Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Ann Surg Oncol ; 25(12): 3747-3754, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30187278

RESUMEN

BACKGROUND: Surgical stress results in a significant reduction in natural killer (NK) cell cytotoxicity (NKC), which has been linked to postoperative cancer metastases. However, few studies have measured the impact of surgical stress upon NK cell IFNγ secretion (NKA), a cytokine with essential roles in controlling infection and metastases. The objective of this study was to investigate the impact of surgical stress on NKA in colorectal cancer (CRC) surgery patients. METHODS: Peripheral blood was collected from CRC surgery patients (n = 42) preoperatively and on postoperative day (POD) 1, 3, 5, 28, and 56. Healthy donor blood (n = 27) was collected for controls. We assessed NKA by production of IFNγ following whole blood cytokine stimulation, NKC by 51Cr-release assay, and immune cell profiling by flow cytometry. RESULTS: The mean reduction in NKA on POD1 compared with baseline was 83.1% (standard deviation 25.2%; confidence interval 75-91), and therefore the study met the primary endpoint of demonstrating a > 75% decrease in a cohort of CRC surgery patients (p < 0.0001). The profound and universal suppression of NKA persisted with 65.5% (19/29) and 33.3% (4/12) of patients with levels measuring < 75% of baseline on POD28 and POD56 respectively. The NKC was significantly reduced on POD1, but the degree was less pronounced (24.6%, p = 0.0024). Immune cell profiling did not reveal differences in the absolute number of NK cells (CD3-CD56+) or the ratio of CD56dim-to-CD56bright subsets. CONCLUSIONS: NKA is significantly suppressed for up to two months following surgery in CRC patients, a degree of surgery-induced immunosuppression far worse than previously reported.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Colorrectales/patología , Cirugía Colorrectal/métodos , Interferón gamma/metabolismo , Células Asesinas Naturales/metabolismo , Complicaciones Posoperatorias , Anciano , Estudios de Casos y Controles , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/cirugía , Femenino , Estudios de Seguimiento , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/patología , Masculino , Persona de Mediana Edad , Cuidados Preoperatorios , Pronóstico , Estudios Prospectivos
2.
BMC Cancer ; 18(1): 277, 2018 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-29530012

RESUMEN

BACKGROUND: Cancer surgery can promote tumour metastases and worsen prognosis, however, the effect of perioperative complications on metastatic disease remains unclear. In this study we sought to evaluate the effect of common perioperative complications including perioperative blood loss, hypothermia, and sepsis on tumour metastases in a murine model. METHODS: Prior to surgery, pulmonary metastases were established by intravenous challenge of CT26LacZ colon cancer cells in BALB/c mice. Surgical stress was generated through partial hepatectomy (PH) or left nephrectomy (LN). Sepsis was induced by puncturing the cecum to express stool into the abdomen. Hemorrhagic shock was induced by removal of 30% of total blood volume (i.e. stage 3 hemorrhage) via the saphenous vein. Hypothermia was induced by removing the heating apparatus during surgery and lowering core body temperatures to 30 °C. Lung tumour burden was quantified 3 days following surgery. RESULTS: Surgically stressed mice subjected to stage 3 hemorrhage or hypothermia did not show an additional increase in lung tumour burden. In contrast, surgically stressed mice subjected to intraoperative sepsis demonstrated an additional 2-fold increase in the number of tumour metastases. Furthermore, natural killer (NK) cell function, as assessed by YAC-1 tumour cell lysis, was significantly attenuated in surgically stressed mice subjected to intraoperative sepsis. Both NK cell-mediated cytotoxic function and lung tumour burden were improved with perioperative administration of polyI:C, which is a toll-like receptor (TLR)-3 ligand. CONCLUSIONS: Perioperative sepsis alone, but not hemorrhage or hypothermia, enhances the prometastatic effect of surgery in murine models of cancer. Understanding the cellular mechanisms underlying perioperative immune suppression will facilitate the development of immunomodulation strategies that can attenuate metastatic disease.


Asunto(s)
Neoplasias del Colon/fisiopatología , Neoplasias Pulmonares/cirugía , Sepsis/fisiopatología , Animales , Ciego/fisiopatología , Ciego/cirugía , Neoplasias del Colon/sangre , Neoplasias del Colon/etiología , Neoplasias del Colon/secundario , Modelos Animales de Enfermedad , Hemorragia/complicaciones , Hemorragia/fisiopatología , Hepatectomía/efectos adversos , Humanos , Células Asesinas Naturales/patología , Neoplasias Pulmonares/sangre , Neoplasias Pulmonares/complicaciones , Neoplasias Pulmonares/fisiopatología , Ratones , Ratones Endogámicos BALB C , Metástasis de la Neoplasia , Nefrectomía/efectos adversos , Periodo Perioperatorio/efectos adversos , Sepsis/sangre , Sepsis/complicaciones
3.
BMC Cancer ; 18(1): 437, 2018 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-29665786

RESUMEN

It has been highlighted that the original manuscript [1] contains a typesetting error in Fig. 1 and the Fig. 1c panel gas been inadvertently duplicated in panel Fig. 1d. This does not affect the results and conclusions of the article. The correct version of Fig. 1 is included with this Correction. The original article has been updated.

4.
Mol Ther ; 22(7): 1320-1332, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24695102

RESUMEN

This study characterizes the ability of novel oncolytic rhabdoviruses (Maraba MG1) to boost natural killer (NK) cell activity. Our results demonstrate that MG1 activates NK cells via direct infection and maturation of conventional dendritic cells. Using NK depletion and conventional dendritic cells ablation studies in vivo, we established that both are required for MG1 efficacy. We further explored the efficacy of attenuated MG1 (nonreplicating MG1-UV(2min) and single-cycle replicating MG1-Gless) and demonstrated that these viruses activate conventional dendritic cells, although to a lesser extent than live MG1. This translates to equivalent abilities to remove tumor metastases only at the highest viral doses of attenuated MG1. In tandem, we characterized the antitumor ability of NK cells following preoperative administration of live and attenuated MG1. Our results demonstrates that a similar level of NK activation and reduction in postoperative tumor metastases was achieved with equivalent high viral doses concluding that viral replication is important, but not necessary for NK activation. Biochemical characterization of a panel of UV-inactivated MG1 (2-120 minutes) revealed that intact viral particle and target cell recognition are essential for NK cell-mediated antitumor responses. These findings provide mechanistic insight and preclinical rationale for safe perioperative virotherapy to effectively reduce metastatic disease following cancer surgery.


Asunto(s)
Células Dendríticas/citología , Células Asesinas Naturales/citología , Melanoma/terapia , Rhabdoviridae/fisiología , Animales , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Viroterapia Oncolítica/métodos
5.
Front Immunol ; 14: 1099459, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36969187

RESUMEN

Introduction: Adipocytes in the tumour microenvironment are highly dynamic cells that have an established role in tumour progression, but their impact on anti-cancer therapy resistance is becoming increasingly difficult to overlook. Methods: We investigated the role of adipose tissue and adipocytes in response to oncolytic virus (OV) therapy in adipose-rich tumours such as breast and ovarian neoplasms. Results: We show that secreted products in adipocyte-conditioned medium significantly impairs productive virus infection and OV-driven cell death. This effect was not due to the direct neutralization of virions or inhibition of OV entry into host cells. Instead, further investigation of adipocyte secreted factors demonstrated that adipocyte-mediated OV resistance is primarily a lipid-driven phenomenon. When lipid moieties are depleted from the adipocyte-conditioned medium, cancer cells are re-sensitized to OV-mediated destruction. We further demonstrated that blocking fatty acid uptake by cancer cells, in a combinatorial strategy with virotherapy, has clinical translational potential to overcome adipocyte-mediated OV resistance. Discussion: Our findings indicate that while adipocyte secreted factors can impede OV infection, the impairment of OV treatment efficacy can be overcome by modulating lipid flux in the tumour milieu.


Asunto(s)
Viroterapia Oncolítica , Virus Oncolíticos , Neoplasias Ováricas , Humanos , Femenino , Microambiente Tumoral , Medios de Cultivo Condicionados , Virus Oncolíticos/fisiología , Neoplasias Ováricas/terapia , Lípidos
6.
Nat Commun ; 13(1): 1898, 2022 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-35393414

RESUMEN

Recent advances in cancer therapeutics clearly demonstrate the need for innovative multiplex therapies that attack the tumour on multiple fronts. Oncolytic or "cancer-killing" viruses (OVs) represent up-and-coming multi-mechanistic immunotherapeutic drugs for the treatment of cancer. In this study, we perform an in-vitro screen based on virus-encoded artificial microRNAs (amiRNAs) and find that a unique amiRNA, herein termed amiR-4, confers a replicative advantage to the VSVΔ51 OV platform. Target validation of amiR-4 reveals ARID1A, a protein involved in chromatin remodelling, as an important player in resistance to OV replication. Virus-directed targeting of ARID1A coupled with small-molecule inhibition of the methyltransferase EZH2 leads to the synthetic lethal killing of both infected and uninfected tumour cells. The bystander killing of uninfected cells is mediated by intercellular transfer of extracellular vesicles carrying amiR-4 cargo. Altogether, our findings establish that OVs can serve as replicating vehicles for amiRNA therapeutics with the potential for combination with small molecule and immune checkpoint inhibitor therapy.


Asunto(s)
Vesículas Extracelulares , MicroARNs , Neoplasias , Viroterapia Oncolítica , Virus Oncolíticos , Humanos , MicroARNs/genética , Neoplasias/terapia , Virus Oncolíticos/genética
7.
Front Immunol ; 11: 963, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32508837

RESUMEN

The majority of data on human Natural Killer (NK) cell phenotype and function has been generated using cryopreserved peripheral blood mononuclear cells (PBMCs). However, cryopreservation can have adverse effects on PBMCs. In contrast, investigating immune cells in whole blood can reduce the time, volume of blood required, and potential artefacts associated with manipulation of the cells. Whole blood collected from healthy donors and cancer patients was processed by three separate protocols that can be used independently or in parallel to assess extracellular receptors, intracellular signaling protein phosphorylation, and intracellular and extracellular cytokine production in human NK cells. To assess extracellular receptor expression, 200 µL of whole blood was incubated with an extracellular staining (ECS) mix and cells were subsequently fixed and RBCs lysed prior to analysis. The phosphorylation status of signaling proteins was assessed in 500 µL of whole blood following co-incubation with interleukin (IL)-2/12 and an ECS mix for 20 min prior to cell fixation, RBC lysis, and subsequent permeabilization for staining with an intracellular staining (ICS) mix. Cytokine production (IFNγ) was similarly assessed by incubating 1 mL of whole blood with PMA-ionomycin or IL-2/12 prior to incubation with ECS and subsequent ICS antibodies. In addition, plasma was collected from stimulated samples prior to ECS for quantification of secreted IFNγ by ELISA. Results were consistent, despite inherent inter-patient variability. Although we did not investigate an exhaustive list of targets, this approach enabled quantification of representative ECS surface markers including activating (NKG2D and DNAM-1) and inhibitory (NKG2A, PD-1, TIGIT, and TIM-3) receptors, cytokine receptors (CD25, CD122, CD132, and CD212) and ICS markers associated with NK cell activation following stimulation, including signaling protein phosphorylation (p-STAT4, p-STAT5, p-p38 MAPK, p-S6) and IFNγ in both healthy donors and cancer patients. In addition, we compared extracellular receptor expression using whole blood vs. cryopreserved PBMCs and observed a significant difference in the expression of almost all receptors. The methods presented permit a relatively rapid parallel assessment of immune cell receptor expression, signaling protein activity, and cytokine production in a minimal volume of whole blood from both healthy donors and cancer patients.


Asunto(s)
Citometría de Flujo , Inmunofenotipificación , Interferón gamma/sangre , Péptidos y Proteínas de Señalización Intracelular/sangre , Células Asesinas Naturales/metabolismo , Neoplasias/sangre , Receptores Inmunológicos/sangre , Anciano , Biomarcadores/sangre , Estudios de Casos y Controles , Criopreservación , Estudios de Factibilidad , Femenino , Humanos , Células Asesinas Naturales/inmunología , Masculino , Persona de Mediana Edad , Neoplasias/inmunología , Fenotipo , Fosforilación , Prueba de Estudio Conceptual , Flujo de Trabajo
8.
Cancer Immunol Res ; 5(3): 211-221, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28159747

RESUMEN

Despite improvements in chemotherapy and radical surgical debulking, peritoneal carcinomatosis (PC) remains among the most common causes of death from abdominal cancers. Immunotherapies have been effective for selected solid malignancies, but their potential in PC has been little explored. Here, we report that intraperitoneal injection of an infected cell vaccine (ICV), consisting of autologous tumor cells infected ex vivo with an oncolytic Maraba MG1 virus expressing IL12, promotes the migration of activated natural killer (NK) cells to the peritoneal cavity in response to the secretion of IFNγ-induced protein-10 (IP-10) from dendritic cells. The recruitment of cytotoxic, IFNγ-secreting NK cells was associated with reduced tumor burden and improved survival in a colon cancer model of PC. Even in mice with bulky PC (tumors > 8 mm), a complete radiologic response was demonstrated within 8 to14 weeks, associated with 100% long-term survival. The impact of MG1-IL12-ICV upon NK-cell recruitment and function observed in the murine system was recapitulated in human lymphocytes exposed to human tumor cell lines infected with MG1-IL12. These findings suggest that an MG1-IL12-ICV is a promising therapy that could provide benefit to the thousands of patients diagnosed with PC each year. Cancer Immunol Res; 5(3); 211-21. ©2017 AACR.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Quimiotaxis/inmunología , Interleucina-12/genética , Células Asesinas Naturales/inmunología , Neoplasias Peritoneales/genética , Neoplasias Peritoneales/inmunología , Animales , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/genética , Línea Celular Tumoral , Citocinas/metabolismo , Citotoxicidad Inmunológica , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Femenino , Vectores Genéticos/genética , Humanos , Interleucina-12/metabolismo , Células Asesinas Naturales/metabolismo , Activación de Linfocitos/inmunología , Melanoma Experimental , Ratones , Virus Oncolíticos/genética , Neoplasias Peritoneales/patología , Neoplasias Peritoneales/terapia , Transducción Genética
10.
Mol Ther Oncolytics ; 3: 16027, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27909702

RESUMEN

The systemic delivery of therapeutic viruses, such as oncolytic viruses or vaccines, is limited by the generation of neutralizing antibodies. While pseudotyping of rhabdoviruses with the lymphocytic choriomeningitis virus glycoprotein has previously allowed for multiple rounds of delivery in mice, this strategy has not translated to other animal models. For the first time, we provide experimental evidence that antibodies generated against the lymphocytic choriomeningitis virus glycoprotein mediate robust complement-dependent viral neutralization via activation of the classical pathway. We show that this phenotype can be capitalized upon to deliver maraba virus pseudotyped with the lymphocytic choriomeningitis virus glycoprotein in a Fischer rat model in the face of neutralizing antibody through the use of complement modulators. This finding changes the understanding of the humoral immune response to arenaviruses, and also describes methodology to deliver viral vectors to their therapeutic sites of action without the interference of neutralizing antibody.

11.
Nat Med ; 21(5): 530-6, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25894825

RESUMEN

Tumors are complex ecosystems composed of networks of interacting 'normal' and malignant cells. It is well recognized that cytokine-mediated cross-talk between normal stromal cells, including cancer-associated fibroblasts (CAFs), vascular endothelial cells, immune cells, and cancer cells, influences all aspects of tumor biology. Here we demonstrate that the cross-talk between CAFs and cancer cells leads to enhanced growth of oncolytic virus (OV)-based therapeutics. Transforming growth factor-ß (TGF-ß) produced by tumor cells reprogrammed CAFs, dampened their steady-state level of antiviral transcripts and rendered them sensitive to virus infection. In turn, CAFs produced high levels of fibroblast growth factor 2 (FGF2), initiating a signaling cascade in cancer cells that reduced retinoic acid-inducible gene I (RIG-I) expression and impeded the ability of malignant cells to detect and respond to virus. In xenografts derived from individuals with pancreatic cancer, the expression of FGF2 correlated with the susceptibility of the cancer cells to OV infection, and local application of FGF2 to resistant tumor samples sensitized them to virotherapy both in vitro and in vivo. An OV engineered to express FGF2 was safe in tumor-bearing mice, showed improved therapeutic efficacy compared to parental virus and merits consideration for clinical testing.


Asunto(s)
Fibroblastos/metabolismo , Virus Oncolíticos/metabolismo , Microambiente Tumoral , Anciano , Animales , Antivirales/química , Línea Celular Tumoral , Chlorocebus aethiops , Técnicas de Cocultivo , Femenino , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Masculino , Ratones , Microscopía Fluorescente , Persona de Mediana Edad , Trasplante de Neoplasias , Viroterapia Oncolítica/métodos , Neoplasias Ováricas/metabolismo , Transducción de Señal , Células del Estroma/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Células Vero
12.
Cancer Res ; 73(1): 97-107, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23090117

RESUMEN

Natural killer (NK) cell clearance of tumor cell emboli following surgery is thought to be vital in preventing postoperative metastases. Using a mouse model of surgical stress, we transferred surgically stressed NK cells into NK-deficient mice and observed enhanced lung metastases in tumor-bearing mice as compared with mice that received untreated NK cells. These results establish that NK cells play a crucial role in mediating tumor clearance following surgery. Surgery markedly reduced NK cell total numbers in the spleen and affected NK cell migration. Ex vivo and in vivo tumor cell killing by NK cells were significantly reduced in surgically stressed mice. Furthermore, secreted tissue signals and myeloid-derived suppressor cell populations were altered in surgically stressed mice. Significantly, perioperative administration of oncolytic parapoxvirus ovis (ORFV) and vaccinia virus can reverse NK cell suppression, which correlates with a reduction in the postoperative formation of metastases. In human studies, postoperative cancer surgery patients had reduced NK cell cytotoxicity, and we show for the first time that oncolytic vaccinia virus markedly increases NK cell activity in patients with cancer. These data provide direct in vivo evidence that surgical stress impairs global NK cell function. Perioperative therapies aimed at enhancing NK cell function will reduce metastatic recurrence and improve survival in surgical cancer patients.


Asunto(s)
Células Asesinas Naturales/inmunología , Metástasis de la Neoplasia/prevención & control , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Células Neoplásicas Circulantes/inmunología , Viroterapia Oncolítica/métodos , Procedimientos Quirúrgicos Operativos/efectos adversos , Animales , Citometría de Flujo , Humanos , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Metástasis de la Neoplasia/inmunología , Neoplasias Experimentales/cirugía , Células Neoplásicas Circulantes/patología , Virus Oncolíticos , Estrés Fisiológico/inmunología
13.
Clin Cancer Res ; 19(18): 5104-15, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23881927

RESUMEN

PURPOSE: Surgical removal of solid primary tumors is an essential component of cancer treatment. Surgery-induced dysfunction in natural killer (NK) cells has been linked to the development of metastases in animal models and patients with cancer. We investigated the activation of NK cells using influenza vaccine in the perioperative period to eradicate micrometastatic disease. EXPERIMENTAL DESIGN: Both the B16lacZ and 4T1 tumor models in immunocompetent mice were used to assess the in vivo efficacy of perioperative influenza vaccine administration. In healthy human donors and cancer surgery patients, we assessed NK cell function pre- and post-influenza vaccination using both in vivo and ex vivo assays. RESULTS: Using the TLR3 agonist poly(I:C), we showed as proof-of-principle that perioperative administration of a nonspecific innate immune stimulant can inhibit surgery-induced dysfunction in NK cells and attenuate metastases. Next, we assessed a panel of prophylactic vaccines for NK cell activation and determined that inactivated influenza vaccine was the best candidate for perioperative administration. Perioperative influenza vaccine significantly reduced tumor metastases and improved NK cytotoxicity in preclinical tumor models. Significantly, IFNα is the main cytokine mediator for the therapeutic effect of influenza vaccination. In human studies, influenza vaccine significantly enhanced NK cell activity in healthy human donors and cancer surgery patients. CONCLUSION: These results provide the preclinical rationale to pursue future clinical trials of perioperative NK cell activation, using vaccination in cancer surgery patients. Research into perioperative immune therapy is warranted to prevent immune dysfunction following surgery and eradicate metastatic disease.


Asunto(s)
Vacunas contra la Influenza/uso terapéutico , Células Asesinas Naturales/inmunología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/prevención & control , Melanoma Experimental/cirugía , Procedimientos Quirúrgicos Operativos/efectos adversos , Animales , Citotoxicidad Inmunológica/inmunología , Humanos , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/cirugía , Activación de Linfocitos , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Atención Perioperativa , Periodo Posoperatorio , Receptor de Interferón alfa y beta/fisiología , Células Tumorales Cultivadas , Vacunación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA