Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Immunol Invest ; 49(7): 808-823, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32498585

RESUMEN

INTRODUCTION: The tumor-draining lymph node (TDLN) plays a role in tumor immunity. Intratumorally administered microspheres (MS) that encapsulate immunomodulatory agents have emerged as a treatment strategy capable of causing profound changes in the tumor microenvironment (TME) and eliciting potent antitumor effects. We hypothesized that local delivery of MS to the TME may also drain to and therefore target the TDLN to initiate antitumor immune responses. METHODS: Fluorescent MS were injected into orthotopically implanted murine pancreatic tumors, and tissues were examined by whole-mount microscopy and imaging flow cytometry. The role of the TDLN was investigated for mice treated with intratumoral interleukin-12 (IL-12)-encapsulated MS in combination with stereotactic body radiotherapy (SBRT) by cytokine profile and TDLN ablation. RESULTS: Fluorescent AF-594 MS delivered intratumorally were detected in the tumor, peritumoral lymphatics, and the TDLN 2 h after injection. Phagocytic cells were observed with internalized fluorescent MS. SBRT + IL-12 MS-induced upregulation of Th1 and antitumor factors IL-12, IFN-γ, CXCL10, and granzyme B in the TDLN, and excision of the TDLN partially abrogated treatment efficacy. CONCLUSIONS: Our results demonstrate that intratumorally administered MS not only target the TME, but also drain to the TDLN. Furthermore, MS encapsulated with a potent antitumor cytokine, IL-12, induce an antitumor cytokine profile in the TDLN, which is essential for treatment efficacy.


Asunto(s)
Antineoplásicos Inmunológicos/administración & dosificación , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/patología , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/patología , Microesferas , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Animales , Biomarcadores , Biomarcadores de Tumor , Carcinoma Ductal Pancreático/diagnóstico por imagen , Terapia Combinada , Manejo de la Enfermedad , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunofenotipificación , Ganglios Linfáticos/inmunología , Ratones , Terapia Molecular Dirigida/métodos , Neoplasias Pancreáticas/diagnóstico por imagen , Neoplasias Pancreáticas/etiología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Gut ; 67(6): 1112-1123, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29196437

RESUMEN

OBJECTIVE: Chemokine pathways are co-opted by pancreatic adenocarcinoma (PDAC) to facilitate myeloid cell recruitment from the bone marrow to establish an immunosuppressive tumour microenvironment (TME). Targeting tumour-associated CXCR2+neutrophils (TAN) or tumour-associated CCR2+ macrophages (TAM) alone improves antitumour immunity in preclinical models. However, a compensatory influx of an alternative myeloid subset may result in a persistent immunosuppressive TME and promote therapeutic resistance. Here, we show CCR2 and CXCR2 combined blockade reduces total tumour-infiltrating myeloids, promoting a more robust antitumour immune response in PDAC compared with either strategy alone. METHODS: Blood, bone marrow and tumours were analysed from PDAC patients and controls. Treatment response and correlative studies were performed in mice with established orthotopic PDAC tumours treated with a small molecule CCR2 inhibitor (CCR2i) and CXCR2 inhibitor (CXCR2i), alone and in combination with chemotherapy. RESULTS: A systemic increase in CXCR2+ TAN correlates with poor prognosis in PDAC, and patients receiving CCR2i showed increased tumour-infiltrating CXCR2+ TAN following treatment. In an orthotopic PDAC model, CXCR2 blockade prevented neutrophil mobilisation from the circulation and augmented chemotherapeutic efficacy. However, depletion of either CXCR2+ TAN or CCR2+ TAM resulted in a compensatory response of the alternative myeloid subset, recapitulating human disease. This was overcome by combined CCR2i and CXCR2i, which augmented antitumour immunity and improved response to FOLFIRINOX chemotherapy. CONCLUSION: Dual targeting of CCR2+ TAM and CXCR2+ TAN improves antitumour immunity and chemotherapeutic response in PDAC compared with either strategy alone.


Asunto(s)
Carcinoma Ductal Pancreático/inmunología , Macrófagos/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Neoplasias Pancreáticas/inmunología , Microambiente Tumoral/inmunología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/metabolismo , Bases de Datos Factuales , Citometría de Flujo , Humanos , Inmunohistoquímica , Inmunomodulación , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Células Mieloides/efectos de los fármacos , Infiltración Neutrófila/efectos de los fármacos , Neutrófilos/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores CCR2/antagonistas & inhibidores , Receptores CCR2/metabolismo , Receptores de Interleucina-8B/antagonistas & inhibidores , Receptores de Interleucina-8B/metabolismo , Análisis de Matrices Tisulares , Microambiente Tumoral/efectos de los fármacos
3.
Nano Lett ; 13(12): 6197-202, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24274657

RESUMEN

Piezoelectric nanowires are an important class of smart materials for next-generation applications including energy harvesting, robotic actuation, and bioMEMS. Lead zirconate titanate (PZT), in particular, has attracted significant attention, owing to its superior electromechanical conversion performance. Yet, the ability to synthesize crystalline PZT nanowires with well-controlled properties remains a challenge. Applications of common nanosynthesis methods to PZT are hampered by issues such as slow kinetics, lack of suitable catalysts, and harsh reaction conditions. Here we report a versatile biomimetic method, in which biotemplates are used to define PZT nanostructures, allowing for rational control over composition and crystallinity. Specifically, stoichiometric PZT nanowires were synthesized using both polysaccharide (alginate) and bacteriophage templates. The wires possessed measured piezoelectric constants of up to 132 pm/V after poling, among the highest reported for PZT nanomaterials. Further, integrated devices can generate up to 0.820 µW/cm(2) of power. These results suggest that biotemplated piezoelectric nanowires are attractive candidates for stimuli-responsive nanosensors, adaptive nanoactuators, and nanoscale energy harvesters.


Asunto(s)
Plomo/química , Sistemas Microelectromecánicos , Nanocables/química , Titanio/química , Circonio/química , Bacteriófagos/química , Fuentes de Energía Bioeléctrica , Nanoestructuras/química , Polisacáridos/química
4.
J Immunother Cancer ; 10(7)2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35851308

RESUMEN

BACKGROUND: Stereotactic body radiotherapy (SBRT) has been increasingly used as adjuvant therapy in pancreatic ductal adenocarcinoma (PDAC), and induces immunogenic cell death, which leads to the release of tumor antigen and damage-associated molecular patterns. However, this induction often fails to generate sufficient response to overcome pre-existing tumor microenvironment (TME) immunosuppression. Toll-like receptor (TLR) 7/8 ligands, such as R848, can amplify the effect of tumor vaccines, with recent evidence showing its antitumor effect in pancreatic cancer by modulating the immunosuppressive TME. Therefore, we hypothesized that the combination of R848 and SBRT would improve local and systemic antitumor immune responses by potentiating the antitumor effects of SBRT and reversing the immunosuppressive nature of the PDAC TME. METHODS: Using murine models of orthotopic PDAC, we assessed the combination of intravenous TLR7/8 agonist R848 and local SBRT on tumor growth and immune response in primary pancreatic tumors. Additionally, we employed a hepatic metastatic model to investigate if the combination of SBRT targeting only the primary pancreatic tumor and systemic R848 is effective in controlling established liver metastases. RESULTS: We demonstrated that intravenous administration of the TLR7/8 agonist R848, in combination with local SBRT, leads to superior tumor control compared with either treatment alone. The combination of R848 and SBRT results in significant immune activation of the pancreatic TME, including increased tumor antigen-specific CD8+ T cells, decreased regulatory T cells, and enhanced antigen-presenting cells maturation, as well as increased interferon gamma, granzyme B, and CCL5 along with decreased levels of interleukin 4 (IL-4), IL-6, and IL-10. Importantly, the combination of SBRT and systemic R848 also resulted in similar immunostimulatory changes in liver metastases, leading to improved metastatic control. CD8+ T cell depletion studies highlighted the necessity of these effector cells at both the local and hepatic metastatic sites. T cell receptor (TCR) clonotype analysis indicated that systemic R848 not only diversified the TCR repertoire but also conditioned the metastatic foci to facilitate entry of CD8+ T cells generated by SBRT therapy. CONCLUSIONS: These findings suggest that systemic administration of TLR7/8 agonists in combination with SBRT may be a promising avenue for metastatic PDAC treatment.


Asunto(s)
Carcinoma Ductal Pancreático , Imidazoles/farmacología , Neoplasias Hepáticas , Neoplasias Pancreáticas , Radiocirugia , Adyuvantes Inmunológicos/farmacología , Animales , Antígenos de Neoplasias , Linfocitos T CD8-positivos , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/radioterapia , Modelos Animales de Enfermedad , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/radioterapia , Ratones , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/radioterapia , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 8/agonistas , Microambiente Tumoral , Neoplasias Pancreáticas
5.
Cancers (Basel) ; 13(10)2021 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-34064795

RESUMEN

Metastatic melanoma portends a poor prognosis and patients may present with multiple, simultaneous tumors. Despite recent advances in systemic immunotherapy, a majority of patients fail to respond, or exhibit lesion-specific responses wherein some metastases respond as others progress within the same patient. While intertumoral heterogeneity has been clinically associated with these mixed lesion-specific therapeutic responses, no clear mechanism has been identified, largely due to the scarcity of preclinical models. We developed a novel murine synchronous melanoma model that recapitulates this intertumoral genetic and microenvironmental heterogeneity. We show that genetic differences between tumors are sufficient to generate distinct tumor immune microenvironments (TIME) simultaneously in the same mouse. Furthermore, these TIMEs lead to the independent regulation of PD-1/PD-L1 (programmed cell death protein 1/PD-1 ligand), a popular axis targeted by immune checkpoint therapy, in response to ongoing anti-tumor immunity and the presence of interferon-gamma. Currently, therapeutic selection for metastatic melanoma patients is guided by a single biopsy, which may not represent the immune status of all tumors. As a result, patients can display heterogeneous lesion-specific responses. Further investigations into this synchronous melanoma model will provide mechanistic insight into the effects of intertumoral heterogeneity and guide therapeutic selection in this challenging patient population.

6.
Cancer Immunol Res ; 8(1): 94-107, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31719057

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) continues to have a dismal prognosis, in part, due to ineffective treatment strategies. The efficacy of some chemotherapies and especially radiotherapy is mediated partially by the immune system. Therefore, we hypothesized that profiling the immune response following chemotherapy and/or irradiation can be used as a readout for treatment efficacy but also to help identify optimal therapeutic schedules for PDAC. Using murine models of PDAC, we demonstrated that concurrent administration of stereotactic body radiotherapy (SBRT) and a modified dose of FOLFIRINOX (mFX) resulted in superior tumor control when compared with single or sequential treatment groups. Importantly, this combined treatment schedule enhanced the magnitude of immunogenic cell death, which in turn amplified tumor antigen presentation by dendritic cells and intratumoral CD8+ T-cell infiltration. Concurrent therapy also resulted in systemic immunity contributing to the control of established metastases. These findings provide a rationale for pursuing concurrent treatment schedules of SBRT with mFX in PDAC.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Muerte Celular Inmunogénica , Neoplasias Pulmonares/secundario , Neoplasias Experimentales/patología , Neoplasias Pancreáticas/patología , Radiocirugia/métodos , Animales , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Terapia Combinada , Células Dendríticas/inmunología , Femenino , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/terapia , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/terapia , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/terapia , Microambiente Tumoral/inmunología
7.
Cell Rep ; 29(2): 406-421.e5, 2019 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-31597100

RESUMEN

Over 80% of pancreatic ductal adenocarcinoma (PDA) patients are diagnosed with non-resectable late-stage disease that lacks effective neoadjuvant therapies. Stereotactic body radiation therapy (SBRT) has shown promise as an emerging neoadjuvant approach for treating PDA, and here, we report that its combination with local interleukin-12 (IL-12) microsphere (MS) immunotherapy results in marked tumor reduction and cures in multiple preclinical mouse models of PDA. Our findings demonstrate an increase of intratumoral interferon gamma (IFNγ) production following SBRT/IL-12 MS administration that initiates suppressor cell reprogramming and a subsequent increase in CD8 T cell activation. Furthermore, SBRT/IL-12 MS therapy results in the generation of systemic tumor immunity that is capable of eliminating established liver metastases, providing a rationale for follow-up studies in advanced metastatic disease.


Asunto(s)
Interleucina-12/uso terapéutico , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/terapia , Radiocirugia , Microambiente Tumoral/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Reprogramación Celular , Humanos , Inmunidad , Interferón gamma/metabolismo , Activación de Linfocitos/inmunología , Ratones Endogámicos C57BL , Microesferas , Modelos Biológicos , Células Mieloides/patología , Análisis de Supervivencia , Carga Tumoral , Neoplasias Pancreáticas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA