Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Cancer Immunol Immunother ; 70(7): 2073-2086, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33439292

RESUMEN

Curaxins are small molecules that bind genomic DNA and interfere with DNA-histone interactions leading to the loss of histones and decondensation of chromatin. We named this phenomenon 'chromatin damage'. Curaxins demonstrated anti-cancer activity in multiple pre-clinical tumor models. Here, we present data which reveals, for the first time, a role for the immune system in the anti-cancer effects of curaxins. Using the lead curaxin, CBL0137, we observed elevated expression of several group of genes in CBL0137-treated tumor cells including interferon sensitive genes, MHC molecules, some embryo-specific antigens suggesting that CBL0137 increases tumor cell immunogenicity and improves recognition of tumor cells by the immune system. In support of this, we found that the anti-tumor activity of CBL0137 was reduced in immune deficient SCID mice when compared to immune competent mice. Anti-tumor activity of CBL0137 was abrogated in CD8+ T cell depleted mice but only partially lost when natural killer or CD4+ T cells were depleted. Further support for a key role for the immune system in the anti-tumor activity of CBL0137 is evidenced by an increased antigen-specific effector CD8+ T cell and NK cell response, and an increased ratio of effector T cells to Tregs in the tumor and spleen. CBL0137 also elevated the number of CXCR3-expressing CTLs in the tumor and the level of interferon-γ-inducible protein 10 (IP-10) in serum, suggesting IP-10/CXCR3 controls CBL0137-elicited recruitment of effector CTLs to tumors. Our collective data underscores a previously unrecognized role for both innate and adaptive immunity in the anti-tumor activity of curaxins.


Asunto(s)
Carbazoles/farmacología , Cromatina/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Inmunidad/inmunología , Animales , Apoptosis , Proliferación Celular , Quimiocinas/metabolismo , Cromatina/genética , Cromatina/metabolismo , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Citocinas/metabolismo , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Proc Natl Acad Sci U S A ; 113(7): E874-83, 2016 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-26831100

RESUMEN

Activation of an anticancer innate immune response is highly desirable because of its inherent ability to generate an adaptive antitumor T-cell response. However, insufficient safety of innate immune modulators limits clinical use to topical applications. Toll-like receptor 5 (TLR5) agonists are favorably positioned as potential systemic immunotherapeutic agents because of unusual tissue specificity of expression, uniquely safe profile of induced cytokines, and antitumor efficacy demonstrated in a number of animal models. Here, we decipher the molecular and cellular events underlying the metastasis suppressive activity of entolimod, a clinical stage TLR5 agonist that activates NF-κB-, AP-1-, and STAT3-driven immunomodulatory signaling pathways specifically within the liver. Used as a single agent in murine colon and mammary metastatic cancer models, entolimod rapidly induces CXCL9 and -10 that support homing of blood-borne CXCR3-expressing NK cells to the liver predominantly through an IFN-γ signaling independent mechanism. NK cell-dependent activation of dendritic cells is followed by stimulation of a CD8(+) T-cell response, which exert both antimetastatic effect of entolimod and establishment of tumor-specific and durable immune memory. These results define systemically administered TLR5 agonists as organ-specific immunoadjuvants, enabling efficient antitumor vaccination that does not depend on identification of tumor-specific antigens.


Asunto(s)
Linfocitos T CD8-positivos/efectos de los fármacos , Células Asesinas Naturales/efectos de los fármacos , Metástasis de la Neoplasia/prevención & control , Péptidos/farmacología , Receptor Toll-Like 5/agonistas , Animales , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Células Asesinas Naturales/inmunología , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
3.
Proc Natl Acad Sci U S A ; 110(20): E1857-66, 2013 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-23630282

RESUMEN

Vertebrate Toll-like receptor 5 (TLR5) recognizes bacterial flagellin proteins and activates innate immune responses to motile bacteria. In addition, activation of TLR5 signaling can inhibit growth of TLR5-expressing tumors and protect normal tissues from radiation and ischemia-reperfusion injuries. To understand the mechanisms behind these phenomena at the organismal level, we assessed nuclear factor kappa B (NF-κB) activation (indicative of TLR5 signaling) in tissues and cells of mice treated with CBLB502, a pharmacologically optimized flagellin derivative. This identified the liver and gastrointestinal tract as primary CBLB502 target organs. In particular, liver hepatocytes were the main cell type directly and specifically responding to systemic administration of CBLB502 but not to that of the TLR4 agonist LPS. To assess CBLB502 impact on other pathways, we created multireporter mice with hepatocytes transduced in vivo with reporters for 46 inducible transcription factor families and found that along with NF-κB, CBLB502 strongly activated STAT3-, phenobarbital-responsive enhancer module (PREM), and activator protein 1 (AP-1-) -driven pathways. Livers of CBLB502-treated mice displayed induction of numerous immunomodulatory factors and massive recruitment of various types of immune cells. This led to inhibition of growth of liver metastases of multiple tumors regardless of their TLR5 status. The changed liver microenvironment was not, however, hepatotoxic, because CBLB502 induced resistance to Fas-mediated apoptosis in normal liver cells. Temporary occlusion of liver blood circulation prevented CBLB502 from protecting hematopoietic progenitors in lethally irradiated mice, indicating involvement of a factor secreted by responding liver cells. These results define the liver as the key mediator of TLR5-dependent effects in vivo and suggest clinical applications for TLR5 agonists as hepatoprotective and antimetastatic agents.


Asunto(s)
Hígado/metabolismo , Péptidos/farmacología , Factor de Transcripción STAT3/metabolismo , Receptor Toll-Like 5/agonistas , Animales , Anticarcinógenos/farmacología , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Neoplasias del Colon/metabolismo , Femenino , Citometría de Flujo , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Células Asesinas Naturales/metabolismo , Hígado/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Trasplante de Neoplasias , Neutrófilos/metabolismo , Protectores contra Radiación/farmacología , Transducción de Señal , Receptor fas/metabolismo
4.
Proc Natl Acad Sci U S A ; 109(33): 13314-8, 2012 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-22847439

RESUMEN

Unlike reversible quiescence, cellular senescence is characterized by a large flat cell morphology, ß-gal staining and irreversible loss of regenerative (i.e., replicative) potential. Conversion from proliferative arrest to irreversible senescence, a process named geroconversion, is driven in part by growth-promoting pathways such as mammalian target of rapamycin (mTOR). During cell cycle arrest, mTOR converts reversible arrest into senescence. Inhibitors of mTOR can suppress geroconversion, maintaining quiescence instead. It was shown that hypoxia inhibits mTOR. Therefore, we suggest that hypoxia may suppress geroconversion. Here we tested this hypothesis. In HT-p21-9 cells, expression of inducible p21 caused cell cycle arrest without inhibiting mTOR, leading to senescence. Hypoxia did not prevent p21 induction and proliferative arrest, but instead inhibited the mTOR pathway and geroconversion. Exposure to hypoxia during p21 induction prevented senescent morphology and loss of regenerative potential, thus maintaining reversible quiescence so cells could restart proliferation after switching p21 off. Suppression of geroconversion was p53- and HIF-1-independent, as hypoxia also suppressed geroconversion in cells lacking functional p53 and HIF-1α. Also, in normal fibroblasts and retinal cells, hypoxia inhibited the mTOR pathway and suppressed senescence caused by etoposide without affecting DNA damage response, p53/p21 induction and cell cycle arrest. Also hypoxia suppressed geroconversion in cells treated with nutlin-3a, a nongenotoxic inducer of p53, in cell lines susceptible to nutlin-3a-induced senescence (MEL-10, A172, and NKE). Thus, in normal and cancer cell lines, hypoxia suppresses geroconversion caused by diverse stimuli. Physiological and clinical implications of the present findings are discussed.


Asunto(s)
Senescencia Celular , Células Epiteliales/citología , Fibroblastos/citología , Hipoxia de la Célula/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Etopósido/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Imidazoles/farmacología , Piperazinas/farmacología , Proteína p53 Supresora de Tumor/metabolismo
5.
J Immunol ; 189(10): 4719-27, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23045613

RESUMEN

Allogeneic hematopoietic cell transplantation is an established treatment for hematologic and nonhematologic malignancies. Donor-derived immune cells can identify and attack host tumor cells, producing a graft-versus-tumor (GVT) effect that is crucial to the effectiveness of the transplantation therapy. CBLB502 is a novel agonist for TLR5 derived from Salmonella flagellin. On the basis of TLR5-mediated immunomodulatory function, we examined the effect of CBLB502 on GVT activity. Using two tumor models that do not express TLR5, and thereby do not directly respond to CBLB502, we found that CBLB502 treatment significantly enhanced allogeneic CD8(+) T cell-mediated GVT activity, which was evidenced by decreased tumor burden and improved host survival. Importantly, histopathologic analyses showed that CBLB502 treatment did not exacerbate the moderate graft-versus-host disease condition caused by the allogeneic CD8(+) T cells. Moreover, mechanistic analyses showed that CBLB502 stimulates CD8(+) T cell proliferation and enhances their tumor killing activity mainly indirectly through a mechanism that involves the IL-12 signaling pathway and the CD11c(+) and CD11b(+) populations in the bone marrow cells. This study demonstrates a new beneficial effect of CBLB502, and suggests that TLR5-mediated immune modulation may be a promising approach to improve GVT immunity without exacerbating graft-versus-host disease.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Flagelina/farmacología , Efecto Injerto vs Tumor/efectos de los fármacos , Trasplante de Células Madre Hematopoyéticas , Inmunidad Celular/efectos de los fármacos , Neoplasias Experimentales/terapia , Salmonella/química , Receptor Toll-Like 5/agonistas , Animales , Linfocitos T CD8-positivos/patología , Proliferación Celular/efectos de los fármacos , Flagelina/química , Flagelina/inmunología , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Efecto Injerto vs Tumor/inmunología , Inmunidad Celular/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos DBA , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Receptor Toll-Like 5/inmunología , Trasplante Homólogo
6.
Infect Immun ; 81(10): 3855-64, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23897616

RESUMEN

Pathogen recognition receptors (PRRs) are essential components of host innate immune systems that detect specific conserved pathogen-associated molecular patterns (PAMPs) presented by microorganisms. Members of two families of PRRs, transmembrane Toll-like receptors (TLRs 1, 2, 4, 5, and 6) and cytosolic NOD receptors (NOD1 and NOD2), are stimulated upon recognition of various bacterial PAMPs. Such stimulation leads to induction of a number of immune defense reactions, mainly triggered via activation of the transcription factor NF-κB. While coordination of responses initiated via different PRRs sensing multiple PAMPS present during an infection makes clear biological sense for the host, such interactions have not been fully characterized. Here, we demonstrate that combined stimulation of NOD1 and TLR5 (as well as other NOD and TLR family members) strongly potentiates activity of NF-κB and induces enhanced levels of innate immune reactions (e.g., cytokine production) both in vitro and in vivo. Moreover, we show that an increased level of NF-κB activity plays a critical role in formation of downstream responses. In live mice, synergy between these receptors resulting in potentiation of NF-κB activity was organ specific, being most prominent in the gastrointestinal tract. Coordinated activity of NOD1 and TLR5 significantly increased protection of mice against enteroinvasive Salmonella infection. Obtained results suggest that cooperation of NOD and TLR receptors is important for effective responses to microbial infection in vivo.


Asunto(s)
Inmunidad Innata/fisiología , FN-kappa B/metabolismo , Proteína Adaptadora de Señalización NOD1/metabolismo , Salmonella typhimurium , Receptor Toll-Like 5/metabolismo , Animales , Femenino , Regulación de la Expresión Génica/inmunología , Ratones , Ratones Endogámicos BALB C , FN-kappa B/genética , Proteína Adaptadora de Señalización NOD1/genética , Salmonelosis Animal/inmunología , Salmonelosis Animal/microbiología , Receptor Toll-Like 5/genética
7.
Nat Med ; 10(1): 48-54, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14702634

RESUMEN

Although tumor progression involves processes such as tissue invasion that can activate inflammatory responses, the immune system largely ignores or tolerates disseminated cancers. The mechanisms that block initiation of immune responses during cancer development are poorly understood. We report here that constitutive activation of Stat-3, a common oncogenic signaling pathway, suppresses tumor expression of proinflammatory mediators. Blocking Stat-3 in tumor cells increases expression of proinflammatory cytokines and chemokines that activate innate immunity and dendritic cells, leading to tumor-specific T-cell responses. In addition, constitutive Stat-3 activity induces production of pleiotropic factors that inhibit dendritic cell functional maturation. Tumor-derived factors inhibit dendritic cell maturation through Stat-3 activation in progenitor cells. Thus, inhibition of antitumor immunity involves a cascade of Stat-3 activation propagating from tumor to dendritic cells. We propose that tumor Stat-3 activity can mediate immune evasion by blocking both the production and sensing of inflammatory signals by multiple components of the immune system.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Inmunidad Innata/fisiología , Melanoma Experimental/inmunología , Transducción de Señal/fisiología , Transactivadores/fisiología , Animales , Línea Celular Tumoral , Células Dendríticas/inmunología , Ensayo de Cambio de Movilidad Electroforética , Ensayo de Inmunoadsorción Enzimática , Melanoma Experimental/metabolismo , Ratones , Factor de Transcripción STAT3
8.
Cell Death Discov ; 7(1): 266, 2021 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-34584068

RESUMEN

Acute radiation syndrome (ARS) is a major cause of lethality following radiation disasters. A TLR5 agonist, entolimod, is among the most powerful experimental radiation countermeasures and shows efficacy in rodents and non-human primates as a prophylactic (radioprotection) and treatment (radiomitigation) modality. While the prophylactic activity of entolimod has been connected to the suppression of radiation-induced apoptosis, the mechanism by which entolimod functions as a radiomitigator remains poorly understood. Uncovering this mechanism has significant and broad-reaching implications for the clinical development and improvement of TLR5 agonists for use as an effective radiation countermeasure in scenarios of mass casualty resulting from accidental exposure to ionizing radiation. Here, we demonstrate that in contrast to radioprotection, neutrophils are essential for the radiomitigative activity of entolimod in a mouse model of lethal ARS. Neutrophils express functional TLR5 and rapidly exit the bone marrow (BM), accumulate in solid tissues, and release MMP-9 following TLR5 stimulation which is accompanied by an increase in the number of active hematopoietic pluripotent precursors (HPPs) in the BM. Importantly, recombinant MMP-9 by itself has radiomitigative activity and, in the absence of neutrophils, accelerates the recovery of the hematopoietic system. Unveiling this novel TLR5-neutrophil-MMP-9 axis of radiomitigation opens new opportunities for the development of efficacious radiation countermeasures to treat ARS following accidental radiation disasters.

9.
Free Radic Biol Med ; 172: 136-151, 2021 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-34097996

RESUMEN

Prostate cancer (PCa) is a major cause of mortality and morbidity in men. Available therapies yield limited outcome. We explored anti-PCa activity in a polyphenol-rich fraction of Bergenia ligulata (PFBL), a plant used in Indian traditional and folk medicine for its anti-inflammatory and antineoplastic properties. PFBL constituted of about fifteen different compounds as per LCMS analysis induced apoptotic death in both androgen-dependent LNCaP and androgen-refractory PC3 and DU145 cells with little effect on NKE and WI38 cells. Further investigation revealed that PFBL mediates its function through upregulating ROS production by enhanced catalytic activity of Monoamine oxidase A (MAO-A). Notably, the differential inactivation of NRF2-antioxidant response pathway by PFBL resulted in death in PC3 versus NKE cells involving GSK-3ß activity facilitated by AKT inhibition. PFBL efficiently reduced the PC3-tumor xenograft in NOD-SCID mice alone and in synergy with Paclitaxel. Tumor tissues in PFBL-treated mice showed upregulation of similar mechanism of cell death as observed in isolated PC3 cells i.e., elevation of MAO-A catalytic activity, ROS production accompanied by activation of ß-TrCP-GSK-3ß axis of NRF2 degradation. Blood counts, liver, and splenocyte sensitivity analyses justified the PFBL safety in the healthy mice. To our knowledge this is the first report of an activity that crippled NRF2 activation both in vitro and in vivo in response to MAO-A activation. Results of this study suggest the development of a novel treatment protocol utilizing PFBL to improve therapeutic outcome for patients with aggressive PCa which claims hundreds of thousands of lives each year.


Asunto(s)
Antioxidantes , Neoplasias de la Próstata , Animales , Glucógeno Sintasa Quinasa 3 beta , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Monoaminooxidasa , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Polifenoles/farmacología , Neoplasias de la Próstata/tratamiento farmacológico
10.
Commun Biol ; 4(1): 466, 2021 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-33846531

RESUMEN

The Toll-like receptor 5 (TLR5) agonist entolimod, a derivative of Salmonella flagellin, has therapeutic potential for several indications including radioprotection and cancer immunotherapy. However, in Phase 1 human studies, entolimod induced a rapid neutralizing immune response, presumably due to immune memory from prior exposure to flagellated enterobacteria. To enable multi-dose applications, we used structure-guided reengineering to develop a next-generation, substantially deimmunized entolimod variant, GP532. GP532 induces TLR5-dependent NF-κB activation like entolimod but is smaller and has mutations eliminating an inflammasome-activating domain and key B- and T-cell epitopes. GP532 is resistant to human entolimod-neutralizing antibodies and shows reduced de novo immunogenicity. GP532 also has improved bioavailability, a stronger effect on key cytokine biomarkers, and a longer-lasting effect on NF-κB. Like entolimod, GP532 demonstrated potent prophylactic and therapeutic efficacy in mouse models of radiation-induced death and tissue damage. These results establish GP532 as an optimized TLR5 agonist suitable for multi-dose therapies and for patients with high titers of preexisting flagellin-neutralizing antibodies.


Asunto(s)
Péptidos/farmacología , Transducción de Señal , Receptor Toll-Like 5/agonistas , Línea Celular Tumoral , Células HEK293 , Humanos
11.
PLoS One ; 15(2): e0227940, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32027657

RESUMEN

Tumor necrosis factor alpha (TNF) is capable of inducing regression of solid tumors. However, TNF released in response to Toll-like receptor 4 (TLR4) activation by bacterial lipopolysaccharide (LPS) is the key mediator of cytokine storm and septic shock that can cause severe tissue damage limiting anticancer applications of this cytokine. In our previous studies, we demonstrated that activation of another Toll-like receptor, TLR5, could protect from tissue damage caused by a variety of stresses including radiation, chemotherapy, Fas-activating antibody and ischemia-reperfusion. In this study, we tested whether entolimod could counteract TNF-induced toxicity in mouse models. We found that entolimod pretreatment effectively protects livers and lungs from LPS- and TNF-induced toxicity and prevents mortality caused by combining either of these agents with the sensitizer, D-galactosamine. While LPS and TNF induced significant activation of apoptotic caspase 3/7, lipid tissue peroxidation and serum ALT accumulation in mice without entolimod treatment, these indicators of toxicity were reduced by entolimod pretreatment to the levels of untreated control mice. Entolimod was effective when injected 0.5-48 hours prior to, but not when injected simultaneously or after LPS or TNF. Using chimeric mice with hematopoiesis differing in its TLR5 status from the rest of tissues, we showed that this protective activity was dependent on TLR5 expression by non-hematopoietic cells. Gene expression analysis identified multiple genes upregulated by entolimod in the liver and cultured hepatocytes as possible mediators of its protective activity. Entolimod did not interfere with the antitumor activity of TNF in mouse hepatocellular and colorectal tumor models. These results support further development of TLR5 agonists to increase tissue resistance to cytotoxic cytokines, reduce the risk of septic shock and enable safe systemic application of TNF as an anticancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Péptidos/farmacología , Receptor Toll-Like 5/agonistas , Factor de Necrosis Tumoral alfa/toxicidad , Animales , Línea Celular Tumoral , Células Cultivadas , Galactosamina , Hematopoyesis/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Lipopolisacáridos/toxicidad , Hígado/efectos de los fármacos , Hígado/patología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Modelos Biológicos , FN-kappa B/metabolismo , Sustancias Protectoras/farmacología , Análisis de Supervivencia , Receptor Toll-Like 5/metabolismo , Factor de Necrosis Tumoral alfa/sangre , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
12.
Radiat Res ; 187(5): 570-580, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28323577

RESUMEN

Radiation treatment of head and neck cancer frequently causes severe collateral damage to normal tissues including mouth mucosa, salivary glands and skin. This toxicity limits the radiation dose that can be delivered and affects the patient's quality of life. Previous studies in mice and nonhuman primates showed that entolimod, a toll-like receptor 5 (TLR5) agonist derived from bacterial flagellin, effectively reduced radiation damage to hematopoietic and gastrointestinal tissues in both total-body and local irradiation scenarios, with no protection of tumors. Here, using a mouse model, we analyzed the efficacy of entolimod administered before or after irradiation in reducing damage to normal tissues. Animals received local fractionated radiation to the head and neck area, thus modeling radiotherapy of head and neck cancer. Tissue damage was evaluated through histomorphological examination of samples collected at different time points up to four weeks, mice were exposed locally to five daily fractions of 5, 6 or 7 Gy. A semiquantitative scoring system was used to assess the severity of observed pathomorphological changes. In this model, radiation damage was most severe in the lips, tongue and skin, moderate in the upper esophagus and minor in salivary glands. The kinetics of injury appearance and recovery of normal morphology varied among tissues, with maximal damage to the tongue, esophagus and salivary glands developing at earlier times (days 8-11 postirradiation) relative to that of lip and skin mucosa (days 11-15 postirradiation). While both tested regimens of entolimod significantly reduced the extent of radiation damage and accelerated restoration of normal structure in all tissues analyzed, administration of entolimod 1 h after each irradiation was more effective than treatment 30 min before irradiation. These results support the potential clinical use of entolimod as an adjuvant for improving the therapeutic index of head and neck cancer radiotherapy by reducing the radiation toxicity in normal tissues.


Asunto(s)
Epitelio/lesiones , Epitelio/patología , Neoplasias de Cabeza y Cuello/radioterapia , Péptidos/administración & dosificación , Traumatismos por Radiación/patología , Traumatismos por Radiación/prevención & control , Animales , Fraccionamiento de la Dosis de Radiación , Relación Dosis-Respuesta a Droga , Epitelio/efectos de la radiación , Femenino , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Ratones , Protectores contra Radiación/administración & dosificación , Receptor Toll-Like 5/antagonistas & inhibidores , Resultado del Tratamiento
13.
Oncotarget ; 7(3): 2936-50, 2016 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-26655090

RESUMEN

Uveal melanoma (UM) is the most common primary cancer of the eye in adults and progresses to metastatic disease predominantly of the liver in ~50% of patients. In these cases, life expectancy averages just 9 months due to the lack of effective treatment options. The Toll-like receptor 5 (TLR5) agonist entolimod (former name CBLB502) rapidly activates TLR5-NF-κB signaling in hepatocytes and suppresses growth of both TLR5-expressing and non-expressing tumors in the liver through mobilization and activation of innate and adaptive immune mechanisms. The goal of this study was to explore the potential of entolimod as an immunotherapeutic agent against hepatic metastasis of UM using the TLR5-positive B16LS9 mouse model of ocular melanoma. Mice were given seven subcutaneous injections of vehicle or entolimod given 72 h apart started one day before, on the same day or three days after intraocular injection of B16LS9 cells. All tested regimens of entolimod treatment resulted in significantly reduced B16LS9 metastasis to the liver. Entolimod induced mobilization of natural killer (NK) cells to the liver and stimulated their maturation, differentiation and activation. Antibody-mediated depletion of NK cells from mice abrogated entolimod's antimetastatic activity in the liver and eliminated the entolimod-elicited in vitro cytotoxic activity of hepatic lymphocytes against B16LS9 cells. These results provide pre-clinical evidence of entolimod's efficacy against hepatometastasis of UM and support its further development as an anticancer immunotherapeutic drug.


Asunto(s)
Antineoplásicos/uso terapéutico , Células Asesinas Naturales/inmunología , Neoplasias Hepáticas , Melanoma Experimental/patología , Melanoma/patología , Péptidos/uso terapéutico , Neoplasias de la Úvea/patología , Inmunidad Adaptativa/efectos de los fármacos , Inmunidad Adaptativa/inmunología , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Hepatocitos/metabolismo , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/inmunología , Inmunoterapia/métodos , Células Asesinas Naturales/efectos de los fármacos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/secundario , Ratones , Ratones Endogámicos C57BL , Receptor Toll-Like 5/agonistas , Factor de Transcripción ReIA/metabolismo
14.
Mol Cancer Ther ; 1(11): 893-9, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12481410

RESUMEN

Constitutive activation of Janus kinases (JAKs) and signal transducers and activators of transcription (STAT) occurs at very high frequency in various hematopoietic malignancies and solid tumors. It has been demonstrated that the tyrosine kinase inhibitor, AG-490, selectively blocks JAK activity and completely eliminates leukemia cells in a severe combined immunodeficient (SCID) mouse model. Because many cytokines, including interleukin (IL)-12, have been shown to signal through JAK/STAT pathways, AG-490 may inhibit cytokine-based cancer therapy. In this study, we evaluated the effects of AG-490 on IL-12 functional signaling and IL-12-mediated antitumor response in vivo. Previous studies have established the critical roles of macrophages and IFN-gamma in mediating IL-12-induced antitumor effects. Our results show that in vivo administration of AG-490 causes tumor cell apoptosis but does not inhibit IL-12-mediated macrophage activation and IFN-gamma production by lymphocytes. Furthermore, our data indicate that combined therapy with AG-490 and IL-12-induces greater antitumor effects than either agent alone in a murine myeloma tumor model. These results suggest that JAK/STAT inhibitors deserve further investigation for use with IL-12 therapy in treating human cancers with elevated JAK/STAT activity.


Asunto(s)
Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica , Interleucina-12/farmacología , Neoplasias/tratamiento farmacológico , Tirfostinos/farmacología , Animales , Apoptosis , Núcleo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Femenino , Humanos , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Interferón gamma/metabolismo , Interleucina-12/metabolismo , Macrófagos/metabolismo , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Factor de Transcripción STAT3 , Transducción de Señal , Bazo/citología , Factores de Tiempo , Transactivadores/metabolismo , Células Tumorales Cultivadas
15.
PLoS One ; 9(1): e85587, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24454895

RESUMEN

Toll-like receptor (TLR) mediated recognition of pathogen associated molecular patterns allows the immune system to rapidly respond to a pathogenic insult. The "danger context" elicited by TLR agonists allows an initially non-immunogenic antigen to become immunogenic. This ability to alter environment is highly relevant in tumor immunity, since it is inherently difficult for the immune system to recognize host-derived tumors as immunogenic. However, immune cells may have encountered certain TLR ligands associated with tumor development, yet the endogenous stimulation is typically not sufficient to induce spontaneous tumor rejection. Of special interest are TLR5 agonists, because there are no endogenous ligands that bind TLR5. CBLB502 is a pharmacologically optimized TLR5 agonist derived from Salmonella enterica flagellin. We examined the effect of CBLB502 on tumor immunity using two syngeneic lymphoma models, both of which do not express TLR5, and thus do not directly respond to CBLB502. Upon challenge with the T-cell lymphoma RMAS, CBLB502 treatment after tumor inoculation protects C57BL/6 mice from death caused by tumor growth. This protective effect is both natural killer (NK) cell- and perforin-dependent. In addition, CBLB502 stimulates clearance of the B-cell lymphoma A20 in BALB/c mice in a CD8(+) T cell-dependent fashion. Analysis on the cellular level via ImageStream flow cytometry reveals that CD11b(+) and CD11c(+) cells, but neither NK nor T cells, directly respond to CBLB502 as determined by NFκB nuclear translocation. Our findings demonstrate that CBLB502 stimulates a robust antitumor response by directly activating TLR5-expressing accessory immune cells, which in turn activate cytotoxic lymphocytes.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Flagelina/química , Linfocitos/efectos de los fármacos , Linfocitos/inmunología , Linfoma de Células T/inmunología , Péptidos/farmacología , Receptor Toll-Like 5/agonistas , Inmunidad Adaptativa/efectos de los fármacos , Adyuvantes Inmunológicos/química , Adyuvantes Inmunológicos/uso terapéutico , Animales , Línea Celular Tumoral , Citocinas/sangre , Inmunidad Innata/efectos de los fármacos , Linfoma de Células T/sangre , Linfoma de Células T/tratamiento farmacológico , Ratones , Péptidos/química , Péptidos/uso terapéutico , Perforina/metabolismo , Regulación hacia Arriba/efectos de los fármacos
16.
Oncotarget ; 5(3): 802-14, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24583651

RESUMEN

Myelosuppression and gastrointestinal damage are common side effects of cancer treatment limiting efficacy of DNA-damaging chemotherapeutic drugs. The Toll-like receptor 5 (TLR5) agonist Entolimod has demonstrated efficacy in mitigating damage to hematopoietic and gastrointestinal tissues caused by radiation. Here, using 5-Fluorouracil (5-FU) treated mice as a model of chemotherapy-induced side effects, we demonstrated significant reduction in the severity of 5-FU-induced morbidity and increased survival accompanied by the improved integrity of intestinal tissue and stimulated the restoration of hematopoiesis. Entolimod-stimulated IL-6 production was essential for Entolimod's ability to rescue mice from death caused by doses of 5-FU associated with hematopoietic failure. In contrast, IL-6 induction was not necessary for protection and restoration of drug-damaged gastrointestinal tissue by Entolimod. In a syngeneic mouse CT26 colon adenocarcinoma model, Entolimod reduced the systemic toxicity of 5-FU, but did not reduce its antitumor efficacy indicating that the protective effect of Entolimod was selective for normal, non-tumor, tissues. These results suggest that Entolimod has clinical potential to broaden the therapeutic window of genotoxic anticancer drugs by reducing their associated hematopoietic and gastrointestinal toxicities.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias del Colon/tratamiento farmacológico , Fluorouracilo/farmacología , Péptidos/farmacología , Receptor Toll-Like 5/agonistas , Adenocarcinoma/tratamiento farmacológico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Sinergismo Farmacológico , Femenino , Fluorouracilo/administración & dosificación , Fluorouracilo/efectos adversos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Distribución Aleatoria
17.
Int J Radiat Oncol Biol Phys ; 83(1): 228-34, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22000579

RESUMEN

PURPOSE: Development of mucositis is a frequent side effect of radiotherapy of patients with head-and-neck cancer. We have recently reported that bacterial flagellin, an agonist of Toll-like receptor 5 (TLR5), can protect rodents and primates from acute radiation syndrome caused by total body irradiation. Here we analyzed the radioprotective efficacy of TLR5 agonist under conditions of local, single dose or fractionated radiation treatment. METHODS AND MATERIALS: Mice received either single-dose (10, 15, 20, or 25 Gy) or fractioned irradiation (cumulative dose up to 30 Gy) of the head-and-neck area with or without subcutaneous injection of pharmacologically optimized flagellin, CBLB502, 30 min before irradiation. RESULTS: CBLB502 significantly reduced the severity of dermatitis and mucositis, accelerated tissue recovery, and reduced the extent of radiation induced weight loss in mice after a single dose of 15 or 20 Gy but not 25 Gy of radiation. CBLB502 was also protective from cumulative doses of 25 and 30 Gy delivered in two (10 + 15 Gy) or three (3 × 10 Gy) fractions, respectively. While providing protection to normal epithelia, CBLB502 did not affect the radiosensitivity of syngeneic squamous carcinoma SCCVII grown orthotopically in mice. Use of CBLB502 also elicited a radiation independent growth inhibitory effect upon TLR5-expressing tumors demonstrated in the mouse xenograft model of human lung adenocarcinoma A549. CONCLUSION: CBLB502 combines properties of supportive care (radiotherapy adjuvant) and anticancer agent, both mediated via activation of TLR5 signaling in the normal tissues or the tumor, respectively.


Asunto(s)
Neoplasias de Cabeza y Cuello/radioterapia , Péptidos/uso terapéutico , Traumatismos Experimentales por Radiación/prevención & control , Protectores contra Radiación/uso terapéutico , Radiodermatitis/prevención & control , Estomatitis/prevención & control , Receptor Toll-Like 5/agonistas , Adenocarcinoma/metabolismo , Animales , Carcinoma de Células Escamosas/radioterapia , Línea Celular Tumoral , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Ratones , Mucosa Bucal/efectos de la radiación , Tolerancia a Radiación/efectos de los fármacos , Estomatitis/etiología , Receptor Toll-Like 5/metabolismo , Pérdida de Peso/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
18.
Science ; 320(5873): 226-30, 2008 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-18403709

RESUMEN

The toxicity of ionizing radiation is associated with massive apoptosis in radiosensitive organs. Here, we investigate whether a drug that activates a signaling mechanism used by tumor cells to suppress apoptosis can protect healthy cells from the harmful effects of radiation. We studied CBLB502, a polypeptide drug derived from Salmonella flagellin that binds to Toll-like receptor 5 (TLR5) and activates nuclear factor-kappaB signaling. A single injection of CBLB502 before lethal total-body irradiation protected mice from both gastrointestinal and hematopoietic acute radiation syndromes and resulted in improved survival. CBLB502 injected after irradiation also enhanced survival, but at lower radiation doses. It is noteworthy that the drug did not decrease tumor radiosensitivity in mouse models. CBLB502 also showed radioprotective activity in lethally irradiated rhesus monkeys. Thus, TLR5 agonists could potentially improve the therapeutic index of cancer radiotherapy and serve as biological protectants in radiation emergencies.


Asunto(s)
FN-kappa B/metabolismo , Péptidos/farmacología , Traumatismos Experimentales por Radiación/prevención & control , Tolerancia a Radiación/efectos de los fármacos , Protectores contra Radiación/farmacología , Receptor Toll-Like 5/agonistas , Secuencia de Aminoácidos , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Quimioterapia Adyuvante , Flagelina/química , Flagelina/farmacología , Rayos gamma , Sistema Hematopoyético/efectos de los fármacos , Sistema Hematopoyético/efectos de la radiación , Intestino Delgado/citología , Intestino Delgado/efectos de los fármacos , Intestino Delgado/efectos de la radiación , Macaca mulatta , Ratones , Ratones Endogámicos ICR , Datos de Secuencia Molecular , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/radioterapia , Péptidos/administración & dosificación , Péptidos/química , Péptidos/toxicidad , Dosis de Radiación , Protectores contra Radiación/administración & dosificación , Protectores contra Radiación/química , Protectores contra Radiación/toxicidad , Salmonella enterica , Transducción de Señal , Receptor Toll-Like 5/metabolismo , Irradiación Corporal Total
19.
Cancer Res ; 66(19): 9356-61, 2006 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17018587

RESUMEN

Inactivation of p53 function, which frequently occurs in tumors, can significantly modulate tumor cell sensitivity to radiation and chemotherapeutic drugs. However, in addition to acting on malignant cells, anticancer agents act on the cells of tumor stroma, causing activation of a p53 response. The effect of this response on treatment outcome has been the subject of the present study. Tumors with p53-deficient stroma were generated using mouse tumorigenic packaging cells that produce a p53 inhibitory retrovirus, encoding a dominant-negative p53 mutant. Tumors maintaining wild-type p53 in their stroma were formed by cells of similar origin but deficient in retroviral production due to the deletion of the packaging signal in the retroviral vector. Comparison of these tumor models, differing only in p53 status of their stromas, showed that tumors with p53-deficient stroma were significantly more sensitive to experimental chemotherapy and radiotherapy. A similar effect was achieved when anticancer treatment was combined with pharmacologic suppression of p53 by the cyclic form of pifithrin alpha, a small-molecule inhibitor of p53. Potentiation of the anticancer effect of chemotherapy and radiotherapy by p53 suppression in the tumor stroma is likely to be due to the increased sensitivity of p53-deficient endothelium to genotoxic stress as shown both in cell culture and in experimental tumors. Thus, reversible pharmacologic suppression of p53 may be a viable approach to improving anticancer treatment via an enhanced antiangiogenic effect of chemotherapy and radiotherapy.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Antineoplásicos Alquilantes/uso terapéutico , Benzotiazoles/uso terapéutico , Carcinoma Pulmonar de Lewis/terapia , Ciclofosfamida/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , Imidazoles/uso terapéutico , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Tiazoles/uso terapéutico , Tolueno/análogos & derivados , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Inhibidores de la Angiogénesis/farmacología , Animales , Antineoplásicos Alquilantes/farmacología , Benzotiazoles/farmacología , Benzotiazoles/toxicidad , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/radioterapia , Línea Celular Transformada , Ciclofosfamida/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Células Endoteliales/efectos de la radiación , Fibroblastos , Imidazoles/farmacología , Imidazoles/toxicidad , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Conformación Proteica , Fármacos Sensibilizantes a Radiaciones/farmacología , Fármacos Sensibilizantes a Radiaciones/toxicidad , Ratas , Células del Estroma/efectos de los fármacos , Células del Estroma/efectos de la radiación , Tiazoles/farmacología , Tiazoles/toxicidad , Tolueno/farmacología , Tolueno/uso terapéutico , Tolueno/toxicidad , Transducción Genética , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/genética
20.
Nat Chem Biol ; 2(9): 474-9, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16862141

RESUMEN

p53-dependent apoptosis contributes to the side effects of cancer treatment, and genetic or pharmacological inhibition of p53 function can increase normal tissue resistance to genotoxic stress. It has recently been shown that p53 can induce apoptosis through a mechanism that does not depend on transactivation but instead involves translocation of p53 to mitochondria. To determine the impact of this p53 activity on normal tissue radiosensitivity, we isolated a small molecule named pifithrin-mu (PFTmu, 1) that inhibits p53 binding to mitochondria by reducing its affinity to antiapoptotic proteins Bcl-xL and Bcl-2 but has no effect on p53-dependent transactivation. PFTmu has a high specificity for p53 and does not protect cells from apoptosis induced by overexpression of proapoptotic protein Bax or by treatment with dexamethasone (2). PFTmu rescues primary mouse thymocytes from p53-mediated apoptosis caused by radiation and protects mice from doses of radiation that cause lethal hematopoietic syndrome. These results indicate that selective inhibition of the mitochondrial branch of the p53 pathway is sufficient for radioprotection in vivo.


Asunto(s)
Rayos gamma/efectos adversos , Mitocondrias/metabolismo , Traumatismos Experimentales por Radiación/prevención & control , Protectores contra Radiación/uso terapéutico , Tiazoles/uso terapéutico , Tolueno/análogos & derivados , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Benzotiazoles , Línea Celular , Dexametasona/farmacología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Transporte de Proteínas , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/patología , Protectores contra Radiación/química , Protectores contra Radiación/farmacología , Tiazoles/química , Tiazoles/farmacología , Timo/efectos de los fármacos , Timo/metabolismo , Timo/patología , Tolueno/química , Tolueno/farmacología , Tolueno/uso terapéutico , Activación Transcripcional , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Rayos Ultravioleta/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA