutic efficacy, and that the ideal timing of administration with radiation is dependent on the mechanism of action of the immunotherapy utilized.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Antígeno CTLA-4/imunologia , Neoplasias Colorretais/terapia , Imunoterapia , Neoplasias Mamárias Animais/terapia , Receptores OX40/imunologia , Linfócitos T Reguladores/imunologia , Animais , Apresentação de Antígeno , Antígeno CTLA-4/metabolismo , Neoplasias Colorretais/imunologia , Terapia Combinada , Fracionamento da Dose de Radiação , Feminino , Neoplasias Mamárias Animais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Fatores de Tempo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Cancer Res ; 76(1): 50-61, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26567136

RESUMO

Cytotoxic therapies prime adaptive immune responses to cancer by stimulating the release of tumor-associated antigens. However, the tumor microenvironment into which these antigens are released is typically immunosuppressed, blunting the ability to initiate immune responses. Recently, activation of the DNA sensor molecule STING by cyclic dinucleotides was shown to stimulate infection-related inflammatory pathways in tumors. In this study, we report that the inflammatory pathways activated by STING ligands generate a powerful adjuvant activity for enhancing adaptive immune responses to tumor antigens released by radiotherapy. In a murine model of pancreatic cancer, we showed that combining CT-guided radiotherapy with a novel ligand of murine and human STING could synergize to control local and distant tumors. Mechanistic investigations revealed T-cell-independent and TNFα-dependent hemorrhagic necrosis at early times, followed by later CD8 T-cell-dependent control of residual disease. Clinically, STING was found to be expressed extensively in human pancreatic tumor and stromal cells. Our findings suggest that this novel STING ligand could offer a potent adjuvant for leveraging radiotherapeutic management of pancreatic cancer.


Assuntos
Carcinoma Ductal Pancreático/terapia , Proteínas de Membrana/genética , Oligonucleotídeos/farmacologia , Neoplasias Pancreáticas/terapia , Animais , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/imunologia , Carcinoma Ductal Pancreático/radioterapia , Linhagem Celular Tumoral , Terapia Combinada , Modelos Animais de Doenças , Proteínas de Membrana/biossíntese , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Terapia de Alvo Molecular , Oligonucleotídeos/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/radioterapia , Distribuição Aleatória , Microambiente Tumoral
8.
Radiat Res ; 182(2): 182-90, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24992164

RESUMO

An accumulating body of evidence demonstrates that radiation therapy can generate adaptive immune responses that contribute to tumor control. However, in the absence of additional immune therapy, the adaptive immune response is insufficient to prevent tumor recurrence or affect distant disease. It has been shown in multiple models that tumor-infiltrating myeloid cells exhibit alternative activation phenotypes and are able to suppress adaptive immune responses, and recent data suggests that the myeloid response in tumors treated with cytotoxic therapy limits tumor control. We hypothesized that tumor myeloid cells inhibit the adaptive immune response after radiation therapy through expression of the enzyme arginase I. Using a myeloid cell-specific deletion of arginase I in mice, we demonstrate an improved tumor control after radiation therapy. However, tumors still recurred despite the conditional knockdown of arginase I. Since multiple alternative factors may combine to inhibit adaptive immunity, we propose that targeting macrophage differentiation may be a more effective strategy than targeting individual suppressive pathways.


Assuntos
Arginase/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Células Mieloides/metabolismo , Células Mieloides/efeitos da radiação , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/radioterapia , Imunidade Adaptativa/efeitos da radiação , Animais , Arginase/genética , Linhagem Celular Tumoral , Fracionamento da Dose de Radiação , Técnicas de Inativação de Genes , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/efeitos da radiação , Camundongos , Células Mieloides/imunologia , Neoplasia Residual , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia
9.
Cancer Immunol Res ; 2(10): 1011-22, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25047233

RESUMO

The immune infiltrate in colorectal cancer has been correlated with outcome, such that individuals with higher infiltrations of T cells have increased survival independent of the disease stage. For patients with lower immune infiltrates, overall survival is limited. Because the patients with colorectal cancer studied have received conventional cancer therapies, these data may indicate that the pretreatment tumor environment increases the efficacy of treatments such as chemotherapy, surgery, and radiotherapy. This study was designed to test the hypothesis that an improved immune environment in the tumor at the time of treatment will increase the efficacy of radiotherapy. We demonstrate that inhibition of TGFß using the orally available small-molecule inhibitor SM16 improved the immune environment of tumors in mice and significantly improved the efficacy of subsequent radiotherapy. This effect was not due to changes in radiosensitivity, epithelial-mesenchymal transition, or changes in vascular function in the tumor; rather, this effect was dependent on adaptive immunity and resulted in long-term protective immunity in cured mice. These data demonstrate that immunotherapy is an option to improve the immune status of patients with poor tumor infiltrates and that pretreatment improves the efficacy of radiotherapy.


Assuntos
Antineoplásicos/uso terapêutico , Compostos Azabicíclicos/uso terapêutico , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Mamárias Experimentais/tratamento farmacológico , Fator de Crescimento Transformador beta/antagonistas & inibidores , Imunidade Adaptativa/efeitos dos fármacos , Imunidade Adaptativa/imunologia , Animais , Quimioterapia Adjuvante/métodos , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/radioterapia , Avaliação Pré-Clínica de Medicamentos/métodos , Feminino , Estimativa de Kaplan-Meier , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/imunologia , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/radioterapia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Terapia Neoadjuvante/métodos , Transplante de Neoplasias , Tolerância a Radiação/efeitos dos fármacos , Células Tumorais Cultivadas
10.
PLoS One ; 8(7): e69527, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23936036

RESUMO

Expansion of myeloid-lineage leukocytes in tumor-bearing mice has been proposed as a cause of systemic immunosuppression. We demonstrate that radiation therapy of tumors leads to a decline in myeloid cell numbers in the blood and a decrease in spleen size. The frequency of myeloid cells does not decline to the level seen in tumor-free mice: we demonstrate that metastatic disease can prevent myeloid cell numbers from returning to baseline, and that tumor recurrence from residual disease correlates with re-expansion of myeloid lineage cells. Radiation therapy results in increased proliferation of T cells in the spleen and while T cell responses to foreign antigens are not altered by tumor burden or myeloid cell expansion, responses to tumor-associated antigens are increased after radiation therapy. These data demonstrate that myeloid cell numbers are directly linked to primary tumor burden, that this population contracts following radiation therapy, and that radiation therapy may open a therapeutic window for immunotherapy of residual disease.


Assuntos
Adenocarcinoma/radioterapia , Raios gama/uso terapêutico , Neoplasias Mamárias Experimentais/radioterapia , Células Mieloides/efeitos da radiação , Neoplasias Pancreáticas/radioterapia , Linfócitos T/efeitos da radiação , Carga Tumoral/efeitos da radiação , Adenocarcinoma/imunologia , Adenocarcinoma/patologia , Animais , Antígenos de Neoplasias/imunologia , Contagem de Células , Proliferação de Células , Feminino , Tolerância Imunológica , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/patologia , Camundongos , Células Mieloides/imunologia , Células Mieloides/patologia , Transplante de Neoplasias , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/patologia , Pele , Baço/imunologia , Baço/patologia , Baço/efeitos da radiação , Linfócitos T/imunologia , Linfócitos T/patologia , Transplante Heterotópico
11.
PLoS One ; 7(6): e39295, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22761754

RESUMO

Radiation therapy aims to kill cancer cells with a minimum of normal tissue toxicity. Dying cancer cells have been proposed to be a source of tumor antigens and may release endogenous immune adjuvants into the tumor environment. For these reasons, radiation therapy may be an effective modality to initiate new anti-tumor adaptive immune responses that can target residual disease and distant metastases. However, tumors engender an environment dominated by M2 differentiated tumor macrophages that support tumor invasion, metastases and escape from immune control. In this study, we demonstrate that following radiation therapy of tumors in mice, there is an influx of tumor macrophages that ultimately polarize towards immune suppression. We demonstrate using in vitro models that this polarization is mediated by transcriptional regulation by NFκB p50, and that in mice lacking NFκB p50, radiation therapy is more effective. We propose that despite the opportunity for increased antigen-specific adaptive immune responses, the intrinsic processes of repair following radiation therapy may limit the ability to control residual disease.


Assuntos
Macrófagos/metabolismo , Subunidade p50 de NF-kappa B/metabolismo , Neoplasias/metabolismo , Neoplasias/radioterapia , Animais , Linhagem Celular Tumoral , Humanos , Camundongos , Camundongos Knockout , Subunidade p50 de NF-kappa B/genética , Transplante de Neoplasias , Neoplasias/genética
12.
Eur J Neurosci ; 35(3): 468-77, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22277045

RESUMO

Stem cells derived from the human brain and grown as neurospheres (HuCNS-SC) have been shown to be effective in treating central neurodegenerative conditions in a variety of animal models. Human safety data in neurodegenerative disorders are currently being accrued. In the present study, we explored the efficacy of HuCNS-SC in a rodent model of retinal degeneration, the Royal College of Surgeons (RCS) rat, and extended our previous cell transplantation studies to include an in-depth examination of donor cell behavior and phenotype post-transplantation. As a first step, we have shown that HuCNS-SC protect host photoreceptors and preserve visual function after transplantation into the subretinal space of postnatal day 21 RCS rats. Moreover, cone photoreceptor density remained relatively constant over several months, consistent with the sustained visual acuity and luminance sensitivity functional outcomes. The novel findings of this study include the characterization and quantification of donor cell radial migration from the injection site and within the subretinal space as well as the demonstration that donor cells maintain an immature phenotype throughout the 7 months of the experiment and undergo very limited proliferation with no evidence of uncontrolled growth or tumor-like formation. Given the efficacy findings and lack of adverse events in the RCS rat in combination with the results from ongoing clinical investigations, HuCNS-SC appear to be a well-suited candidate for cell therapy in retinal degenerative conditions.


Assuntos
Células-Tronco Neurais/transplante , Fármacos Neuroprotetores , Degeneração Retiniana/prevenção & controle , Degeneração Retiniana/cirurgia , Transplante de Células-Tronco , Animais , Movimento Celular/fisiologia , Células Cultivadas , Modelos Animais de Doenças , Humanos , Células-Tronco Neurais/citologia , Ratos , Ratos Endogâmicos , Retina/citologia , Retina/metabolismo , Retina/patologia , Células Fotorreceptoras Retinianas Cones/citologia , Células Fotorreceptoras Retinianas Cones/fisiologia , Transplante Heterólogo , Visão Ocular/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...