Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Int J Mol Sci ; 24(14)2023 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-37511431

RESUMEN

Therapeutic options for metastatic colorectal cancer (mCRC) are very limited, and the prognosis using combination therapy with a chemotherapeutic drug and a targeted agent, e.g., epidermal growth factor receptor or tyrosine kinase, remains poor. Therefore, mCRC is associated with a poor median overall survival (mOS) of only 25-30 months. Current immunotherapies with checkpoint inhibitor blockade (ICB) have led to a substantial change in the treatment of several cancers, such as melanoma and non-small cell lung cancer. In CRC, ICB has only limited effects, except in patients with microsatellite instability-high (MSI-H) or mismatch repair-deficient (dMMR) tumors, which comprise about 15% of sporadic CRC patients and about 4% of patients with metastatic CRC. The vast majority of sporadic CRCs are microsatellite-stable (MSS) tumors with low levels of infiltrating immune cells, in which immunotherapy has no clinical benefit so far. Immunotherapy with checkpoint inhibitors requires the presence of infiltrating T cells into the tumor microenvironment (TME). This makes T cells the most important effector cells in the TME, as evidenced by the establishment of the immunoscore-a method to estimate the prognosis of CRC patients. The microenvironment of a tumor contains several types of T cells that are anti-tumorigenic, such as CD8+ T cells or pro-tumorigenic, such as regulatory T cells (Tregs) or T helper 17 (Th17) cells. However, even CD8+ T cells show marked heterogeneity, e.g., they can become exhausted, enter a state of hyporesponsiveness or become dysfunctional and express high levels of checkpoint molecules, the targets for ICB. To kill cancer cells, CD8+ T cells need the recognition of the MHC class I, which is often downregulated on colorectal cancer cells. In this case, a population of unconventional T cells with a γδ T cell receptor can overcome the limitations of the conventional CD8+ T cells with an αßT cell receptor. γδ T cells recognize antigens in an MHC-independent manner, thus acting as a bridge between innate and adaptive immunity. Here, we discuss the effects of different T cell subsets in colorectal cancer with a special emphasis on γδ T cells and the possibility of using them in CAR-T cell therapy. We explain T cell exclusion in microsatellite-stable colorectal cancer and the possibilities to overcome this exclusion to enable immunotherapy even in these "cold" tumors.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Colorrectales , Neoplasias Pulmonares , Humanos , Linfocitos T CD8-positivos/metabolismo , Microambiente Tumoral , Subgrupos de Linfocitos T/metabolismo , Neoplasias Colorrectales/metabolismo , Inestabilidad de Microsatélites , Reparación de la Incompatibilidad de ADN
2.
Nature ; 494(7437): 361-5, 2013 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-23376950

RESUMEN

Cancer control by adaptive immunity involves a number of defined death and clearance mechanisms. However, efficient inhibition of exponential cancer growth by T cells and interferon-γ (IFN-γ) requires additional undefined mechanisms that arrest cancer cell proliferation. Here we show that the combined action of the T-helper-1-cell cytokines IFN-γ and tumour necrosis factor (TNF) directly induces permanent growth arrest in cancers. To safely separate senescence induced by tumour immunity from oncogene-induced senescence, we used a mouse model in which the Simian virus 40 large T antigen (Tag) expressed under the control of the rat insulin promoter creates tumours by attenuating p53- and Rb-mediated cell cycle control. When combined, IFN-γ and TNF drive Tag-expressing cancers into senescence by inducing permanent growth arrest in G1/G0, activation of p16INK4a (also known as CDKN2A), and downstream Rb hypophosphorylation at serine 795. This cytokine-induced senescence strictly requires STAT1 and TNFR1 (also known as TNFRSF1A) signalling in addition to p16INK4a. In vivo, Tag-specific T-helper 1 cells permanently arrest Tag-expressing cancers by inducing IFN-γ- and TNFR1-dependent senescence. Conversely, Tnfr1(-/-)Tag-expressing cancers resist cytokine-induced senescence and grow aggressively, even in TNFR1-expressing hosts. Finally, as IFN-γ and TNF induce senescence in numerous murine and human cancers, this may be a general mechanism for arresting cancer progression.


Asunto(s)
Senescencia Celular/inmunología , Citocinas/inmunología , Neoplasias/inmunología , Neoplasias/patología , Células TH1/inmunología , Animales , Antígenos Transformadores de Poliomavirus/genética , Antígenos Transformadores de Poliomavirus/metabolismo , Ciclo Celular , Proliferación Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/deficiencia , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Humanos , Interferón gamma/inmunología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Oncogenes/genética , Fosfoserina/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Proteína de Retinoblastoma/química , Proteína de Retinoblastoma/metabolismo , Factor de Transcripción STAT1/metabolismo , Factores de Tiempo , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/inmunología , Proteína p53 Supresora de Tumor/metabolismo
3.
Cancer Metastasis Rev ; 36(2): 357-365, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28391403

RESUMEN

The immune response is a first-line systemic defense to curb tumorigenesis and metastasis. Much effort has been invested to design antitumor interventions that would boost the immune system in its fight to defeat or contain cancerous growth. Tumor vaccination protocols, transfer of tumor-associated-antigen-specific T cells, T cell activity-regulating antibodies, and recombinant cytokines are counted among a toolbox filled with immunotherapeutic options. Although the mechanistic underpinnings of tumor immune control remain to be deciphered, these are studied with the goal of cancer cell destruction. In contrast, tumor dormancy is considered as a dangerous equilibrium between cell proliferation and cell death. There is, however, emerging evidence that tumor immune control can be achieved in the absence of overt cancer cell death. Here, we propose cytokine-induced senescence (CIS) by transfer of T helper-1 cells (TH1) or by recombinant cytokines as a novel therapeutic intervention for cancer treatment. Immunity-induced senescence triggers a stable cell cycle arrest of cancer cells. It engages the immune system to construct defensive, isolating barriers around tumors, and prevents tumor growth through the delivery or induction of TH1-cytokines in the tumor microenvironment. Keeping cancer cells in a non-proliferating state is a strategy, which directly copes with the lost homeostasis of aggressive tumors. As most studies show that even after efficient cancer therapies minimal residual disease persists, we suggest that therapies should include immune-mediated senescence for cancer surveillance. CIS has the goal to control the residual tumor and to transform a deadly disease into a state of silent tumor persistence.


Asunto(s)
Citocinas/inmunología , Neoplasias/inmunología , Animales , Procesos de Crecimiento Celular/inmunología , Senescencia Celular/inmunología , Citocinas/farmacología , Humanos , Monitorización Inmunológica , Neoplasias/patología , Neoplasias/terapia , Linfocitos T Colaboradores-Inductores/inmunología
4.
Cell Physiol Biochem ; 51(3): 1103-1118, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30476917

RESUMEN

BACKGROUND/AIMS: Cellular senescence, or permanent growth arrest, is known as an effective tumor suppressor mechanism that can be induced by different stressors, such as oncogenes, chemotherapeutics or cytokine cocktails. Previous studies demonstrated that the growth-repressing state of oncogene-induced senescent cells depends on argonaute protein 2 (Ago2)-mediated transcriptional gene silencing and Ago2/Rb corepression of E2F-dependent cell cycle genes. Cytokine-induced senescence (CIS) likewise depends on activation of the p16Ink4a/Rb pathway, and consecutive inactivation of the E2F family of transcription factors. In the present study, we therefore analyzed the role of Ago2 in CIS. METHODS: Human cancer cell lines were treated with interferon-gamma (IFN-γ) and tumor necrosis factor (TNF) to induce senescence. Senescence was determined by growth assays and measurement of senescence-associated ß-galactosidase (SA-ß-gal) activity, Ago2 translocation by Ago2/ Ki67 immunofluorescence staining and western blot analysis, and gene transcription by quantitative polymerase chain reaction (qPCR). RESULTS: IFN-γ and TNF permanently stopped cell proliferation and time-dependently increased SA-ß-gal activity. After 24 - 48 h of cytokine treatment, Ago2 translocated from the cytoplasm into the nucleus of Ki67-negative cells, an effect which was shown to be reversible. Importantly, the proinflammatory cytokine cocktail suppressed Ago2-regulated cell cycle control genes, and siRNA-mediated depletion of Ago2 interfered with cytokine-induced growth inhibition. CONCLUSION: IFN-γ and TNF induce a stable cell cycle arrest of cancer cells that is accompanied by a fast nuclear Ago2 translocation and repression of Ago2-regulated cell cycle control genes. As Ago2 downregulation impairs cytokine-induced growth regulation, Ago2 may contribute to tissue homeostasis in human cancers.


Asunto(s)
Proteínas Argonautas/metabolismo , Senescencia Celular , Citocinas/metabolismo , Neoplasias/metabolismo , Transporte Activo de Núcleo Celular , Proliferación Celular , Supervivencia Celular , Humanos , Interferón gamma/metabolismo , Células MCF-7 , Factores de Necrosis Tumoral/metabolismo
5.
J Dtsch Dermatol Ges ; 14(1): 28-37, 2016 Jan.
Artículo en Inglés, Alemán | MEDLINE | ID: mdl-26713633

RESUMEN

Forty years of research have brought about the development of antibodies that induce effective antitumor immune responses through sustained activation of the immune system. These "immune checkpoint inhibitors" are directed against immune inhibitory molecules, such as cytotoxic T lymphocyte antigen 4 (CTLA-4), programmed death 1 (PD-1) or programmed death ligand 1 (PD-L1). Disruption of the PD-1/PD-L1 interaction improves the intermediate-term prognosis even in patients with advanced stage IV melanoma. One and a half years after treatment initiation, 30-60 % of these patients are still alive. While cancer immunotherapies usually do not eradicate metastases completely, they do cause a regression by 20-80 %. It is well established that the immune system is able to kill tumor cells, and this has also been demonstrated for immunotherapies. Preclinical data, however, has shown that anti-cancer immunity is not limited to killing cancer cells. Thus, through interferon gamma and tumor necrosis factor, the immune system is able to induce stable tumor growth arrest, referred to as senescence. Ensuring patient survival by long-term stabilization of metastatic growth will therefore become a central goal of antitumor immunotherapies. This therapeutic approach is effective in melanoma and non-small-cell lung cancer. Once immunotherapies also have an indication for common cancer types, drug prices will have to drop considerably in order to be able to keep them available to those dependent on such therapies.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Inmunoterapia/métodos , Melanoma/terapia , Terapia Molecular Dirigida/métodos , Neoplasias Cutáneas/terapia , Medicina Basada en la Evidencia , Humanos , Resultado del Tratamiento
6.
Cancers (Basel) ; 14(6)2022 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-35326515

RESUMEN

In contrast to surgical excision, chemotherapy or radiation therapy, immune checkpoint blockade therapies primarily influence cells in the tumor microenvironment, especially the tumor-associated lymphocytes and antigen-presenting cells. Besides complete remission of tumor lesions, in some patients, early tumor regression is followed by a consolidation phase where residing tumors remain dormant. Whereas the cytotoxic mechanisms of the regression phase (i.e., apoptosis, necrosis, necroptosis, and immune cell-mediated cell death) have been extensively described, the mechanisms underlying the dormant state are still a matter of debate. Here, we propose immune-mediated induction of senescence in cancers as one important player. Senescence can be achieved by tumor-associated antigen-specific T helper 1 cells, cytokines or antibodies targeting immune checkpoints. This concept differs from cytotoxic treatment, which often targets the genetic makeup of cancer cells. The immune system's ability to establish "defensive walls" around tumors also places the tumor microenvironment into the fight against cancer. Those "defensive walls" isolate the tumor cells instead of increasing the selective pressure. They also keep the tumor cells in a non-proliferating state, thereby correcting the derailed tissue homeostasis. In conclusion, strengthening the senescence surveillance of tumors by the immune cells of the microenvironment is a future goal to dampen this life-threatening disease.

7.
Cells ; 12(1)2022 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-36611932

RESUMEN

Colorectal cancer (CRC) is one of the most frequent tumor entities worldwide with only limited therapeutic options. CRC is not only a genetic disease with several mutations in specific oncogenes and/or tumor suppressor genes such as APC, KRAS, PIC3CA, BRAF, SMAD4 or TP53 but also a multifactorial disease including environmental factors. Cancer cells communicate with their environment mostly via soluble factors such as cytokines, chemokines or growth factors to generate a favorable tumor microenvironment (TME). The TME, a heterogeneous population of differentiated and progenitor cells, plays a critical role in regulating tumor development, growth, invasion, metastasis and therapy resistance. In this context, cytokines from cancer cells and cells of the TME influence each other, eliciting an inflammatory milieu that can either enhance or suppress tumor growth and metastasis. Additionally, several lines of evidence exist that the composition of the microbiota regulates inflammatory processes, controlled by cytokine secretion, that play a role in carcinogenesis and tumor progression. In this review, we discuss the cytokine networks between cancer cells and the TME and microbiome in colorectal cancer and the related treatment strategies, with the goal to discuss cytokine-mediated strategies that could overcome the common therapeutic resistance of CRC tumors.


Asunto(s)
Neoplasias Colorrectales , Citocinas , Humanos , Citocinas/genética , Neoplasias Colorrectales/patología , Oncogenes , Mutación , Quimiocinas/genética , Microambiente Tumoral
8.
Front Immunol ; 13: 908449, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35844581

RESUMEN

More than half of all patients with colorectal cancer (CRC) develop distant metastasis and, depending on the local stage of the primary tumor, up to 48% of patients present peritoneal carcinomatosis (PC). PC is often considered as a widespread metastatic disease, which is almost resistant to current systemic therapies like chemotherapeutic and immunotherapeutic regimens. Here we could show that tumor cells of PC besides being senescent also exhibit stem cell features. To investigate these surprising findings in more detail, we established a murine model based on tumor organoids that resembles the clinical setting. In this murine orthotopic transplantation model for peritoneal carcinomatosis, we could show that the metastatic site in the peritoneum is responsible for senescence and stemness induction in tumor cells and that induction of senescence is not due to oncogene activation or therapy. In both mouse and human PC, senescence is associated with a senescence-associated secretory phenotype (SASP) influencing the tumor microenvironment (TME) of PC. SASP factors are able to induce a senescence phenotype in neighbouring cells. Here we could show that SASP leads to enhanced immunosenescence in the TME of PC. Our results provide a new immunoescape mechanism in PC explaining the resistance of PC to known chemo- and immunotherapeutic approaches. Therefore, senolytic approaches may represent a novel roadmap to target this terminal stage of CRC.


Asunto(s)
Inmunosenescencia , Neoplasias Peritoneales , Animales , Humanos , Ratones , Peritoneo/patología , Fenotipo , Microambiente Tumoral
9.
Blood ; 113(15): 3494-502, 2009 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-19188665

RESUMEN

Experimental tumor vaccination and adoptive T-cell therapies show that interferon-gamma (IFN-gamma)-producing CD4(+) T helper cells (Th1) can be highly effective in tumor prevention and therapy. Unexpectedly, first vaccine trials in humans revealed that tumor immune therapy may not only be protective, but, on the contrary, even promote tumor progression. Here, we analyzed T-cell immune responses to the epithelial cell adhesion molecule (EpCAM), one of the most common tumor-associated antigens (TAA) serving as immune target in colon cancer patients. Th-cell priming against EpCAM inevitably resulted in interleukin-4 (IL-4)-dominated Th2 responses, even under most stringent Th1-inducing conditions. These EpCAM-reactive Th2 cells rather promoted growth of EpCAM-expressing tumors. To analyze the role of IL-4 in tumor immune evasion, we generated EpCAM-reactive Th1 cells from IL-4.ko mice. These Th1 cells provided tumor-specific protection and established highly protective Th1 memory responses, even in naive BALB/c mice. Inhibition of tumor growth by Th1 cells resulted in intra-tumoral expression of cytokines of the IL-12 family and of IFN-gamma. Preventing activation-associated death of Th1 cells further increased intratumoral IFN-gamma expression and improved therapeutic efficacy. Thus, human TAA may promote tumor immune evasion by strongly favoring Th2 development.


Asunto(s)
Traslado Adoptivo/métodos , Antígenos de Neoplasias/inmunología , Moléculas de Adhesión Celular/inmunología , Neoplasias del Colon/inmunología , Neoplasias del Colon/terapia , Células Th2/citología , Animales , Antígenos de Neoplasias/genética , Vacunas contra el Cáncer/inmunología , Moléculas de Adhesión Celular/genética , Muerte Celular/inmunología , Diferenciación Celular/inmunología , División Celular/inmunología , Línea Celular Tumoral , Células Cultivadas , Neoplasias del Colon/patología , Molécula de Adhesión Celular Epitelial , Femenino , Regulación Neoplásica de la Expresión Génica/inmunología , Técnicas In Vitro , Interleucina-4/genética , Interleucina-4/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Trasplante de Neoplasias , Células TH1/citología , Células TH1/inmunología , Células TH1/metabolismo , Células Th2/inmunología , Células Th2/metabolismo
10.
Blood ; 114(8): 1696-706, 2009 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-19546478

RESUMEN

Signaling through tumor necrosis factor receptor 1 (TNFR1) controls bacterial infections and the induction of inflammatory Th1 cell-mediated autoimmune diseases. By dissecting Th1 cell-mediated delayed-type hypersensitivity responses (DTHRs) into single steps, we localized a central defect to the missing TNFR1 expression by endothelial cells (ECs). Adoptive transfer and mast cell knockin experiments into Kit(W)/Kit(W-v), TNF(-/-), and TNFR1(-/-) mice showed that the signaling defect exclusively affects mast cell-EC interactions but not T cells or antigen-presenting cells. As a consequence, TNFR1(-/-) mice had strongly reduced mRNA and protein expression of P-selectin, E-selectin, ICAM-1, and VCAM-1 during DTHR elicitation. In consequence, intravital fluorescence microscopy revealed up to 80% reduction of leukocyte rolling and firm adhesion in TNFR1(-/-) mice. As substitution of TNF(-/-) mice with TNF-producing mast cells fully restored DTHR in these mice, signaling of mast cell-derived TNF through TNFR1-expressing ECs is essential for the recruitment of leukocytes into sites of inflammation.


Asunto(s)
Endotelio Vascular/patología , Inflamación/etiología , Mastocitos/fisiología , Receptor Cross-Talk/fisiología , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Células Cultivadas , Endotelio Vascular/inmunología , Endotelio Vascular/metabolismo , Haptenos/efectos adversos , Hipersensibilidad Tardía/inducido químicamente , Hipersensibilidad Tardía/genética , Hipersensibilidad Tardía/inmunología , Inflamación/genética , Inflamación/metabolismo , Mastocitos/inmunología , Mastocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Cloruro de Picrilo/efectos adversos , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/fisiología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/fisiología
11.
Nat Commun ; 11(1): 1335, 2020 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-32165639

RESUMEN

Immune checkpoint blockade (ICB)-based or natural cancer immune responses largely eliminate tumours. Yet, they require additional mechanisms to arrest those cancer cells that are not rejected. Cytokine-induced senescence (CIS) can stably arrest cancer cells, suggesting that interferon-dependent induction of senescence-inducing cell cycle regulators is needed to control those cancer cells that escape from killing. Here we report in two different cancers sensitive to T cell-mediated rejection, that deletion of the senescence-inducing cell cycle regulators p16Ink4a/p19Arf (Cdkn2a) or p21Cip1 (Cdkn1a) in the tumour cells abrogates both the natural and the ICB-induced cancer immune control. Also in humans, melanoma metastases that progressed rapidly during ICB have losses of senescence-inducing genes and amplifications of senescence inhibitors. Metastatic cells also resist CIS. Such genetic and functional alterations are infrequent in metastatic melanomas regressing during ICB. Thus, activation of tumour-intrinsic, senescence-inducing cell cycle regulators is required to stably arrest cancer cells that escape from eradication.


Asunto(s)
Ciclo Celular , Senescencia Celular , Interferones/metabolismo , Melanoma/inmunología , Melanoma/patología , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Línea Celular Tumoral , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Humanos , Inmunoterapia , Antígeno Ki-67/metabolismo , Ganglios Linfáticos/patología , Melanoma/terapia , Melanoma/ultraestructura , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Transcripción STAT1/metabolismo , Análisis de Supervivencia , Carga Tumoral
12.
ChemMedChem ; 12(23): 1927-1930, 2017 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-29094797

RESUMEN

Autoimmune diseases are characterized by dendritic cell (DC)-driven activation of pro-inflammatory T cell responses. Therapeutic options for these severe diseases comprise small molecules such as dimethyl fumarate, or "gasotransmitters" such as CO. Herein we describe the synthesis of bifunctional enzyme-triggered CO-releasing molecules (ET-CORMs) that allow the simultaneous intracellular release of both CO and methyl fumarate. Using bone-marrow-derived DCs the impressive therapeutic potential of these methyl fumarate-derived compounds (FumET-CORMs) is demonstrated by strong inhibition of lipopolysaccharide-induced pro-inflammatory signaling pathways and blockade of downstream interleukin-12 or -23 production. The data also show that FumET-CORMs are able to transform DCs into an anti-inflammatory phenotype. Thus, these novel compounds have great clinical potential, for example, for the treatment of psoriasis or other inflammatory conditions of the skin.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Monóxido de Carbono/metabolismo , Esterasas/metabolismo , Ácido Fusárico/análogos & derivados , Inflamación/tratamiento farmacológico , Compuestos de Hierro Carbonilo/farmacología , Animales , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/metabolismo , Monóxido de Carbono/química , Cristalografía por Rayos X , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Esterasas/química , Ácido Fusárico/química , Ácido Fusárico/metabolismo , Ácido Fusárico/farmacología , Inflamación/metabolismo , Interleucina-12/antagonistas & inhibidores , Interleucina-12/biosíntesis , Interleucina-23/antagonistas & inhibidores , Interleucina-23/biosíntesis , Compuestos de Hierro Carbonilo/química , Compuestos de Hierro Carbonilo/metabolismo , Ratones , Modelos Moleculares , Estructura Molecular , Polisacáridos/antagonistas & inhibidores , Polisacáridos/farmacología
13.
J Nucl Med ; 46(1): 184-9, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15632051

RESUMEN

UNLABELLED: Radiolabeled cyclic peptides containing the amino acid sequence arginine-glycine-aspartate (RGD peptides) have successfully been used to image the expression of the alpha(v)beta(3) integrin in malignant tumors. However, the alpha(v)beta(3) integrin also plays an important role in angiogenesis induced by chronic inflammatory processes. Therefore, the aim of this study was to evaluate whether radiolabeled RGD peptides may also be used to assess alpha(v)beta(3) expression in inflammatory diseases. We studied a hapten-induced delayed-type hypersensitivity reaction (DTHR) as a model for inflammatory processes, since DTHRs are involved in many human autoimmune disorders. METHODS: The abdominal skin of mice was sensitized by application of 2,4,6-trinitrochlorobenzene (TNCB). One week later, a DTHR was elicited by challenging the right ear with TNCB. Application of TNCB was then repeated every 48 h to induce chronic skin inflammation. Small-animal PET and autoradiography with the alpha(v)beta(3) ligands (18)F-galacto-RGD and (125)I-gluco-RGD were performed at various times after TNCB application. The time course of tracer uptake by the treated ears was compared with histologic skin changes. RESULTS: The first challenge with TNCB caused, within 12 h, an acute inflammatory response with dense dermal infiltrates of polymorphonuclear leukocytes and lymphocytes. However, autoradiography revealed no significant increase in (125)I-gluco-RGD uptake at that time (mean uptake ratio for treated ear to untreated ear, 1.02 +/- 0.1 [SD]). Further challenges with TNCB resulted in chronic skin inflammation with markedly increased small-vessel density in the ear tissue. This was paralleled by a continuous increase in uptake of (125)I-gluco-RGD. After 13 challenges, the uptake ratio had increased to 2.30 +/- 0.27 (P < 0.005 compared with baseline). Enhanced uptake of radiolabeled RGD peptides in chronic inflammation was also demonstrated noninvasively by PET with (18)F-galacto-RGD. Pretreatment of the mice with nonradiolabeled cyclic peptide c(RGDfV) almost completely blocked uptake of (18)F-galacto-RGD by the challenged ear, thus confirming the specificity of tracer uptake. CONCLUSION: Radiolabeled RGD peptides allow a noninvasive assessment of alpha(v)beta(3) expression in inflammatory processes. PET with (18)F-galacto-RGD might become a powerful tool to distinguish between the acute and chronic phases of T cell-mediated immune responses and may represent a new biomarker for disease activity in autoimmune disorders.


Asunto(s)
Dermatitis por Contacto/diagnóstico por imagen , Dermatitis por Contacto/metabolismo , Galactosa/análogos & derivados , Galactosa/farmacocinética , Glucósidos/farmacocinética , Hipersensibilidad Tardía/diagnóstico por imagen , Hipersensibilidad Tardía/metabolismo , Oligopéptidos/farmacocinética , Péptidos Cíclicos/farmacocinética , Tomografía de Emisión de Positrones/métodos , Animales , Autorradiografía , Dermatitis por Contacto/complicaciones , Hipersensibilidad Tardía/inducido químicamente , Hipersensibilidad Tardía/complicaciones , Tasa de Depuración Metabólica , Ratones , Ratones Endogámicos C57BL , Cloruro de Picrilo , Radiofármacos/farmacocinética
14.
Oncoimmunology ; 4(7): e1014760, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26140238

RESUMEN

Stimulating the immune system to attack cancer is a promising approach, even for the control of advanced cancers. Several cytokines that promote interferon-γ-dominated immune responses show antitumor activity, with interleukin 12 (IL-12) being of major importance. Here, we used an antibody-IL-12 fusion protein (NHS-IL12) that binds histones of necrotic cells to treat human sarcoma in humanized mice. Following sarcoma engraftment, NHS-IL12 therapy was combined with either engineered IL-7 (FcIL-7) or IL-2 (IL-2MAB602) for continuous cytokine bioavailability. NHS-IL12 strongly induced innate and adaptive antitumor immunity when combined with IL-7 or IL-2. NHS-IL12 therapy significantly improved survival of sarcoma-bearing mice and caused long-term remissions when combined with IL-2. NHS-IL12 induced pronounced cancer cell senescence, as documented by strong expression of senescence-associated p16INK4a and nuclear translocation of p-HP1γ, and permanent arrest of cancer cell proliferation. In addition, this cancer immunotherapy initiated the induction of myogenic differentiation, further promoting the hypothesis that efficient antitumor immunity includes mechanisms different from cytotoxicity for efficient cancer control in vivo.

15.
J Nucl Med ; 55(2): 301-7, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24434289

RESUMEN

UNLABELLED: Although T cells can be labeled for noninvasive in vivo imaging, little is known about the impact of such labeling on T-cell function, and most imaging methods do not provide holistic information about trafficking kinetics, homing sites, or quantification. METHODS: We developed protocols that minimize the inhibitory effects of (64)Cu-pyruvaldehyde-bis(N4-methylthiosemicarbazone) ((64)Cu-PTSM) labeling on T-cell function and permit the homing patterns of T cells to be followed by PET. Thus, we labeled ovalbumin (OVA) T-cell receptor transgenic interferon (IFN)-γ-producing CD4(+) T (Th1) cells with 0.7-2.2 MBq of (64)Cu-PTSM and analyzed cell viability, IFN-γ production, proliferation, apoptosis, and DNA double-strand breaks and identified intracellular (64)Cu accumulation sites by energy dispersive x-ray analysis. To elucidate the fate of Th1 cell homing by PET, 10(7 64)Cu-OVA-Th1 cells were injected intraperitoneally or intravenously into healthy mice. To test the functional capacities of (64)Cu-OVA-Th1 cells during experimental OVA-induced airway hyperreactivity, we injected 10(7 64)Cu-OVA-Th1 cells intraperitoneally into OVA-immunized or nonimmunized healthy mice, which were challenged with OVA peptide or phosphate-buffered saline or remained untreated. In vivo PET investigations were followed by biodistribution, autoradiography, and fluorescence-activated cell sorting analysis. RESULTS: PET revealed unexpected homing patterns depending on the mode of T-cell administration. Within 20 min after intraperitoneal administration, (64)Cu-OVA-Th1 cells homed to the perithymic lymph nodes (LNs) of naive mice. Interestingly, intravenously administered (64)Cu-OVA-Th1 cells homed predominantly into the lung and spleen but not into the perithymic LNs. The accumulation of (64)Cu-OVA-Th1 cells in the pulmonary LNs (6.8 ± 1.1 percentage injected dose per cubic centimeter [%ID/cm(3)]) 24 h after injection was highest in the OVA-immunized and OVA-challenged OVA airway hyperreactivity-diseased littermates 24 h after intraperitoneal administration and lowest in the untreated littermates (3.7 ± 0.4 %ID/cm(3)). As expected, (64)Cu-OVA-Th1 cells also accumulated significantly in the pulmonary LNs of nonimmunized OVA-challenged animals (6.1 ± 0.5 %ID/cm(3)) when compared with phosphate-buffered saline-challenged animals (4.6 ± 0.5 %ID/cm(3)). CONCLUSION: Our protocol permits the detection of Th1 cells in single LNs and enables temporal in vivo monitoring of T-cell homing over 48 h. This work enables future applications for (64)Cu-PTSM-labeled T cells in clinical trials and novel therapy concepts focusing on T-cell-based immunotherapies of autoimmune diseases or cancer.


Asunto(s)
Tejido Linfoide/efectos de los fármacos , Tejido Linfoide/diagnóstico por imagen , Compuestos Organometálicos , Tomografía de Emisión de Positrones/métodos , Células TH1/citología , Tiosemicarbazonas , Animales , Apoptosis , Autoinmunidad , Movimiento Celular , Proliferación Celular , Separación Celular , Supervivencia Celular , Radioisótopos de Cobre , Roturas del ADN de Doble Cadena , Citometría de Flujo , Inmunoterapia/métodos , Interferón gamma/metabolismo , Ganglios Linfáticos/patología , Ratones , Péptidos/química , Factores de Tiempo , Distribución Tisular
16.
Cell Cycle ; 12(19): 3146-53, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24013429

RESUMEN

Data from different laboratories and theoretical considerations challenge our current view on anticancer immunity. Immune cells are capable of destroying cancer cells under in vitro and in vivo conditions. Therefore, cellular immunity is considered to control cancers through mechanisms that kill cancers. Yet, therapeutic anticancer immune responses rarely delete cancers. If efficient, they rather establish a life with stable disease. This raises the question of whether killing is the sole mechanism by which immune therapy attacks cancers. Here, we show that, besides cancer eradication by cytotoxic lymphocytes, other modes of action are operative and strictly required for cancer control. We show that T helper-1 cells arrest cancer growth by driving cancers into a state of stable or permanent growth arrest, called senescence. Such immune cells establish cytokine-producing walls around developing cancers. When producing interferon-γ and tumor necrosis factor, this cytokine-induced tumor immune-surveillance keeps the cancer cells in a permanently non-proliferating state. Simultaneously, antiangiogenic chemokines cut their connections to the surrounding tissues. This strategy significantly reduces tumor burden and prolongs life of cancer-bearing animals. As human cancers also undergo senescence, the current data suggest tumor-immune surveillance through cytokine-induced senescence, instead of tumor eradication, as the more realistic and primary goal of cancer control.


Asunto(s)
Neoplasias/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Antígenos de Neoplasias/metabolismo , Linfocitos T CD8-positivos/inmunología , Puntos de Control del Ciclo Celular , Senescencia Celular , Quimiocinas/metabolismo , Humanos , Inmunidad Celular , Inmunoterapia , Interferón gamma/metabolismo , Neoplasias/prevención & control , Neoplasias/terapia , Factor de Necrosis Tumoral alfa/metabolismo
17.
Mol Imaging Biol ; 15(2): 186-93, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22752651

RESUMEN

PURPOSE: Assessing information on tumor progression in the RIP1-Tag2 mouse in vivo is a great challenge because the tumors form spontaneously throughout the pancreas and are difficult to detect with current imaging modalities. In this study, we focused on non-invasive magnetic resonance imaging, providing information on tumor growth. PROCEDURES: Tissue relaxation times were measured over time and were compared between tumors and healthy pancreatic tissue. The effects of age and body temperature on these relaxation times, possibly influencing image contrast and therefore detection capabilities, were evaluated. RESULTS: Tumors appeared hyperintense in T2-weighted images when they exceeded 0.8 mm in diameter, and both relaxation times showed significantly higher values in tumors than in the healthy pancreas. CONCLUSION: Visualization and monitoring of these small tumors in vivo is feasible, even under adverse conditions of permanent gut movement. In the mouse model studied, the relaxation times of tumors differed significantly from healthy pancreatic tissue.


Asunto(s)
Imagen por Resonancia Magnética/métodos , Neoplasias Pancreáticas/diagnóstico , Animales , Temperatura Corporal , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Histocitoquímica , Ratones , Ratones Endogámicos C3H , Ratones Transgénicos , Neoplasias Pancreáticas/química , Neoplasias Pancreáticas/patología , Análisis de Regresión
18.
Diabetes ; 59(1): 119-27, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19826167

RESUMEN

OBJECTIVE: In vitro models suggest that free fatty acid-induced apoptotic beta-cell death is mediated through protein kinase C (PKC)delta. To examine the role of PKCdelta signaling in vivo, transgenic mice overexpressing a kinase-negative PKCdelta (PKCdeltaKN) selectively in beta-cells were generated and analyzed for glucose homeostasis and beta-cell survival. RESEARCH DESIGN AND METHODS: Mice were fed a standard or high-fat diet (HFD). Blood glucose and insulin levels were determined after glucose loads. Islet size, cleaved caspase-3, and PKCdelta expression were estimated by immunohistochemistry. In isolated islet cells apoptosis was assessed with TUNEL/TO-PRO3 DNA staining and the mitochondrial potential by rhodamine-123 staining. Changes in phosphorylation and subcellular distribution of forkhead box class O1 (FOXO1) were analyzed by Western blotting and immunohistochemistry. RESULTS: PKCdeltaKN mice were protected from HFD-induced glucose intolerance. This was accompanied by increased insulin levels in vivo, by an increased islet size, and by a reduced staining of beta-cells for cleaved caspase-3 compared with wild-type littermates. In accordance, long-term treatment with palmitate increased apoptotic cell death of isolated islet cells from wild-type but not from PKCdeltaKN mice. PKCdeltaKN overexpression protected islet cells from palmitate-induced mitochondrial dysfunction and inhibited nuclear accumulation of FOXO1 in mouse islet and INS-1E cells. The inhibition of nuclear accumulation of FOXO1 by PKCdeltaKN was accompanied by an increased phosphorylation of FOXO1 at Ser256 and a significant reduction of FOXO1 protein. CONCLUSIONS: Overexpression of PKCdeltaKN in beta-cells protects from HFD-induced beta-cell failure in vivo by a mechanism that involves inhibition of fatty acid-mediated apoptosis, inhibition of mitochondrial dysfunction, and inhibition of FOXO1 activation.


Asunto(s)
Intolerancia a la Glucosa/prevención & control , Células Secretoras de Insulina/enzimología , Células Secretoras de Insulina/fisiología , Proteína Quinasa C-delta/genética , Animales , Apoptosis , Glucemia/metabolismo , Técnicas de Cultivo de Célula , Muerte Celular , Dieta , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Regulación de la Expresión Génica , Insulina/análisis , Insulina/sangre , Insulina/genética , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Ratones , Ratones Noqueados , Ratones Transgénicos , Mitocondrias/efectos de los fármacos , Mitocondrias/fisiología , Mitocondrias/ultraestructura , Proteína Quinasa C-delta/deficiencia , Rodamina 123/farmacología
19.
Cell Cycle ; 7(19): 2974-7, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18838866

RESUMEN

Adoptive transfer of tumor antigen-specific T helper (Th) cells is a surprisingly potent anti-tumor therapy. Even in RIP1-Tag2 mice with a rapidly growing, aggressive endogenous beta cell tumor Th can significantly extend life time and are more efficient than any other therapy studied. The therapeutic effect of Th cells seems to be independent of tumor cell destruction. It critically relies on three principles: (1) inhibition of tumor angiogenesis, (2) inhibition of beta cell proliferation, and (3) induction of tumor dormancy. As tumor cell destruction by cytotoxic CD8(+) T cells (CTL) largely failed in tumor therapy, induction of tumor dormancy by Th cell-mediated immune responses represents a novel therapeutic option that may be combined with other cytotoxic regimens, e.g., radio- and/or chemotherapy, as it is established for bone marrow transplantation. Importantly, Th cell efficacy strictly requires interferon gamma (IFNgamma) signaling, and in the absence of IFNgamma, Th cells may even worsen tumor diseases. Therefore, using the immune system to control tumor dormancy represents a novel approach, especially as therapy of tumors resistant to conventional therapies. Yet, it is important to underline that Th cell-based antitumor effects critically depend on a functional cytokine network, especially appropriate IFNgamma signaling.


Asunto(s)
Neoplasias/inmunología , Linfocitos T/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Proteínas Activadoras de GTPasa/metabolismo , Humanos , Interferón gamma/inmunología , Ratones , Modelos Animales , Modelos Biológicos , Neoplasias/metabolismo , Fenotipo , Células TH1/inmunología
20.
Cancer Cell ; 13(6): 507-18, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18538734

RESUMEN

Immune responses may arrest tumor growth by inducing tumor dormancy. The mechanisms leading to either tumor dormancy or promotion of multistage carcinogenesis by adaptive immunity are poorly characterized. Analyzing T antigen (Tag)-induced multistage carcinogenesis in pancreatic islets, we show that Tag-specific CD4+ T cells home selectively into the tumor microenvironment around the islets, where they either arrest or promote transition of dysplastic islets into islet carcinomas. Through combined TNFR1 signaling and IFN-gamma signaling, Tag-specific CD4+ T cells induce antiangiogenic chemokines and prevent alpha(v)beta(3) integrin expression, tumor angiogenesis, tumor cell proliferation, and multistage carcinogenesis, without destroying Tag-expressing islet cells. In the absence of either TNFR1 signaling or IFN-gamma signaling, the same T cells paradoxically promote angiogenesis and multistage carcinogenesis. Thus, tumor-specific T cells can directly survey multistage carcinogenesis through cytokine signaling.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Proliferación Celular , Transformación Celular Neoplásica/inmunología , Insulinoma/inmunología , Interferón gamma/metabolismo , Neoplasias Pancreáticas/inmunología , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Transducción de Señal , Animales , Antígenos Virales de Tumores/genética , Antígenos Virales de Tumores/metabolismo , Glucemia/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/patología , Linfocitos T CD4-Positivos/trasplante , Movimiento Celular , Supervivencia Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Células Cultivadas , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Inmunoterapia/métodos , Insulinoma/irrigación sanguínea , Insulinoma/genética , Insulinoma/metabolismo , Insulinoma/patología , Insulinoma/terapia , Integrina alfaVbeta3/metabolismo , Ratones , Ratones Endogámicos C3H , Ratones Noqueados , Ratones Transgénicos , Neovascularización Patológica/inmunología , Neovascularización Patológica/patología , Neoplasias Pancreáticas/irrigación sanguínea , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/terapia , Receptores Tipo I de Factores de Necrosis Tumoral/deficiencia , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Células TH1/inmunología , Células TH1/patología , Factores de Tiempo , Irradiación Corporal Total
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA