Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.548
Filtrar
1.
Front Immunol ; 12: 770080, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34925340

RESUMEN

Enhancer of Zeste Homolog 2 (EZH2) inhibitors (EZH2i) are approved to treat certain cancer types. Previous studies have suggested the potential to combine EZH2i with immune checkpoint blockade targeting coinhibitory receptors like PD-(L)1 and CTLA-4, but whether it can also enhance the activity of agents targeting costimulatory receptors is not known. Here, we explore the combination between EZH2i and an agonist antibody targeting the T cell costimulatory receptor 4-1BB (α4-1BB). Our data show that EZH2i compromise the efficacy of α4-1BB in both CT26 colon carcinoma and in an in vivo protein immunization model. We link this to reduced effector survival and increased BIM expression in CD8+ T cells upon EZH2i treatment. These data support the requirement of EZH2 function in 4-1BB-mediated CD8+ T cell expansion and effector programming and emphasize the consideration that must be given when combining such antitumoral therapies.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Linfocitos T CD8-positivos/efectos de los fármacos , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Neoplasias Experimentales/prevención & control , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/agonistas , Animales , Anticuerpos Monoclonales/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Proteína Potenciadora del Homólogo Zeste 2/inmunología , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Humanos , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neoplasias Experimentales/genética , Neoplasias Experimentales/inmunología , Carga Tumoral/efectos de los fármacos , Carga Tumoral/genética , Carga Tumoral/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo
2.
Front Immunol ; 12: 769799, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34745146

RESUMEN

Tumor Associated Antigens (TAAs) may suffer from an immunological tolerance due to expression on normal cells. In order to potentiate their immunogenicity, heteroclitic peptides (htcPep) were designed according to prediction algorithms. In particular, specific modifications were introduced in peptide residues facing to TCR. Moreover, a MHC-optimized scaffold was designed for improved antigen presentation to TCR by H-2Db allele. The efficacy of such htcPep was assessed in C57BL/6 mice injected with syngeneic melanoma B16F10 or lung TC1 tumor cell lines, in combination with metronomic chemotherapy and immune checkpoint inhibitors. The immunogenicity of htcPep was significantly stronger than the corresponding wt peptide and the modification involving both MHC and TCR binding residues scored the strongest. In particular, the H-2Db-specific scaffold significantly potentiated the peptides' immunogenicity and control of tumor growth was comparable to wt peptide in a therapeutic setting. Overall, we demonstrated that modified TAAs show higher immunogenicity compared to wt peptide. In particular, the MHC-optimized scaffold can present different antigen sequences to TCR, retaining the conformational characteristics of the corresponding wt. Cross-reacting CD8+ T cells are elicited and efficiently kill tumor cells presenting the wild-type antigen. This novel approach can be of high clinical relevance in cancer vaccine development.


Asunto(s)
Presentación de Antígeno/inmunología , Vacunas contra el Cáncer/inmunología , Antígenos de Histocompatibilidad/inmunología , Neoplasias Experimentales/inmunología , Péptidos/inmunología , Vacunas de Subunidad/inmunología , Animales , Presentación de Antígeno/efectos de los fármacos , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Vacunas contra el Cáncer/administración & dosificación , Línea Celular Tumoral , Terapia Combinada , Femenino , Humanos , Ratones Endogámicos C57BL , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/prevención & control , Péptidos/metabolismo , Unión Proteica , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Resultado del Tratamiento , Carga Tumoral/efectos de los fármacos , Carga Tumoral/inmunología , Vacunas de Subunidad/administración & dosificación
3.
Immunol Lett ; 240: 137-148, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34710507

RESUMEN

Immune checkpoint inhibitors (ICIs), including programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) and cytotoxic T-lymphocyte-associated protein 4 have shown promising cancer clinical outcomes. However, IC therapy has low patient response rates (10%-15%). Thus, ICIs and sufficient antigen combinations into the tumor microenvironment (TME) is important to produce strong tumor-specific adaptive immune responses. Mice were treated with cisplatin, and human cancer cells were exposed to inflammatory cytokines, to confirm increased PD-L1 and major histocompatibility complex (MHC) I expression by tumor cells or dendritic cells. TC-1, CT26, B16-F1, or B16-F10 tumor cells, and bone marrow-derived dendritic cells, were treated with interferon (IFN)-ß, IFN-γ, or tumor necrosis factor-α to identify the molecular mechanisms underlying tumor PD-L1 and MHC I upregulation, and to examine MHC I, CD40, CD80, CD86, or PD-L1 levels, respectively. For synergistic combination therapy, αPD-L1 monoclonal antibody (mAb) covalently linked to the long E7 peptide was generated. Chemotherapy shifted the TME to express high PD-L1 and MHC I, resulting in targeted ICI cargo delivery and enhanced generation and activation of tumor antigen-specific T cells. Synergistic effects of vaccination and IC blockade in the TME were demonstrated using an anti-PD-L1 mAb covalently conjugated to the E7 long peptide.


Asunto(s)
Antígenos/farmacología , Antígeno B7-H1/antagonistas & inhibidores , Inmunoconjugados/farmacología , Inmunoterapia , Neoplasias Experimentales/prevención & control , Péptidos/farmacología , Animales , Antígeno B7-H1/inmunología , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Neoplasias Experimentales/inmunología
4.
Food Funct ; 12(21): 10632-10643, 2021 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-34585698

RESUMEN

Safflower yellow (SY) is the main active ingredient isolated from the traditional Chinese medicine Carthamus tinctorius, which is a valuable natural edible pigment that is widely used to treat cerebrovascular and cardiovascular diseases. However, the effect of SY on hepatocellular carcinoma (HCC) remains unclear. In this study, we showed that SY decreased the degree of injury and inhibited the release of inflammatory factors in the liver of a diethylnitrosamine (DEN)-induced HCC mouse model. Flow cytometry and immunoblotting showed that SY increased the infiltration of CD8+ T cells and Gr-1+ macrophages to improve the immune microenvironment by affecting the expression of collagen fibers. Further cellular experiments showed that SY degraded the collagens in the liver cells through the TGF-ß/Smad signalling pathway. SY also regulated the gut microbiota which may contribute to the immune microenvironment. In conclusion, SY exhibited a potent effect on the development of HCC by enhancing liver immune infiltration by promoting collagen degradation and modulating the gut microbiota. This study provides novel insights into the mechanism of SY as a candidate for the treatment of HCC in the future.


Asunto(s)
Carcinoma Hepatocelular/inducido químicamente , Chalcona/análogos & derivados , Dietilnitrosamina/toxicidad , Microbioma Gastrointestinal/efectos de los fármacos , Neoplasias Hepáticas/inducido químicamente , Hígado/efectos de los fármacos , Animales , Carcinoma Hepatocelular/prevención & control , Línea Celular Tumoral , Chalcona/farmacología , Colágeno/metabolismo , Humanos , Hígado/inmunología , Hígado/metabolismo , Neoplasias Hepáticas/prevención & control , Macrófagos Peritoneales/efectos de los fármacos , Masculino , Ratones , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/prevención & control , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
5.
Int J Biol Sci ; 17(11): 2944-2956, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34345218

RESUMEN

The generation of successful anticancer vaccines relies on the ability to induce efficient and long-lasting immune responses to tumor antigens. In this scenario, dendritic cells (DCs) are essential cellular components in the generation of antitumor immune responses. Thus, delivery of tumor antigens to specific DC populations represents a promising approach to enhance the efficiency of antitumor immunotherapies. In the present study, we employed antibody-antigen conjugates targeting a specific DC C-type lectin receptor. For that purpose, we genetically fused the anti-DEC205 monoclonal antibody to the type 16 human papillomavirus (HPV-16) E7 oncoprotein to create a therapeutic vaccine to treat HPV-associated tumors in syngeneic mouse tumor models. The therapeutic efficacy of the αDEC205-E7 mAb was investigated in three distinct anatomical tumor models (subcutaneous, lingual and intravaginal). The immunization regimen comprised two doses of the αDEC205-E7 mAb coadministered with a DC maturation stimulus (Polyinosinic:polycytidylic acid, poly (I:C)) as an adjuvant. The combined immunotherapy produced robust antitumor effects on both the subcutaneous and orthotopic tumor models, stimulating rapid tumor regression and long-term survival. These outcomes were related to the activation of tumor antigen-specific CD8+ T cells in both systemic compartments and lymphoid tissues. The αDEC205-E7 antibody plus poly (I:C) administration induced long-lasting immunity and controlled tumor relapses. Our results highlight that the delivery of HPV tumor antigens to DCs, particularly via the DEC205 surface receptor, is a promising therapeutic approach, providing new opportunities for the development of alternative immunotherapies for patients with HPV-associated tumors at different anatomical sites.


Asunto(s)
Antígenos CD/inmunología , Vacunas contra el Cáncer/administración & dosificación , Células Dendríticas/inmunología , Lectinas Tipo C/inmunología , Antígenos de Histocompatibilidad Menor/inmunología , Neoplasias Experimentales/prevención & control , Proteínas E7 de Papillomavirus/inmunología , Infecciones por Papillomavirus/prevención & control , Receptores de Superficie Celular/inmunología , Adyuvantes Inmunológicos , Animales , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Femenino , Humanos , Memoria Inmunológica , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/virología , Infecciones por Papillomavirus/inmunología , Infecciones por Papillomavirus/virología , Poli I-C/administración & dosificación
6.
Proc Natl Acad Sci U S A ; 118(23)2021 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-34074778

RESUMEN

Tumors frequently express unmutated self-tumor-associated antigens (self-TAAs). However, trial results using self-TAAs as vaccine targets against cancer are mixed, often attributed to deletion of T cells with high-affinity receptors (TCRs) for self-TAAs during T cell development. Mutating these weak self-TAAs to produce higher affinity, effective vaccines is challenging, since the mutations may not benefit all members of the broad self-TAA-specific T cell repertoire. We previously identified a common weak murine self-TAA that we converted to a highly effective antitumor vaccine by a single amino acid substitution. In this case the modified and natural self-TAAs still raised very similar sets of CD8 T cells. Our structural studies herein show that the modification of the self-TAA resulted in a subtle change in the major histocompatibility complex I-TAA structure. This amino acid substitution allowed a dramatic conformational change in the peptide during subsequent TCR engagement, creating a large increase in TCR affinity and accounting for the efficacy of the modified self-TAA as a vaccine. These results show that carefully selected, well-characterized modifications to a poorly immunogenic self-TAA can rescue the immune response of the large repertoire of weakly responding natural self-TAA-specific CD8 T cells, driving them to proliferate and differentiate into functional effectors. Subsequently, the unmodified self-TAA on the tumor cells, while unable to drive this response, is nevertheless a sufficient target for the CD8 cytotoxic effectors. Our results suggest a pathway for more efficiently identifying variants of common self-TAAs, which could be useful in vaccine development, complementing other current nonantigen-specific immunotherapies.


Asunto(s)
Antígenos de Neoplasias/inmunología , Autoantígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Neoplasias Experimentales/inmunología , Péptidos/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Animales , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Femenino , Ratones , Ratones Endogámicos BALB C , Neoplasias Experimentales/prevención & control , Células Sf9 , Spodoptera
7.
J Drug Target ; 29(10): 1102-1110, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33926356

RESUMEN

It has been previously reported that targeting and retaining antigens in the endoplasmic reticulum (ER) can induce an ER stress response. In this study, we evaluated the antitumor effect of E7 antigen fused to an ERresident protein, cyclooxygenase-2, which possesses a 19-aminoacid cassette that directs it to the endoplasmic reticulum-associated protein degradation (ERAD) pathway. The featured DNA constructs, COX2-E7 and COX2-E7ΔERAD, with a deletion in the 19-aminoacid cassette, were used to evaluate the importance of this sequence. In vitro analysis of protein expression and ER localisation were verified. We observed that both constructs induced an ER stress response. This finding correlated with the antitumor effect in mice injected with TC-1 cells and treated with different DNA constructs by biolistic vaccination. Immunisation with COX2-E7 and COX2-E7ΔERAD DNA constructs induced a significant antitumor effect in mice, without a significant difference between them, although the COX2-E7 construct induced a significant E7-specific immune response. These results demonstrate that targeting the E7 antigen to the ERAD pathway promotes a potent therapeutic antitumor effect. This strategy could be useful for the design of other antigen-specific therapies.


Asunto(s)
Vacunas contra el Cáncer/administración & dosificación , Ciclooxigenasa 2/química , Estrés del Retículo Endoplásmico/inmunología , Proteínas E7 de Papillomavirus/inmunología , Animales , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Ciclooxigenasa 2/administración & dosificación , Retículo Endoplásmico/inmunología , Degradación Asociada con el Retículo Endoplásmico/inmunología , Femenino , Células HEK293 , Humanos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/prevención & control , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/prevención & control , Vacunas de ADN/administración & dosificación , Vacunas de ADN/inmunología
8.
Cancer Prev Res (Phila) ; 14(5): 527-540, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33648941

RESUMEN

Skin cancer is the most common malignancy worldwide and is rapidly rising in incidence, representing a significant public health challenge. The ß-blocker, carvedilol, has shown promising effects in preventing skin cancer. However, as a potent ß-blocker, repurposing carvedilol to an anticancer agent is limited by cardiovascular effects. Carvedilol is a racemic mixture consisting of equimolar S- and R-carvedilol, whereas the R-carvedilol enantiomer does not possess ß-blocking activity. Because previous studies suggest that carvedilol's cancer preventive activity is independent of ß-blockade, we examined the skin cancer preventive activity of R-carvedilol compared with S-carvedilol and the racemic carvedilol. R- and S-carvedilol were equally effective in preventing EGF-induced neoplastic transformation of the mouse epidermal JB6 Cl 41-5a (JB6 P+) cells and displayed similar attenuation of EGF-induced ELK-1 activity. R-carvedilol appeared slightly better than S-carvedilol against UV-induced intracellular oxidative stress and release of prostaglandin E2 from the JB6 P+ cells. In an acute UV-induced skin damage and inflammation mouse model using a single irradiation of 300 mJ/cm2 UV, topical treatment with R-carvedilol dose dependently attenuated skin edema and reduced epidermal thickening, Ki-67 staining, COX-2 protein, and IL6 and IL1ß mRNA levels similar to carvedilol. In a chronic UV (50-150 mJ/cm2) induced skin carcinogenesis model in mice with pretreatment of test agents, topical treatment with R-carvedilol, but not racemic carvedilol, significantly delayed and reduced skin squamous cell carcinoma development. Therefore, as an enantiomer present in an FDA-approved agent, R-carvedilol may be a better option for developing a safer and more effective preventive agent for skin carcinogenesis. PREVENTION RELEVANCE: In this study, we demonstrated the skin cancer preventive activity of R-carvedilol, the non-ß-blocking enantiomer present in the racemic ß-blocker, carvedilol. As R-carvedilol does not have ß-blocking activity, such a preventive treatment would not lead to common cardiovascular side effects of ß-blockers.


Asunto(s)
Carcinogénesis/efectos de los fármacos , Carvedilol/administración & dosificación , Epidermis/efectos de los fármacos , Neoplasias Experimentales/prevención & control , Neoplasias Cutáneas/prevención & control , Animales , Carcinogénesis/inducido químicamente , Carcinogénesis/patología , Carcinogénesis/efectos de la radiación , Carvedilol/química , Células Epidérmicas , Factor de Crecimiento Epidérmico/toxicidad , Epidermis/patología , Epidermis/efectos de la radiación , Femenino , Células HEK293 , Humanos , Ratones , Neoplasias Experimentales/etiología , Neoplasias Experimentales/patología , Neoplasias Cutáneas/etiología , Neoplasias Cutáneas/patología , Estereoisomerismo , Rayos Ultravioleta/efectos adversos
9.
Nat Commun ; 12(1): 773, 2021 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-33536439

RESUMEN

Macrophages are plastic and, in response to different local stimuli, can polarize toward multi-dimensional spectrum of phenotypes, including the pro-inflammatory M1-like and the anti-inflammatory M2-like states. Using a high-throughput phenotypic screen in a library of ~4000 FDA-approved drugs, bioactive compounds and natural products, we find ~300 compounds that potently activate primary human macrophages toward either M1-like or M2-like state, of which ~30 are capable of reprogramming M1-like macrophages toward M2-like state and another ~20 for the reverse repolarization. Transcriptional analyses of macrophages treated with 34 non-redundant compounds identify both shared and unique targets and pathways through which the tested compounds modulate macrophage activation. One M1-activating compound, thiostrepton, is able to reprogram tumor-associated macrophages toward M1-like state in mice, and exhibit potent anti-tumor activity. Our compound-screening results thus help to provide a valuable resource not only for studying the macrophage biology but also for developing therapeutics through modulating macrophage activation.


Asunto(s)
Antiinflamatorios/farmacología , Productos Biológicos/farmacología , Ensayos Analíticos de Alto Rendimiento/métodos , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Animales , Antiinflamatorios/química , Productos Biológicos/química , Línea Celular Tumoral , Células Cultivadas , Expresión Génica/efectos de los fármacos , Ontología de Genes , Humanos , Macrófagos/clasificación , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Neoplasias Experimentales/prevención & control , Fenotipo , Células THP-1 , Tioestreptona/química , Tioestreptona/farmacología
10.
Sci China Life Sci ; 64(4): 548-562, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32737851

RESUMEN

The interaction of PD-1/PD-L1 allows tumor cells to escape from immune surveillance. Clinical success of the antibody drugs has proven that blockade of PD-1/PD-L1 pathway is a promising strategy for cancer immunotherapy. Here, we developed a cyclic peptide C8 by using Ph.D.-C7C phage display technology. C8 showed high binding affinity with hPD-1 and could effectively interfere the interaction of PD-1/PD-L1. Furthermore, C8 could stimulate CD8+ T cell activation in human peripheral blood mononuclear cells (PBMCs). We also observed that C8 could suppress tumor growth in CT26 and B16-OVA, as well as anti-PD-1 antibody resistant B16 mouse model. CD8 T cells infiltration significantly increased in tumor microenvironment, and IFN-γ secretion by CD8+ T cells in draining lymph nodes also increased. Simultaneously, we exploited T cells depletion models and confirmed that C8 exerted anti-tumor effects via activating CD8+ T cells dependent manner. The interaction model of C8 with hPD-1 was simulated and confirmed by alanine scanning. In conclusion, C8 shows anti-tumor capability by blockade of PD-1/PD-L1 interaction, and C8 may provide an alternative candidate for cancer immunotherapy.


Asunto(s)
Antígeno B7-H1/metabolismo , Inmunoterapia/métodos , Neoplasias/terapia , Péptidos Cíclicos/farmacología , Receptor de Muerte Celular Programada 1/metabolismo , Animales , Antígeno B7-H1/genética , Antígeno B7-H1/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Células CHO , Línea Celular Tumoral , Cricetinae , Cricetulus , Femenino , Humanos , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Ratones Endogámicos BALB C , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/prevención & control , Péptidos Cíclicos/química , Péptidos Cíclicos/metabolismo , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología , Unión Proteica/efectos de los fármacos , Unión Proteica/inmunología , Carga Tumoral/efectos de los fármacos , Carga Tumoral/inmunología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
11.
Gastroenterology ; 160(4): 1179-1193.e14, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32920015

RESUMEN

BACKGROUND & AIMS: Streptococcus thermophilus was identified to be depleted in patients with colorectal cancer (CRC) by shotgun metagenomic sequencing of 526 multicohort fecal samples. Here, we aim to investigate whether this bacterium could act as a prophylactic for CRC prevention. METHODS: The antitumor effects of S thermophilus were assessed in cultured colonic epithelial cells and in 2 murine models of intestinal tumorigenesis. The tumor-suppressive protein produced by S thermophilus was identified by mass spectrometry and followed by ß-galactosidase activity assay. The mutant strain of S thermophilus was constructed by homologous recombination. The effect of S thermophilus on the gut microbiota composition was assessed by shotgun metagenomic sequencing. RESULTS: Oral gavage of S thermophilus significantly reduced tumor formation in both Apcmin/+ and azoxymethane-injected mice. Coincubation with S thermophilus or its conditioned medium decreased the proliferation of cultured CRC cells. ß-Galactosidase was identified as the critical protein produced by S thermophilus by mass spectrometry screening and ß-galactosidase activity assay. ß-Galactosidase secreted by S thermophilus inhibited cell proliferation, lowered colony formation, induced cell cycle arrest, and promoted apoptosis of cultured CRC cells and retarded the growth of CRC xenograft. The mutant S thermophilus without functional ß-galactosidase lost its tumor-suppressive effect. Also, S thermophilus increased the gut abundance of known probiotics, including Bifidobacterium and Lactobacillus via ß-galactosidase. ß-Galactosidase-dependent production of galactose interfered with energy homeostasis to activate oxidative phosphorylation and downregulate the Hippo pathway kinases, which partially mediated the anticancer effects of S thermophilus. CONCLUSION: S thermophilus is a novel prophylactic for CRC prevention in mice. The tumor-suppressive effect of S thermophilus is mediated at least by the secretion of ß-galactosidase.


Asunto(s)
Proteínas Bacterianas/metabolismo , Neoplasias Colorrectales/prevención & control , Probióticos/administración & dosificación , Streptococcus thermophilus/enzimología , beta-Galactosidasa/metabolismo , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Azoximetano/administración & dosificación , Azoximetano/toxicidad , Proteínas Bacterianas/genética , Línea Celular Tumoral , Transformación Celular Neoplásica/inducido químicamente , Colon/microbiología , Neoplasias Colorrectales/inducido químicamente , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/microbiología , Humanos , Mucosa Intestinal/microbiología , Masculino , Ratones , Ratones Transgénicos , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/genética , Neoplasias Experimentales/microbiología , Neoplasias Experimentales/prevención & control , Probióticos/metabolismo , Streptococcus thermophilus/genética , beta-Galactosidasa/genética
12.
Proc Natl Acad Sci U S A ; 117(50): 31993-32004, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33262282

RESUMEN

Effective cancer prevention requires the discovery and intervention of a factor critical to cancer development. Here we show that ovarian progesterone is a crucial endogenous factor inducing the development of primary tumors progressing to metastatic ovarian cancer in a mouse model of high-grade serous carcinoma (HGSC), the most common and deadliest ovarian cancer type. Blocking progesterone signaling by the pharmacologic inhibitor mifepristone or by genetic deletion of the progesterone receptor (PR) effectively suppressed HGSC development and its peritoneal metastases. Strikingly, mifepristone treatment profoundly improved mouse survival (∼18 human years). Hence, targeting progesterone/PR signaling could offer an effective chemopreventive strategy, particularly in high-risk populations of women carrying a deleterious mutation in the BRCA gene.


Asunto(s)
Proteína BRCA1/genética , Cistadenocarcinoma Seroso/prevención & control , Mifepristona/farmacología , Neoplasias Ováricas/prevención & control , Progesterona/antagonistas & inhibidores , Adulto , Animales , Mama/patología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Neoplasias de la Mama/prevención & control , Cistadenocarcinoma Seroso/química , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/patología , Modelos Animales de Enfermedad , Estradiol/administración & dosificación , Femenino , Humanos , Ratones , Persona de Mediana Edad , Mifepristona/uso terapéutico , Mutación , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Neoplasias Experimentales/prevención & control , Neoplasias Ováricas/inducido químicamente , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Ovario/patología , Ovario/cirugía , Progesterona/administración & dosificación , Progesterona/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Salpingooforectomía , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
13.
Front Immunol ; 11: 581301, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33193386

RESUMEN

Murine and human invariant natural killer T (iNKT) lymphocytes are activated by α-galactosylceramide (α-GalCer) presented on CD1d. α-GalCer was first described as a lipid that had strong anti-metastatic effects in a mouse melanoma model, and it has subsequently been shown to induce efficient iNKT cell dependent tumor immunity in several tumor models. We have shown that α-GalCer treatment leads to a weak reduction of polyp burden in the autochthonous ApcMin/+ mouse model for human colon cancer, however this treatment resulted in upregulation of the inhibitory receptor PD-1 on iNKT cells. While anti-PD-1 treatment can prevent immune-suppression in other cancer types, human colon cancer is generally resistant to this treatment. Here we have used the ApcMin/+ model to investigate whether a combined treatment with α-GalCer and PD-1 blockade results in improved effects on polyp development. We find that PD-1 expression was high on T cells in polyps and lamina propria (LP) of ApcMin/+ mice compared to polyp free Apc+/+ littermates. Anti-PD-1 treatment alone promoted Tbet expression in iNKT cells and CD4 T cells, but did not significantly reduce polyp numbers. However, the combined treatment with anti-PD-1 and α-GalCer had synergistic effects, resulting in highly significant reduction of polyp numbers in the small and large intestine. Addition of PD-1 blockade to α-GalCer treatment prevented loss of iNKT cells that were skewed towards a TH1-like iNKT1 phenotype specifically in polyps. It also resulted in TH1 skewing and increased granzyme B expression of CD4 T cells. Taken together this demonstrates that a combination of immune stimulation targeting iNKT cells and checkpoint blockade may be a promising approach to develop for improved tumor immunotherapy.


Asunto(s)
Neoplasias del Colon/inmunología , Neoplasias del Colon/prevención & control , Galactosilceramidas/administración & dosificación , Células T Asesinas Naturales/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Proteína de la Poliposis Adenomatosa del Colon/deficiencia , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Anticuerpos Bloqueadores/administración & dosificación , Femenino , Humanos , Mucosa Intestinal/inmunología , Pólipos Intestinales/inmunología , Pólipos Intestinales/prevención & control , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/prevención & control , Receptor de Muerte Celular Programada 1/inmunología , Linfocitos T Reguladores/inmunología , Células TH1/inmunología
14.
Cancer Chemother Pharmacol ; 86(5): 681-686, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32980903

RESUMEN

PURPOSE: Beta-adrenergic signaling can influence cancer progression and the use of beta blockers as adjuvant drugs in oncologic patients has been suggested. However, the involvement of beta-adrenergic blockers in tumorigenesis is poorly understood. This study investigated the action of beta-adrenergic blocker propranolol on tumor onset using a preclinical model of chemically induced oral cancer. METHODS: Thirty-two male Wistar rats were subjected to daily subcutaneous injection of beta-blocker propranolol (10 mg/kg; SubQ), while another 32 rats received only a PBS injection (sham group). One week after starting propranolol treatment, all rats were submitted to chemical induction of oral carcinogenesis with 4-nitroquinoline-1-oxide (4NQO). After 16 weeks, they were assessed for occurrence of oral squamous cell carcinoma (OSCC), in addition to measurement of tumor volume and thickness, and tissue levels of cytokines IL-6, TNF-alpha and IL-10 in the tumor microenvironment. RESULTS: Propranolol treatment reduced the occurrence of OSCC by 31%, 95% CI ( - 127, 216). Beta-adrenergic blocker significantly decreased thickness of OSCC when compared with PBS. Rats treated with propranolol exhibited a lower tumor volume when compared with control rats, but this result did not reach statistical significance. Tumors from propranolol-treated rats exhibited reduced concentrations of pro-inflammatory cytokines IL-6 and TNF-α. There was no difference in the IL-10 levels between tumors from propranolol- and sham-treated rats. CONCLUSION: Beta-adrenergic signaling may be one of the mechanisms associated with chemically induced oral carcinogenesis.


Asunto(s)
Antagonistas Adrenérgicos beta/administración & dosificación , Carcinogénesis/efectos de los fármacos , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias Experimentales/tratamiento farmacológico , Propranolol/administración & dosificación , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , 4-Nitroquinolina-1-Óxido/administración & dosificación , 4-Nitroquinolina-1-Óxido/toxicidad , Animales , Carcinogénesis/inducido químicamente , Carcinógenos/administración & dosificación , Carcinógenos/toxicidad , Citocinas/inmunología , Citocinas/metabolismo , Progresión de la Enfermedad , Humanos , Masculino , Mucosa Bucal/efectos de los fármacos , Mucosa Bucal/inmunología , Mucosa Bucal/patología , Neoplasias de la Boca/inducido químicamente , Neoplasias de la Boca/patología , Neoplasias de la Boca/prevención & control , Invasividad Neoplásica/prevención & control , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/patología , Neoplasias Experimentales/prevención & control , Ratas , Ratas Wistar , Receptores Adrenérgicos beta/metabolismo , Transducción de Señal/efectos de los fármacos , Carcinoma de Células Escamosas de Cabeza y Cuello/inducido químicamente , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Carcinoma de Células Escamosas de Cabeza y Cuello/prevención & control , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
15.
Front Immunol ; 11: 1750, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32922389

RESUMEN

Anogenital and oropharyngeal cancers caused by human papillomavirus (HPV) infections account for 4.5% of all cancer cases worldwide. So far, only the initial infection with selected high-risk types can be prevented by prophylactic vaccination. Already existing persistent HPV infections, however, can currently only be treated by surgical removal of resulting lesions. Therapeutic HPV vaccination, promoting cell-based anti-HPV immunity, would be ideal to eliminate and protect against HPV-induced lesions and tumors. A multitude of vaccination approaches has been tested to date, many of which led to high amounts of HPV-specific T cells in vivo. However, growing evidence suggests that not the induction of systemic but of local immunity is paramount for tackling mucosal infections and tumors. Therefore, recent therapeutic vaccination studies have focused on how to induce tissue-resident T cells in the anogenital and oropharyngeal mucosa. These approaches include direct mucosal vaccinations and influencing the migration of systemic T cells toward the mucosa. The efficacy of these new vaccination approaches is best tested in vivo by utilizing orthotopic tumor models, i.e. HPV-positive tumors being located in the animal's mucosa. In line with this, we here review existing HPV tumor models and describe two novel tumorigenic cell lines for the MHC-humanized mouse model A2.DR1. These were used for the establishment of an HPV16 E6/E7-positive vaginal tumor model, suitable for testing therapeutic vaccines containing HLA-A2-restricted HPV16-derived epitopes. The newly developed MHC-humanized orthotopic HPV16-positive tumor model is likely to improve the translatability of in vivo findings to the clinical setting.


Asunto(s)
Alphapapillomavirus/patogenicidad , Vacunas contra el Cáncer/uso terapéutico , Neoplasias Experimentales/prevención & control , Infecciones por Papillomavirus/prevención & control , Vacunas contra Papillomavirus/uso terapéutico , Alphapapillomavirus/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Línea Celular Tumoral , Interacciones Huésped-Patógeno , Humanos , Inmunidad Mucosa , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/virología , Ratones Transgénicos , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/virología , Infecciones por Papillomavirus/inmunología , Infecciones por Papillomavirus/virología , Vacunación
16.
Cancer Prev Res (Phila) ; 13(9): 735-746, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32655003

RESUMEN

The STAT3 pathway is frequently overactive in non-small cell lung cancer (NSCLC), an often fatal disease with known risk factors including tobacco and chemical exposures. Whether STAT3 can be downmodulated to delay or prevent development of lung cancer resulting from an environmental exposure has not been previously tested. A circular oligonucleotide STAT3 decoy (CS3D) was used to treat mice previously exposed to the tobacco carcinogen nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone. CS3D contains a double-stranded STAT3 DNA response element sequence and interrupts STAT3 signaling by binding to STAT3 dimers, rendering them unable to initiate transcription at native STAT3 DNA binding sites. An intermittent course of CS3D decreased the development of airway preneoplasias by 42% at 1 week posttreatment, reduced the progression of preneoplasia to adenomas by 54% at 8 weeks posttreatment, and reduced the size and number of resulting lung tumors by 49.7% and 29.5%, respectively, at 20 weeks posttreatment. No toxicity was detected. A mutant cyclic oligonucleotide with no STAT3 binding ability was used as a control. Chemopreventive effects were independent of the KRAS mutational status of the tumors. In lungs harvested during and after the treatment course with CS3D, airway preneoplasias had reduced STAT3 signaling. Chemopreventive effects were accompanied by decreased VEGFA expression, ablated IL6, COX-2, and p-NF-κB, and decreased pulmonary M2 macrophages and myeloid-derived suppressor cells. Thus, downmodulation of STAT3 activity using a decoy molecule both reduced oncogenic signaling in the airway epithelium and favored a lung microenvironment with reduced immunosuppression.


Asunto(s)
Anticarcinógenos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/prevención & control , Neoplasias Pulmonares/prevención & control , Nicotiana/toxicidad , Factor de Transcripción STAT3/antagonistas & inhibidores , Animales , Anticarcinógenos/uso terapéutico , Butanonas/toxicidad , Carcinogénesis/inducido químicamente , Carcinogénesis/efectos de los fármacos , Carcinógenos/toxicidad , Carcinoma de Pulmón de Células no Pequeñas/inducido químicamente , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Pulmón/efectos de los fármacos , Pulmón/patología , Neoplasias Pulmonares/inducido químicamente , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Ratones , Mutación , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Neoplasias Experimentales/prevención & control , Nitrosaminas/toxicidad , Oligonucleótidos/farmacología , Oligonucleótidos/uso terapéutico , Proteínas Proto-Oncogénicas p21(ras)/genética , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Nicotiana/química , Activación Transcripcional/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
17.
J Immunother Cancer ; 8(1)2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32581060

RESUMEN

BACKGROUND: In vivo targeting of human papillomavirus (HPV) derived antigens to dendritic cells might constitute an efficient immunotherapeutic strategy against cervical cancer. In previous works, we have shown that the extra domain A from murine fibronectin (mEDA) can be used to target antigens to toll-like receptor 4 (TLR4) expressing dendritic cells and induce strong antigen-specific immune responses. In the present study, we have produced a bivalent therapeutic vaccine candidate consisting of the human EDA (hEDA) fused to E7 proteins from HPV16 and HPV18 (hEDA-HPVE7-16/18) and evaluate its potential as a therapeutic vaccine against cervical cancer. MATERIALS AND METHODS: Recombinant fusion proteins containing HPV E7 proteins from HPV16 and HPV18 virus subtypes fused to hEDA were produced and tested in vitro on their capacity to bind TLR4 and induce the production of tumor necrosis factor-α or interleukin (IL)-12 by human monocytes and dendritic cells. The immunogenicity and potential therapeutic activity of the vaccine in combination with cisplatin or with the TLR3 agonist molecules polyinosinic-polycytidylic acid (Poly IC) or Poly ICLC was evaluated in mice bearing subcutaneous or genital orthotopic HPV16 TC-1 tumors. RESULTS: hEDA-HPVE7-16/18 prototype vaccine binds human TLR4 and stimulate TLR4-dependent signaling pathways and IL-12 production by human monocyte-derived dendritic cell. Vaccination with hEDA-HPVE7-16/18 induced strong HPVE7-specific Cytotoxic T lymphocyte (CTL) responses and eliminated established tumors in the TC-1-based tumor model. The antitumor efficacy was significantly improved by combining the fusion protein with cisplatin or with the TLR-3 ligand Poly IC and especially with the stabilized analog Poly ICLC. Moreover, hEDA-HPVE7-16/18+Poly ICLC induced full tumor regression in 100% of mice bearing orthotopic genital HPV tumors. CONCLUSION: Our results suggest that this therapeutic vaccine formulation may be an effective treatment for cervical tumors that do not respond to current therapies.


Asunto(s)
Vacunas contra el Cáncer/administración & dosificación , Proteínas de Unión al ADN/inmunología , Fibronectinas/inmunología , Neoplasias Experimentales/prevención & control , Proteínas Oncogénicas Virales/inmunología , Proteínas E7 de Papillomavirus/inmunología , Infecciones por Papillomavirus/prevención & control , Animales , Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Femenino , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/inmunología , Papillomavirus Humano 18/genética , Papillomavirus Humano 18/inmunología , Humanos , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/virología , Infecciones por Papillomavirus/inmunología , Infecciones por Papillomavirus/virología , Linfocitos T Citotóxicos/inmunología
18.
World J Gastroenterol ; 26(17): 2064-2081, 2020 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-32536775

RESUMEN

BACKGROUND: Epigallocatechin gallate (EGCG) is a polyhydroxy phenolic compound extracted from tea and its antitumor effect has received widespread attention. We explored the inhibitory effect of EGCG on dimethylhydrazine (DMH)-induced colorectal cancer (CRC) using a rat model, predicted the interaction between EGCG and CRC target genes using a database, and explained the EGCG associated target pathways and mechanisms in CRC. AIM: To understand the inhibitory mechanisms of EGCG on CRC cell proliferation and identify its pharmacological targets by network pharmacology analysis. METHODS: DMH (40 mg/kg, s.c., twice weekly for eight weeks) was used to induce CRC in rats. After model establishment, the rats were administered with EGCG (50, 100, or 200 mg/kg, p.o., once daily for eight weeks) and killed 12 and 20 wk after the start of the experiment. Formation of aberrant crypt foci and tumor was studied by histological analysis. Using network pharmacology analysis, candidate and collective targets of EGCG and CRC were identified, and Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes analyses were used to predict the pathways altered by EGCG. RESULTS: At week 12, high-dose EGCG treatment significantly reduced the tumor formation rate, total number of tumors, cancerous and non-cancerous tumors, tumor volume, ascites formation, and aberrant crypt foci count. At week 20, all three doses of EGCG were effective. Seventy-eight collective targets of EGCG and CRC were identified, of which 28 genes were dysregulated in CRC. Kyoto Encyclopedia of Genes and Genomes and GO analyses showed that the dysregulated genes were enriched in hsa05210 (CRC), hsa04115 (p53 signaling pathway), and hsa04151 (PI3K-Akt signaling pathway), GO:0043124 (negative regulation of I-kappaB kinase/NF-kappaB signaling pathway), GO:0043409 (negative regulation of mitogen-activated protein kinase cascade), and GO:2001244 (positive regulation of intrinsic apoptotic signaling pathway) respectively. CONCLUSION: EGCG inhibits the formation of DMH-induced CRC by regulating key pathways involved in tumorigenesis.


Asunto(s)
Focos de Criptas Aberrantes/prevención & control , Anticarcinógenos/farmacología , Catequina/análogos & derivados , Neoplasias Colorrectales/prevención & control , Neoplasias Experimentales/prevención & control , Focos de Criptas Aberrantes/inducido químicamente , Focos de Criptas Aberrantes/genética , Focos de Criptas Aberrantes/patología , Animales , Anticarcinógenos/uso terapéutico , Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Catequina/farmacología , Catequina/uso terapéutico , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Colon/efectos de los fármacos , Colon/patología , Neoplasias Colorrectales/inducido químicamente , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Dimetilhidrazinas/toxicidad , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Masculino , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Mapas de Interacción de Proteínas/efectos de los fármacos , Mapas de Interacción de Proteínas/genética , Ratas , Recto/efectos de los fármacos , Recto/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
19.
J Immunother Cancer ; 8(1)2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32561637

RESUMEN

BACKGROUND: Envisioned as a similar process to tumorigenesis in terms of biological behaviors and molecular basis, embryogenesis necessitates an immune surveillance system to eliminate erratically transformed cells. Our previous study demonstrated that fetal macrophage-like phagocytes triggered Th2-skewed immunity following endocytosing prenatally administered ovalbumin to facilitate postnatal allergic airway responses, highlighting the critical role fetal phagocytes played in dealing with antigens present in developing fetuses and shaping subsequent immune responses. It prompted us to examine whether fetuses could mount Th1 tumoricidal immunity against tumorigenesis following in utero exposure to tumor antigens. METHODS: Gestational day 14 murine fetuses underwent in utero injection of Th1-promoting human papilloma virus (HPV) E7 peptides. Postnatally, recipients were examined for immunological consequences and the resistance to TC-1 tumorigenesis. RESULTS: Fetal exposure to HPV E7 did not cause tolerance but rather immunization in the recipients, characterized by proinflammatory Th1 polarization of their lymphocytes. Fetal macrophage-like phagocytes were responsible for taking up HPV E7 and triggering HPV E7-specific T-cell cytotoxicity and humoral immunity that rendered recipients resistant to TC-1 tumorigenesis in postnatal life. Adoptive transfer of HPV E7-loaded fetal phagocytes also elicited Th1 immunity with rapid expansion of HPV E7-specific cytotoxic CD8+ T-cell clones in response to TC-1 cell challenge so as to protect the recipients from TC-1 tumorigenesis, but failed to completely eliminate pre-existing TC-1 cells despite perceptible attenuation of local TC-1 tumor growth. CONCLUSIONS: Our study revealed that Th2-biasing fetus was not immune-privileged to foreign peptides, but competent to mount Th1 cytotoxic immunity and generate immunoglobulins against tumorigenesis following in utero exposure to Th1-promoting oncoantigen. It shed light on the role of fetal macrophage-like phagocytes in bridging toward tumor antigen-specific cellular and humoral immunity potentially as an immune surveillance system to eliminate transformed cells that might be egressing during embryogenesis and leftover until postnatal life.


Asunto(s)
Inmunidad Adaptativa/inmunología , Antígenos de Neoplasias/inmunología , Feto/inmunología , Neoplasias Experimentales/inmunología , Papillomaviridae/inmunología , Proteínas E7 de Papillomavirus/inmunología , Infecciones por Papillomavirus/complicaciones , Animales , Femenino , Feto/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales/prevención & control , Neoplasias Experimentales/virología , Infecciones por Papillomavirus/virología , Embarazo
20.
Pediatr Blood Cancer ; 67(8): e28372, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32459399

RESUMEN

BACKGROUND: Neurofibromatosis type 1 (NF1) is a common genetic disorder characterized by plexiform neurofibromas (pNF), which are thought to be congenital tumors that arise in utero and enlarge throughout life. Genetic studies in murine models delineated an indispensable role for the stem cell factor (SCF)/c-kit pathway in pNF initiation and progression. A subsequent phase 2 clinical trial using imatinib mesylate to inhibit SCF/c-kit demonstrated tumor shrinkage in a subset of preexisting pNF; however, imatinib's role on preventing pNF development has yet to be explored. PROCEDURE: We evaluated the effect of imatinib dosed at 10-100 mg/kg/day for 12 weeks to one-month-old Nf1flox/flox ;PostnCre(+) mice, prior to onset of pNF formation. To determine durability of response, we then monitored for pNF growth at later time points, comparing imatinib- with vehicle-treated mice. We assessed gross and histopathological analysis of tumor burden. RESULTS: Imatinib administered preventatively led to a significant decrease in pNF number, even at doses as low as 10 mg/kg/day. Tumor development continued to be significantly inhibited after cessation of imatinib dosed at 50 and 100 mg/kg/day. In the cohort of treated mice that underwent prolonged follow-up, the size of residual tumors was significantly reduced as compared with age-matched littermates that received vehicle control. CONCLUSIONS: Early administration of imatinib inhibits pNF genesis in vivo, and effects are sustained after discontinuation of therapy. These findings may guide clinical use of imatinib in young NF1 patients prior to the substantial development of pNF.


Asunto(s)
Mesilato de Imatinib/administración & dosificación , Neoplasias Experimentales/prevención & control , Neurofibroma Plexiforme/prevención & control , Neurofibromatosis 1/prevención & control , Animales , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Ratones , Ratones Transgénicos , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Neurofibroma Plexiforme/genética , Neurofibroma Plexiforme/metabolismo , Neurofibroma Plexiforme/patología , Neurofibromatosis 1/genética , Neurofibromatosis 1/metabolismo , Neurofibromatosis 1/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...