Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
J Transl Med ; 21(1): 562, 2023 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-37608347

RESUMEN

Alpha-melanocyte stimulating hormone (α-MSH) and its receptor, melanocortin 1 receptor (MC1R), have been proposed as potential target for anti-cancer strategies in melanoma research, due to their tissue specific expression and involvement in melanocyte homeostasis. However, their role in prevention and treatment of melanoma is still debated and controversial. Although a large body of evidence supports α-MSH in preventing melanoma development, some preclinical findings suggest that the α-MSH downstream signalling may promote immune escape and cancer resistance to therapy. Additionally, in metastatic melanoma both MC1R and α-MSH have been reported to be overexpressed at levels much higher than normal cells. Furthermore, targeted therapy (e.g. BRAF inhibition in BRAFV600E mutant tumours) has been shown to enhance this phenomenon. Collectively, these data suggest that targeting MC1R could serve as an approach in the treatment of metastatic melanoma. In this review, we explore the molecular biology of α-MSH with particular emphasis into its tumor-related properties, whilst elaborating the experimental evidence currently available regarding the interplay between α-MSH/MC1R axis, melanoma and antitumor strategies.


Asunto(s)
Melanoma , Receptor de Melanocortina Tipo 1 , alfa-MSH , Humanos , Relevancia Clínica , Melanoma/genética , Proteínas Proto-Oncogénicas B-raf/genética , Receptor de Melanocortina Tipo 1/genética
2.
Biomacromolecules ; 24(9): 4229-4239, 2023 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-37638739

RESUMEN

In this study, a circular conjugate of granulocyte colony-stimulating factor (G-CSF) was prepared by conjugating the two end-chains of poly(ethylene glycol) (PEG) to two different sites of the protein. For the orthogonal conjugation, a heterobifunctional PEG chain was designed and synthesized, bearing the dipeptide ZGln-Gly (ZQG) at one end-chain, for transglutaminase (TGase) enzymatic selective conjugation at Lys41 of G-CSF, and an aldehyde group at the opposite end-chain, for N-terminal selective reductive alkylation of the protein. The cPEG-Nter/K41-G-CSF circular conjugate was characterized by physicochemical methods and compared with native G-CSF and the corresponding linear monoconjugates of G-CSF, PEG-Nter-G-CSF, and PEG-K41-G-CSF. The results demonstrated that the circular conjugate had improved physicochemical and thermal stability, prolonged pharmacokinetic interaction, and retained the biological activity of G-CSF. The PEGylation strategy employed in this study has potential applications in the design of novel protein-based therapeutics.


Asunto(s)
Aldehídos , Factor Estimulante de Colonias de Granulocitos , Alquilación , Fenómenos Químicos , Dipéptidos
3.
J Nanobiotechnology ; 18(1): 31, 2020 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-32066449

RESUMEN

BACKGROUND: Myeloid derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs) are two of the major players involved in the inhibition of anti-tumor immune response in cancer patients, leading to poor prognosis. Selective targeting of myeloid cells has therefore become an attractive therapeutic strategy to relieve immunosuppression and, in this frame, we previously demonstrated that lipid nanocapsules (LNCs) loaded with lauroyl-modified gemcitabine efficiently target monocytic MDSCs in melanoma patients. In this study, we investigated the impact of the physico-chemical characteristics of LNCs, namely size and surface potential, towards immunosuppressive cell targeting. We exploited myeloid cells isolated from glioblastoma patients, which play a relevant role in the immunosuppression, to demonstrate that tailored nanosystems can target not only tumor cells but also tumor-promoting cells, thus constituting an efficient system that could be used to inhibit their function. RESULTS: The incorporation of different LNC formulations with a size of 100 nm, carrying overall positive, neutral or negative charge, was evaluated on leukocytes and tumor-infiltrating cells freshly isolated from glioblastoma patients. We observed that the maximum LNC uptake was obtained in monocytes with neutral 100 nm LNCs, while positively charged 100 nm LNCs were more effective on macrophages and tumor cells, maintaining at low level the incorporation by T cells. The mechanism of uptake was elucidated, demonstrating that LNCs are incorporated mainly by caveolae-mediated endocytosis. CONCLUSIONS: We demonstrated that LNCs can be directed towards immunosuppressive cells by simply modulating their size and charge thus providing a novel approach to exploit nanosystems for anticancer treatment in the frame of immunotherapy.


Asunto(s)
Antimetabolitos Antineoplásicos/química , Desoxicitidina/análogos & derivados , Glioblastoma/tratamiento farmacológico , Inmunosupresores/química , Lípidos/química , Macrófagos/metabolismo , Células Supresoras de Origen Mieloide/metabolismo , Nanocápsulas/química , Antimetabolitos Antineoplásicos/farmacología , Línea Celular Tumoral , Permeabilidad de la Membrana Celular , Desoxicitidina/química , Desoxicitidina/farmacología , Composición de Medicamentos , Endocitosis , Humanos , Inmunosupresores/farmacología , Inmunoterapia/métodos , Leucocitos/metabolismo , Tamaño de la Partícula , Transducción de Señal , Propiedades de Superficie , Gemcitabina
4.
Immunity ; 32(6): 790-802, 2010 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-20605485

RESUMEN

Tumor growth is associated with a profound alteration in myelopoiesis, leading to recruitment of immunosuppressive cells known as myeloid-derived suppressor cells (MDSCs). We showed that among factors produced by various experimental tumors, the cytokines GM-CSF, G-CSF, and IL-6 allowed a rapid generation of MDSCs from precursors present in mouse and human bone marrow (BM). BM-MDSCs induced by GM-CSF+IL-6 possessed the highest tolerogenic activity, as revealed by the ability to impair the priming of CD8(+) T cells and allow long term acceptance of pancreatic islet allografts. Cytokines inducing MDSCs acted on a common molecular pathway and the immunoregulatory activity of both tumor-induced and BM-derived MDSCs was entirely dependent on the C/EBPbeta transcription factor. Adoptive transfer of tumor antigen-specific CD8(+) T lymphocytes resulted in therapy of established tumors only in mice lacking C/EBPbeta in the myeloid compartment, suggesting that C/EBPbeta is a critical regulator of the immunosuppressive environment created by growing cancers.


Asunto(s)
Células de la Médula Ósea/inmunología , Proteína beta Potenciadora de Unión a CCAAT/inmunología , Tolerancia Inmunológica/inmunología , Neoplasias/inmunología , Escape del Tumor/inmunología , Traslado Adoptivo , Animales , Proteína beta Potenciadora de Unión a CCAAT/genética , Separación Celular , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Factor Estimulante de Colonias de Granulocitos/biosíntesis , Factor Estimulante de Colonias de Granulocitos/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Tolerancia Inmunológica/genética , Interleucina-6/biosíntesis , Interleucina-6/inmunología , Ratones , Neoplasias/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Escape del Tumor/genética
5.
J Immunol ; 198(10): 4166-4177, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28396317

RESUMEN

Myeloid cells play a key role in tumor progression and metastasis by providing nourishment and immune protection, as well as facilitating cancer invasion and seeding to distal sites. Although advances have been made in understanding the biology of these tumor-educated myeloid cells (TEMCs), their intrinsic plasticity challenges our further understanding of their biology. Indeed, in vitro experiments only mimic the in vivo setting, and current gene-knockout technologies do not allow the simultaneous, temporally controlled, and cell-specific silencing of multiple genes or pathways. In this article, we describe the 4PD nanoplatform, which allows the in vivo preferential transfection and in vivo tracking of TEMCs with the desired RNAs. This platform is based on the conjugation of CD124/IL-4Rα-targeting peptide with G5 PAMAM dendrimers as the loading surface and can convey therapeutic or experimental RNAs of interest. When injected i.v. in mice bearing CT26 colon carcinoma or B16 melanoma, the 4PD nanoparticles predominantly accumulate at the tumor site, transfecting intratumoral myeloid cells. The use of 4PD to deliver a combination of STAT3- and C/EBPß-specific short hairpin RNA or miR-142-3p confirmed the importance of these genes and microRNAs in TEMC biology and indicates that silencing of both genes is necessary to increase the efficacy of immune interventions. Thus, the 4PD nanoparticle can rapidly and cost effectively modulate and assess the in vivo function of microRNAs and mRNAs in TEMCs.


Asunto(s)
Dendrímeros/metabolismo , Silenciador del Gen , Células Mieloides/metabolismo , Nanotecnología/métodos , Animales , Línea Celular Tumoral , Neoplasias del Colon , Dendrímeros/administración & dosificación , Subunidad alfa del Receptor de Interleucina-4/inmunología , Subunidad alfa del Receptor de Interleucina-4/metabolismo , Melanoma Experimental , Ratones , MicroARNs , Células Mieloides/inmunología , Nanopartículas/administración & dosificación , Nanopartículas/metabolismo , Nanotecnología/normas , Receptores de Interleucina-4/inmunología , Receptores de Interleucina-4/metabolismo
6.
Biochim Biophys Acta ; 1865(1): 35-48, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26255541

RESUMEN

The incomplete clinical efficacy of anti-tumor immunotherapy can depend on the presence of an immunosuppressive environment in the host that supports tumor progression. Tumor-derived cytokines and growth factors induce an altered hematopoiesis that modifies the myeloid cell differentiation process, promoting proliferation and expansion of cells with immunosuppressive skills, namely myeloid derived suppressor cells (MDSCs). MDSCs promote tumor growth not only by shaping immune responses towards tumor tolerance, but also by supporting several processes necessary for the neoplastic progression such as tumor angiogenesis, cancer stemness, and metastasis dissemination. Thus, MDSC targeting represents a promising tool to eliminate host immune dysfunctions and increase the efficacy of immune-based cancer therapies.


Asunto(s)
Tolerancia Inmunológica , Células Mieloides/inmunología , Neoplasias/inmunología , Animales , Diferenciación Celular , Progresión de la Enfermedad , Humanos , Neoplasias/terapia , Pronóstico , Transducción de Señal
7.
Haematologica ; 102(5): 818-825, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28183849

RESUMEN

Bone marrow microenvironment is fundamental for hematopoietic homeostasis. Numerous efforts have been made to reproduce or manipulate its activity to facilitate engraftment after hematopoietic stem cell transplantation but clinical results remain unconvincing. This probably reflects the complexity of the hematopoietic niche. Recent data have demonstrated the fundamental role of stromal and myeloid cells in regulating hematopoietic stem cell self-renewal and mobilization in the bone marrow. In this study we unveil a novel interaction by which bone marrow mesenchymal stromal cells induce the rapid differentiation of CD11b+ myeloid cells from bone marrow progenitors. Such an activity requires the expression of nitric oxide synthase-2. Importantly, the administration of these mesenchymal stromal cell-educated CD11b+ cells accelerates hematopoietic reconstitution in bone marrow transplant recipients. We conclude that the liaison between mesenchymal stromal cells and myeloid cells is fundamental in hematopoietic homeostasis and suggests that it can be harnessed in clinical transplantation.


Asunto(s)
Células de la Médula Ósea/metabolismo , Diferenciación Celular , Hematopoyesis , Células Madre Mesenquimatosas/metabolismo , Células Mieloides/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Animales , Células de la Médula Ósea/citología , Antígeno CD11b/metabolismo , Autorrenovación de las Células , Trasplante de Células Madre Hematopoyéticas , Homeostasis , Humanos , Células Madre Mesenquimatosas/citología , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/citología , Óxido Nítrico Sintasa de Tipo II/genética
8.
Blood ; 123(5): 697-705, 2014 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-24335499

RESUMEN

The bromodomain and extraterminal (BET) protein BRD2-4 inhibitors hold therapeutic promise in preclinical models of hematologic malignancies. However, translation of these data to molecules suitable for clinical development has yet to be accomplished. Herein we expand the mechanistic understanding of BET inhibitors in multiple myeloma by using the chemical probe molecule I-BET151. I-BET151 induces apoptosis and exerts strong antiproliferative effect in vitro and in vivo. This is associated with contrasting effects on oncogenic MYC and HEXIM1, an inhibitor of the transcriptional activator P-TEFb. I-BET151 causes transcriptional repression of MYC and MYC-dependent programs by abrogating recruitment to the chromatin of the P-TEFb component CDK9 in a BRD2-4-dependent manner. In contrast, transcriptional upregulation of HEXIM1 is BRD2-4 independent. Finally, preclinical studies show that I-BET762 has a favorable pharmacologic profile as an oral agent and that it inhibits myeloma cell proliferation, resulting in survival advantage in a systemic myeloma xenograft model. These data provide a strong rationale for extending the clinical testing of the novel antimyeloma agent I-BET762 and reveal insights into biologic pathways required for myeloma cell proliferation.


Asunto(s)
Antineoplásicos/uso terapéutico , Benzodiazepinas/uso terapéutico , Compuestos Heterocíclicos de 4 o más Anillos/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Benzodiazepinas/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Ratones , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas de Unión al ARN/genética , Factores de Transcripción , Activación Transcripcional/efectos de los fármacos , Células Tumorales Cultivadas
9.
Cells ; 13(11)2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38891109

RESUMEN

Chondrosarcoma (ChS), a malignant cartilage-producing tumor, is the second most frequently diagnosed osseous sarcoma after osteosarcoma. It represents a very heterogeneous group of malignant chemo- and radiation-resistant neoplasms, accounting for approximately 20% of all bone sarcomas. The majority of ChS patients have a good prognosis after a complete surgical resection, as these tumors grow slowly and rarely metastasize. Conversely, patients with inoperable disease, due to the tumor location, size, or metastases, represent a great clinical challenge. Despite several genetic and epigenetic alterations that have been described in distinct ChS subtypes, very few therapeutic options are currently available for ChS patients. Therefore, new prognostic factors for tumor progression as well as new treatment options have to be explored, especially for patients with unresectable or metastatic disease. Recent studies have shown that a correlation between immune infiltrate composition, tumor aggressiveness, and survival does exist in ChS patients. In addition, the intra-tumor microvessel density has been proven to be associated with aggressive clinical behavior and a high metastatic potential in ChS. This review will provide an insight into the ChS microenvironment, since immunotherapy and antiangiogenic agents are emerging as interesting therapeutic options for ChS patients.


Asunto(s)
Condrosarcoma , Microambiente Tumoral , Humanos , Condrosarcoma/patología , Condrosarcoma/genética , Condrosarcoma/metabolismo , Neoplasias Óseas/patología , Neoplasias Óseas/terapia , Neoplasias Óseas/metabolismo , Neoplasias Óseas/genética , Inmunoterapia , Inhibidores de la Angiogénesis/uso terapéutico , Inhibidores de la Angiogénesis/farmacología
10.
Biomater Adv ; 161: 213881, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38749213

RESUMEN

Nanoparticle's success as drug delivery systems for cancer treatment has been achieved through passive targeting mechanisms. However, tumor heterogeneity and rapid drug clearance limit the treatment efficacy. Improved outcomes and selective drug release can be achieved by grafting ligands at the surface of nanocarriers that bind molecules overexpressed in the tumor microenvironment (TME). In this work, we developed a docetaxel-loaded nanoemulsions (NEs) binding an anti-netrin-1 monoclonal antibody (NP137) to selectively target the netrin-1 protein overexpressed in many different tumors. The goal is to refine a combined approach utilizing NP137 and docetaxel as an improved tumor-targeting chemotherapeutic agent for addressing triple-negative breast cancer (TNBC). Several factors have been considered for the optimization of the active targeted drug delivery system via the click-chemistry conjugation, as the impact of PEGylated surfactant that stabilize the NEs shell on conjugation efficiency, cytocompatibility with EMT6 cell line and colloidal stability over time of NEs. Results showed that a 660 Da PEG chain length contributed to NEs colloidal stability and had no impact on cell viability or on the antibody binding ability for its ligand after surface conjugation. Moreover, docetaxel was encapsulated into the oily core of NEs, with an encapsulation efficiency of 70 %. To validate our treatment strategy in vivo, the 4T1 murine breast cancer model was used. As a result, the comparison of active-targeted and non-targeted NEs revealed that only active-targeted NE could decrease the tumor growth rate.


Asunto(s)
Docetaxel , Nanopartículas , Neoplasias de la Mama Triple Negativas , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología , Animales , Femenino , Nanopartículas/química , Línea Celular Tumoral , Ratones , Docetaxel/farmacología , Docetaxel/uso terapéutico , Docetaxel/administración & dosificación , Humanos , Antineoplásicos/farmacología , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/uso terapéutico
11.
J Immunol ; 186(1): 264-74, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21135171

RESUMEN

The interaction between cancer vaccine adjuvants and myeloid-derived suppressor cells (MDSCs) is currently poorly understood. Very small size proteoliposomes (VSSP) are a nanoparticulated adjuvant under investigation in clinical trials in patients with renal carcinoma, breast cancer, prostate cancer, and cervical intraepithelial neoplasia grade III. We found that VSSP adjuvant induced a significant splenomegaly due to accumulation of CD11b(+)Gr-1(+) cells. However, VSSP-derived MDSCs showed a reduced capacity to suppress both allogeneic and Ag-specific CTL response compared with that of tumor-induced MDSCs. Moreover, splenic MDSCs isolated from tumor-bearing mice treated with VSSP were phenotypically more similar to those isolated from VSSP-treated tumor-free mice and much less suppressive than tumor-induced MDSCs, both in vitro and in vivo. Furthermore, different from dendritic cell vaccination, inoculation of VSSP-based vaccine in EG.7-OVA tumor-bearing mice was sufficient to avoid tumor-induced tolerance and stimulate an immune response against OVA Ag, similar to that observed in tumor-free mice. This effect correlated with an accelerated differentiation of MDSCs into mature APCs that was promoted by VSSP. VSSP used as a cancer vaccine adjuvant might thus improve antitumor efficacy not only by stimulating a potent immune response against tumor Ags but also by reducing tumor-induced immunosuppression.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Antineoplásicos/administración & dosificación , Vacunas contra el Cáncer/administración & dosificación , Linfoma/prevención & control , Células Mieloides/inmunología , Nanopartículas/administración & dosificación , Proteolípidos/administración & dosificación , Sarcoma Experimental/prevención & control , Animales , Proteínas de la Membrana Bacteriana Externa/administración & dosificación , Proteínas de la Membrana Bacteriana Externa/inmunología , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Femenino , Gangliósido G(M3)/administración & dosificación , Gangliósido G(M3)/inmunología , Inhibidores de Crecimiento/administración & dosificación , Linfoma/inmunología , Linfoma/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Mieloides/patología , Neisseria meningitidis/inmunología , Proteolípidos/inmunología , Sarcoma Experimental/inmunología , Sarcoma Experimental/patología
12.
Immunol Rev ; 222: 162-79, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18364001

RESUMEN

Emerging evidence indicates that the Achilles' heel of cancer immunotherapies is often the complex interplay of tumor-derived factors and deviant host properties, which involve a wide range of immune elements in the lymphoid and myeloid compartments. Regulatory lymphocytes, tumor-conditioned myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages, and dysfunctional and immature dendritic cells take part in a complex immunoregulatory network. Despite the fact that some mechanisms governing tumor-induced immune tolerance and suppression are starting to be better understood and their complexity dissected, little is known about the diachronic picture of immune tolerance. Based on observations of MDSCs, we present a time-structured and topologically consistent idea of tumor-dependent tolerance progression in tumor-bearing hosts.


Asunto(s)
Tolerancia Inmunológica , Activación de Linfocitos , Células Mieloides/inmunología , Neoplasias/inmunología , Linfocitos T Reguladores/inmunología , Escape del Tumor , Animales , Antígenos CD34/inmunología , Antígenos CD11/inmunología , Linfocitos T CD8-positivos/inmunología , Comunicación Celular , Regulación hacia Abajo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Terapia de Inmunosupresión , Subunidad alfa del Receptor de Interleucina-4/inmunología , Ratones , Modelos Inmunológicos , Mielopoyesis/inmunología , Neoplasias/patología , Óxido Nítrico Sintasa de Tipo II/inmunología , Transducción de Señal/inmunología
13.
J Exp Med ; 201(8): 1257-68, 2005 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-15824085

RESUMEN

Immunotherapy may provide valid alternative therapy for patients with hormone-refractory metastatic prostate cancer. However, if the tumor environment exerts a suppressive action on antigen-specific tumor-infiltrating lymphocytes (TIL), immunotherapy will achieve little, if any, success. In this study, we analyzed the modulation of TIL responses by the tumor environment using collagen gel matrix-supported organ cultures of human prostate carcinomas. Our results indicate that human prostatic adenocarcinomas are infiltrated by terminally differentiated cytotoxic T lymphocytes that are, however, in an unresponsive status. We demonstrate the presence of high levels of nitrotyrosines in prostatic TIL, suggesting a local production of peroxynitrites. By inhibiting the activity of arginase and nitric oxide synthase, key enzymes of L-arginine metabolism that are highly expressed in malignant but not in normal prostates, reduced tyrosine nitration and restoration of TIL responsiveness to tumor were achieved. The metabolic control exerted by the tumor on TIL function was confirmed in a transgenic mouse prostate model, which exhibits similarities with human prostate cancer. These results identify a novel and dominant mechanism by which cancers induce immunosuppression in situ and suggest novel strategies for tumor immunotherapy.


Asunto(s)
Adenocarcinoma/inmunología , Arginasa/antagonistas & inhibidores , Linfocitos T CD8-positivos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Neoplasias de la Próstata/inmunología , Subgrupos de Linfocitos T/inmunología , Tirosina/análogos & derivados , Adenocarcinoma/sangre , Adenocarcinoma/enzimología , Animales , Arginina/antagonistas & inhibidores , Arginina/metabolismo , Línea Celular Tumoral , Humanos , Interferón gamma/biosíntesis , Recuento de Linfocitos , Linfocitos Infiltrantes de Tumor/química , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Técnicas de Cultivo de Órganos , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/enzimología , Subgrupos de Linfocitos T/química , Tirosina/biosíntesis
14.
Eur J Immunol ; 40(1): 22-35, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19941314

RESUMEN

CD11b+/Gr-1+ myeloid-derived suppressor cells (MDSC) contribute to tumor immune evasion by restraining the activity of CD8+ T-cells. Two major MDSC subsets were recently shown to play an equal role in MDSC-induced immune dysfunctions: monocytic- and granulocytic-like. We isolated three fractions of MDSC, i.e. CD11b+/Gr-1high, CD11b+/Gr-1int, and CD11b+/Gr-1low populations that were characterized morphologically, phenotypically and functionally in different tumor models. In vitro assays showed that CD11b+/Gr-1int cell subset, mainly comprising monocytes and myeloid precursors, was always capable to suppress CD8+ T-cell activation, while CD11b+/Gr-1high cells, mostly granulocytes, exerted appreciable suppression only in some tumor models and when present in high numbers. The CD11b+/Gr-1int but not CD11b+/Gr-1high cells were also immunosuppressive in vivo following adoptive transfer. CD11b+/Gr-1low cells retained the immunosuppressive potential in most tumor models. Gene silencing experiments indicated that GM-CSF was necessary to induce preferential expansion of both CD11b+/Gr-1int and CD11b+/Gr-1low subsets in the spleen of tumor-bearing mice and mediate tumor-induced tolerance whereas G-CSF, which preferentially expanded CD11b+/Gr-1high cells, did not create such immunosuppressive environment. GM-CSF also acted on granulocyte-macrophage progenitors in the bone marrow inducing local expansion of CD11b+/Gr-1low cells. These data unveil a hierarchy of immunoregulatory activity among MDSC subsets that is controlled by tumor-released GM-CSF.


Asunto(s)
Antígeno CD11b/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Tolerancia Inmunológica , Células Mieloides/inmunología , Receptores de Quimiocina/inmunología , Animales , Diferenciación Celular , Línea Celular Tumoral , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Mieloides/citología , Trasplante de Neoplasias , Neoplasias/inmunología , Interferencia de ARN
15.
Front Immunol ; 12: 718098, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34675917

RESUMEN

Reactive oxygen species, including RNS, contribute to the control of multiple immune cell functions within the tumor microenvironment (TME). Tumor-infiltrating myeloid cells (TIMs) represent the archetype of tolerogenic cells that actively contribute to dismantle effective immunity against cancer. TIMs inhibit T cell functions and promote tumor progression by several mechanisms including the amplification of the oxidative/nitrosative stress within the TME. In tumors, TIM expansion and differentiation is regulated by the granulocyte-macrophage colony-stimulating factor (GM-CSF), which is produced by cancer and immune cells. Nevertheless, the role of GM-CSF in tumors has not yet been fully elucidated. In this study, we show that GM-CSF activity is significantly affected by RNS-triggered post-translational modifications. The nitration of a single tryptophan residue in the sequence of GM-CSF nourishes the expansion of highly immunosuppressive myeloid subsets in tumor-bearing hosts. Importantly, tumors from colorectal cancer patients express higher levels of nitrated tryptophan compared to non-neoplastic tissues. Collectively, our data identify a novel and selective target that can be exploited to remodel the TME and foster protective immunity against cancer.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo , Neoplasias/etiología , Neoplasias/metabolismo , Procesamiento Proteico-Postraduccional , Animales , Biomarcadores , Diferenciación Celular , Línea Celular Tumoral , Citocinas/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Inmunomodulación , Ratones , Neoplasias/patología , Especies de Nitrógeno Reactivo/metabolismo , Transducción de Señal , Microambiente Tumoral/inmunología
16.
Front Immunol ; 11: 1590, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32793228

RESUMEN

Immune checkpoint inhibitors are becoming standard treatments in several cancer types, profoundly changing the prognosis of a fraction of patients. Currently, many efforts are being made to predict responders and to understand how to overcome resistance in non-responders. Given the crucial role of myeloid cells as modulators of T effector cell function in tumors, it is essential to understand their impact on the clinical outcome of immune checkpoint blockade and on the mechanisms of immune evasion. In this review we focus on the existing clinical evidence of the relation between the presence of myeloid cell subsets and the response to anti-PD(L)1 and anti-CTLA-4 treatment. We highlight how circulating and tumor-infiltrating myeloid populations can be used as predictive biomarkers for immune checkpoint inhibitors in different human cancers, both at baseline and on treatment. Moreover, we propose to follow the dynamics of myeloid cells during immunotherapy as pharmacodynamic biomarkers. Finally, we provide an overview of the current strategies tested in the clinic that use myeloid cell targeting together with immune checkpoint blockade with the aim of uncovering the most promising approaches for effective combinations.


Asunto(s)
Biomarcadores , Inhibidores de Puntos de Control Inmunológico/farmacología , Proteínas de Punto de Control Inmunitario/metabolismo , Células Mieloides/metabolismo , Animales , Estudios Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Terapia Molecular Dirigida , Células Mieloides/efectos de los fármacos , Células Mieloides/inmunología , Células Supresoras de Origen Mieloide/efectos de los fármacos , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo , Especificidad de Órganos/genética , Especificidad de Órganos/inmunología , Resultado del Tratamiento
17.
Eur J Pharm Biopharm ; 150: 1-13, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32113915

RESUMEN

This work here presented provides insights over a novel biodegradable polymeric nanosystem made of hyaluronic acid and polyarginine for diaminocyclohexane-platinum (DACHPt) encapsulation. Using mild conditions based on ionic gelation technique, monodispersed blank and DACHPt-loaded nanoparticles (NP) with a size of around 200 nm and negative ζ potential (-35 mV) were obtained. The freeze-drying process was optimized to improve the stability and shelf-life of the developed nanoparticles. After reconstitution, nanoparticles maintained their size showing an association efficiency of around 70% and a high drug loading (8%). In vitro cytotoxicity studies revealed that DACHPt-loaded nanoparticles had a superior anticancer activity compared with oxaliplatin solution. The IC50 was reduced by a factor of two in HT-29 cells (IC50 39 µM vs 74 µM, respectively), and resulted almost 1.3 fold lower in B6KPC3 cells (18 µM vs 23 µM respectively). Whereas toxic effects of both drug and DACHPt-loaded nanoparticles were comparable in the A549 cell line (IC50 11 µM vs 12 µM). DACHPt-loaded nanoparticles were also able to modulate immunogenic cell death (ICD) in vitro. After incubation with B6KPC3 cells, an increase in HMGB1 (high-mobility group box 1) production associated with ATP release occurred. Then, in vivo pharmacokinetic studies were performed after intravenous injection (IV) of DACHPt-loaded nanoparticles and oxaliplatin solution in healthy mice (35.9 µg of platinum equivalent/mouse). An AUC six times higher (24 h * mg/L) than the value obtained following the administration of oxaliplatin solution (3.76 h * mg/L) was found. Cmax was almost five times higher than the control (11.4 mg/L for NP vs 2.48 mg/L). Moreover, the reduction in volume of distribution and clearance clearly indicated a more limited tissue distribution. A simulated repeated IV regimen was performed in silico and showed no accumulation of platinum from the nanoparticles. Overall, the proposed approach discloses a novel nano-oncological treatment based on platinum derivative with improved antitumor activity in vitro and in vivo stability as compared to the free drug.


Asunto(s)
Antineoplásicos/administración & dosificación , Portadores de Fármacos , Ácido Hialurónico/química , Nanopartículas , Oxaliplatino/administración & dosificación , Péptidos/química , Células A549 , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Supervivencia Celular/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , Composición de Medicamentos , Estabilidad de Medicamentos , Femenino , Células HT29 , Humanos , Inyecciones Intravenosas , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Ratones Endogámicos C57BL , Modelos Biológicos , Oxaliplatino/química , Oxaliplatino/farmacocinética , Distribución Tisular
18.
J Control Release ; 324: 228-237, 2020 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-32413454

RESUMEN

Poly(L-glutamic acid)-co-poly(ethylene glycol) block copolymers (PLE-PEG) are here investigated as polymers for conjugation to therapeutic proteins such as granulocyte colony stimulating factor (G-CSF) and human growth hormone (hGH). PLE-PEG block copolymers are able to stabilize and protect proteins from degradation and to prolong their residence time in the blood stream, features that are made possible thanks to PEG's intrinsic properties and the simultaneous presence of the biodegradable anionic PLE moiety. When PLE-PEG copolymers are selectively tethered to the N-terminus of G-CSF and hGH, they yield homogeneous monoconjugates that preserve the protein's secondary structure. During the current study the pharmacokinetics of PLE10-PEG20k-G-CSF and PLE20-PEG20k-G-CSF derivatives and their ability to induce granulopoiesis were, respectively, assessed in Sprague-Dawley rats and in C57BL6 mice. Our results show that the bioavailability and bioactivity of the derivatives are comparable to or better than those of PEG20k-Nter-G-CSF (commercially known as Pegfilgrastim). The therapeutic effects of PLE10-PEG20k-hGH and PLE20-PEG20k-hGH derivatives tested in hypophysectomized rats demonstrate that the presence of a negatively charged PLE block enhances the biological properties of the conjugates additionally with respect to PEG20k-Nter-hGH.


Asunto(s)
Ácido Glutámico , Polietilenglicoles , Animales , Ratones , Ratones Endogámicos C57BL , Polímeros , Ratas , Ratas Sprague-Dawley
19.
Cancer Discov ; 10(11): 1758-1773, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32651166

RESUMEN

Tumor-associated macrophages (TAM) are regulators of extracellular matrix (ECM) remodeling and metastatic progression, the main cause of cancer-associated death. We found that disabled homolog 2 mitogen-responsive phosphoprotein (DAB2) is highly expressed in tumor-infiltrating TAMs and that its genetic ablation significantly impairs lung metastasis formation. DAB2-expressing TAMs, mainly localized along the tumor-invasive front, participate in integrin recycling, ECM remodeling, and directional migration in a tridimensional matrix. DAB2+ macrophages escort the invasive dissemination of cancer cells by a mechanosensing pathway requiring the transcription factor YAP. In human lobular breast and gastric carcinomas, DAB2+ TAMs correlated with a poor clinical outcome, identifying DAB2 as potential prognostic biomarker for stratification of patients with cancer. DAB2 is therefore central for the prometastatic activity of TAMs. SIGNIFICANCE: DAB2 expression in macrophages is essential for metastasis formation but not primary tumor growth. Mechanosensing cues, activating the complex YAP-TAZ, regulate DAB2 in macrophages, which in turn controls integrin recycling and ECM remodeling in 3-D tissue matrix. The presence of DAB2+ TAMs in patients with cancer correlates with worse prognosis.This article is highlighted in the In This Issue feature, p. 1611.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Neoplasias/genética , Macrófagos Asociados a Tumores/metabolismo , Línea Celular Tumoral , Humanos
20.
J Clin Invest ; 116(10): 2777-90, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17016559

RESUMEN

Active suppression of tumor-specific T lymphocytes can limit the efficacy of immune surveillance and immunotherapy. While tumor-recruited CD11b+ myeloid cells are known mediators of tumor-associated immune dysfunction, the true nature of these suppressive cells and the fine biochemical pathways governing their immunosuppressive activity remain elusive. Here we describe a population of circulating CD11b+IL-4 receptor alpha+ (CD11b+IL-4Ralpha+), inflammatory-type monocytes that is elicited by growing tumors and activated by IFN-gamma released from T lymphocytes. CD11b+IL-4Ralpha+ cells produced IL-13 and IFN-gamma and integrated the downstream signals of these cytokines to trigger the molecular pathways suppressing antigen-activated CD8+ T lymphocytes. Analogous immunosuppressive circuits were active in CD11b+ cells present within the tumor microenvironment. These suppressor cells challenge the current idea that tumor-conditioned immunosuppressive monocytes/macrophages are alternatively activated. Moreover, our data show how the inflammatory response elicited by tumors had detrimental effects on the adaptive immune system and suggest novel approaches for the treatment of tumor-induced immune dysfunctions.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Monocitos/inmunología , Neoplasias/inmunología , Animales , Arginasa/genética , Arginasa/metabolismo , Antígeno CD11b/análisis , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Expresión Génica/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Inmunidad Celular/inmunología , Interferón gamma/genética , Interferón gamma/metabolismo , Interferón gamma/farmacología , Interleucina-13/metabolismo , Interleucina-13/farmacología , Interleucina-4/genética , Interleucina-4/metabolismo , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Modelos Inmunológicos , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Células Mieloides/efectos de los fármacos , Células Mieloides/inmunología , Células Mieloides/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Receptores de Superficie Celular/análisis , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Bazo/citología , Bazo/inmunología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA