Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Methods Mol Biol ; 2614: 81-91, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36587120

RESUMO

Tumor-associated macrophages (TAMs) play a key immunosuppressive role that limits the ability of the immune system to fight cancer and hinder the anti-tumoral efficacy of most treatments currently applied in the clinic. However, a key feature of macrophages is their phenotypical and functional plasticity, which called their attention as promising targets for therapeutic intervention based on their elimination or reprogramming toward M1-like cytotoxic effector cells, with anti-tumor functions. This polarization status of macrophages can be studied in terms of molecular markers and functional activities, using an appropriate combination of experimental methodologies, both in vitro and in vivo. Here we focus on describing in vitro protocols to isolate primary monocytes from buffy coats and to study macrophage phenotype and function, after exposure to new therapies, by a combination of flow cytometry, RT-PCR, and ELISA analysis. We also provide the methodology to evaluate in vitro the cytotoxic activity of treated macrophages toward cancer cells.


Assuntos
Neoplasias , Humanos , Monócitos , Macrófagos , Fenótipo , Biomarcadores , Microambiente Tumoral
2.
Drug Deliv Transl Res ; 13(7): 1896-1911, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36472784

RESUMO

Tumor-associated macrophages (TAMs), a class of immune cells that play a key role in tumor immunosuppression, are recognized as important targets to improve cancer prognosis and treatment. Consequently, the engineering of drug delivery nanocarriers that can reach TAMs has acquired special relevance. This work describes the development and biological evaluation of a panel of hyaluronic acid (HA) nanocapsules (NCs), with different compositions and prepared by different techniques, designed to target macrophages. The results showed that plain HA NCs did not significantly influence the polarization of M0 and M2-like macrophages towards an M1-like pro-inflammatory phenotype; however, the chemical functionalization of HA with mannose (HA-Man) led to a significant increase of NCs uptake by M2 macrophages in vitro and to an improved biodistribution in a MN/MNCA1 fibrosarcoma mouse model with high infiltration of TAMs. These functionalized HA-Man NCs showed a higher accumulation in the tumor compared to non-modified HA NCs. Finally, the pre-administration of the liposomal liver occupying agent Nanoprimer™ further increased the accumulation of the HA-Man NCs in the tumor. This work highlights the promise shown by the HA-Man NCs to target TAMs and thus provides new options for the development of nanomedicine and immunotherapy-based cancer treatments.


Assuntos
Nanocápsulas , Neoplasias , Camundongos , Animais , Nanocápsulas/química , Ácido Hialurônico/química , Manose , Macrófagos Associados a Tumor/patologia , Distribuição Tecidual , Neoplasias/patologia
3.
Front Immunol ; 14: 1334800, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38259462

RESUMO

Background: In the tumor microenvironment (TME), tumor-associated macrophages (TAMs) play a key immunosuppressive role that limits the ability of the immune system to fight cancer. Toll-like receptors (TLRs) ligands, such as poly(I:C) or resiquimod (R848) are able to reprogram TAMs towards M1-like antitumor effector cells. The objective of our work has been to develop and evaluate polymeric nanocapsules (NCs) loaded with poly(I:C)+R848, to improve drug stability and systemic toxicity, and evaluate their targeting and therapeutic activity towards TAMs in the TME of solid tumors. Methods: NCs were developed by the solvent displacement and layer-by-layer methodologies and characterized by dynamic light scattering and nanoparticle tracking analysis. Hyaluronic acid (HA) was chemically functionalized with mannose for the coating of the NCs to target TAMs. NCs loaded with TLR ligands were evaluated in vitro for toxicity and immunostimulatory activity by Alamar Blue, ELISA and flow cytometry, using primary human monocyte-derived macrophages. For in vivo experiments, the CMT167 lung cancer model and the MN/MCA1 fibrosarcoma model metastasizing to lungs were used; tumor-infiltrating leukocytes were evaluated by flow cytometry and multispectral immunophenotyping. Results: We have developed polymeric NCs loaded with poly(I:C)+R848. Among a series of 5 lead prototypes, protamine-NCs were selected based on their physicochemical properties (size, charge, stability) and in vitro characterization, showing good biocompatibility on primary macrophages and ability to stimulate their production of T-cell attracting chemokines (CXCL10, CCL5) and to induce M1-like macrophages cytotoxicity towards tumor cells. In mouse tumor models, the intratumoral injection of poly(I:C)+R848-protamine-NCs significantly prevented tumor growth and lung metastasis. In an orthotopic murine lung cancer model, the intravenous administration of poly(I:C)+R848-prot-NCs, coated with an additional layer of HA-mannose to improve TAM-targeting, resulted in good antitumoral efficacy with no apparent systemic toxicity. While no significant alterations were observed in T cell numbers (CD8, CD4 or Treg), TAM-reprogramming in treated mice was confirmed by the relative decrease of interstitial versus alveolar macrophages, having higher CD86 expression but lower CD206 and Arg1 expression in the same cells, in treated mice. Conclusion: Mannose-HA-protamine-NCs loaded with poly(I:C)+R848 successfully reprogram TAMs in vivo, and reduce tumor progression and metastasis spread in mouse tumors.


Assuntos
Imidazóis , Neoplasias Pulmonares , Nanocápsulas , Humanos , Animais , Camundongos , Macrófagos Associados a Tumor , Manose , Neoplasias Pulmonares/tratamento farmacológico , Modelos Animais de Doenças , Protaminas , Microambiente Tumoral
4.
Biomedicines ; 10(7)2022 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-35884895

RESUMO

Toll-like receptors (TLRs) are natural initial triggers of innate and adaptive immune responses. With the advent of cancer immunotherapy, nucleic acids engineered as ligands of endosomal TLRs have been investigated for the treatment of solid tumors. Despite promising results, their systemic administration, similarly to other immunotherapies, raises safety issues. To overcome these problems, recent studies have applied the direct injection of endosomal TLR agonists in the tumor and/or draining lymph nodes, achieving high local drug exposure and strong antitumor response. Importantly, intratumoral delivery of TLR agonists showed powerful effects not only against the injected tumors but also often against uninjected lesions (abscopal effects), resulting in some cases in cure and antitumoral immunological memory. Herein, we describe the structure and function of TLRs and their role in the tumor microenvironment. Then, we provide our vision on the potential of intratumor versus systemic delivery or vaccination approaches using TLR agonists, also considering the use of nanoparticles to improve their targeting properties. Finally, we collect the preclinical and clinical studies applying intratumoral injection of TLR agonists as monotherapies or in combination with: (a) other TLR or STING agonists; (b) other immunotherapies; (c) radiotherapy or chemotherapy; (d) targeted therapies.

5.
J Immunother Cancer ; 9(9)2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34531246

RESUMO

BACKGROUND: Tumor-associated macrophages (TAMs) play a key immunosuppressive role that limits the ability of the immune system to fight cancer and hinder the antitumoral efficacy of most treatments currently applied in the clinic. Previous studies have evaluated the antitumoral immune response triggered by (TLR) agonists, such as poly(I:C), imiquimod (R837) or resiquimod (R848) as monotherapies; however, their combination for the treatment of cancer has not been explored. This study investigates the antitumoral efficacy and the macrophage reprogramming triggered by poly(I:C) combined with R848 or with R837, versus single treatments. METHODS: TLR agonist treatments were evaluated in vitro for toxicity and immunostimulatory activity by Alamar Blue, ELISA and flow cytometry using primary human and murine M-CSF-differentiated macrophages. Cytotoxic activity of TLR-treated macrophages toward cancer cells was evaluated with an in vitro functional assay by flow cytometry. For in vivo experiments, the CMT167 lung cancer model and the MN/MCA1 fibrosarcoma model metastasizing to lungs were used; tumor-infiltrating leukocytes were evaluated by flow cytometry, RT-qPCR, multispectral immunophenotyping, quantitative proteomic experiments, and protein-protein interaction analysis. RESULTS: Results demonstrated the higher efficacy of poly(I:C) combined with R848 versus single treatments or combined with R837 to polarize macrophages toward M1-like antitumor effectors in vitro. In vivo, the intratumoral synergistic combination of poly(I:C)+R848 significantly prevented tumor growth and metastasis in lung cancer and fibrosarcoma immunocompetent murine models. Regressing tumors showed increased infiltration of macrophages with a higher M1:M2 ratio, recruitment of CD4+ and CD8+ T cells, accompanied by a reduction of immunosuppressive CD206+ TAMs and FOXP3+/CD4+ T cells. The depletion of both CD4+ and CD8+ T cells resulted in complete loss of treatment efficacy. Treated mice acquired systemic antitumoral response and resistance to tumor rechallenge mediated by boosted macrophage cytotoxic activity and T-cell proliferation. Proteomic experiments validate the superior activation of innate immunity by poly(I:C)+R848 combination versus single treatments or poly(I:C)+R837, and protein-protein-interaction network analysis reveal the key activation of the STAT1 pathway. DISCUSSION: These findings demonstrate the antitumor immune responses mediated by macrophage activation on local administration of poly(I:C)+R848 combination and support the intratumoral application of this therapy to patients with solid tumors in the clinic.


Assuntos
Antivirais/uso terapêutico , Terapia Combinada/métodos , Imidazóis/uso terapêutico , Imunoterapia/métodos , Neoplasias/tratamento farmacológico , Poli I-C/uso terapêutico , Macrófagos Associados a Tumor/metabolismo , Animais , Antivirais/farmacologia , Linhagem Celular Tumoral , Sinergismo Farmacológico , Humanos , Imidazóis/farmacologia , Camundongos , Poli I-C/farmacologia
6.
Nanomaterials (Basel) ; 11(4)2021 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-33808372

RESUMO

Engineered nanomaterials are potentially very useful for a variety of applications, but studies are needed to ascertain whether these materials pose a risk to human health. Here, we studied three benchmark nanomaterials (Ag nanoparticles, TiO2 nanoparticles, and multi-walled carbon nanotubes, MWCNTs) procured from the nanomaterial repository at the Joint Research Centre of the European Commission. Having established a sub-lethal concentration of these materials using two human cell lines representative of the immune system and the lungs, respectively, we performed RNA sequencing of the macrophage-like cell line after exposure for 6, 12, and 24 h. Downstream analysis of the transcriptomics data revealed significant effects on chemokine signaling pathways. CCR2 was identified as the most significantly upregulated gene in MWCNT-exposed cells. Using multiplex assays to evaluate cytokine and chemokine secretion, we could show significant effects of MWCNTs on several chemokines, including CCL2, a ligand of CCR2. The results demonstrate the importance of evaluating sub-lethal concentrations of nanomaterials in relevant target cells.

7.
Clin Cancer Res ; 27(12): 3291-3297, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-33531428

RESUMO

The stroma of most solid tumors is populated by myeloid cells, which mostly represent macrophages. Tumor-associated macrophages (TAMs), strongly influenced by cancer cell-derived factors, are key drivers of immunosuppression and support tumor growth and spread to distant sites. Their accurate quantification and characterization in the tumor microenvironment are gaining prognostic value: increasing evidence demonstrates their ability to hamper cancer patients' response to chemotherapy, as well as to immunotherapies based on checkpoint inhibition. Therefore, strategies to counteract their negative effects are nowadays gaining momentum at preclinical, translational, and clinical levels. Our knowledge of the biology of TAMs has greatly advanced in the last years; several strategies to target and reprogram their functions to become antitumor effectors have proven successful in experimental preclinical tumor models; on the other hand, few approaches have so far been effectively translated into clinic practice. A growing interest in the therapeutic manipulation of TAMs is evidenced by numerous early-phase clinical trials, which are continuously fueled by new discoveries from basic research. This gives us hope that the targeting and sustained reprogramming of TAMs will be more specific to synergize with current therapies and maximize antitumor responses in patients.


Assuntos
Neoplasias , Macrófagos Associados a Tumor , Humanos , Imunoterapia , Macrófagos/patologia , Neoplasias/tratamento farmacológico , Microambiente Tumoral
8.
Cells ; 9(1)2019 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-31878087

RESUMO

: Established evidence demonstrates that tumor-infiltrating myeloid cells promote rather than stop-cancer progression. Tumor-associated macrophages (TAMs) are abundantly present at tumor sites, and here they support cancer proliferation and distant spreading, as well as contribute to an immune-suppressive milieu. Their pro-tumor activities hamper the response of cancer patients to conventional therapies, such as chemotherapy or radiotherapy, and also to immunotherapies based on checkpoint inhibition. Active research frontlines of the last years have investigated novel therapeutic strategies aimed at depleting TAMs and/or at reprogramming their tumor-promoting effects, with the goal of re-establishing a favorable immunological anti-tumor response within the tumor tissue. In recent years, numerous clinical trials have included pharmacological strategies to target TAMs alone or in combination with other therapies. This review summarizes the past and current knowledge available on experimental tumor models and human clinical studies targeting TAMs for cancer treatment.


Assuntos
Imunoterapia/métodos , Macrófagos/imunologia , Neoplasias/imunologia , Neoplasias/terapia , Animais , Transformação Celular Neoplásica/imunologia , Humanos , Imunoterapia/tendências , Macrófagos/patologia , Neoplasias/patologia , Microambiente Tumoral/imunologia
9.
Semin Immunol ; 34: 103-113, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28941641

RESUMO

The engineering of new nanomedicines with ability to target and kill or re-educate Tumor Associated Macrophages (TAMs) stands up as a promising strategy to induce the effective switching of the tumor-promoting immune suppressive microenvironment, characteristic of tumors rich in macrophages, to one that kills tumor cells, is anti-angiogenic and promotes adaptive immune responses. Alternatively, the loading of monocytes/macrophages in blood circulation with nanomedicines, may be used to profit from the high infiltration ability of myeloid cells and to allow the drug release in the bulk of the tumor. In addition, the development of TAM-targeted imaging nanostructures, can be used to study the macrophage content in solid tumors and, hence, for a better diagnosis and prognosis of cancer disease. The major challenges for the effective targeting of TAM with nanomedicines and their application in the clinic have already been identified. These challenges are associated to the undesirable clearance of nanomedicines by, the mononuclear phagocyte system (macrophages) in competing organs (liver, lung or spleen), upon their intravenous injection; and also to the difficult penetration of nanomedicines across solid tumors due to the abnormal vasculature and the excessive extracellular matrix present in stromal tumors. In this review we describe the recent nanotechnology-base strategies that have been developed to target macrophages in tumors.


Assuntos
Imunoterapia/métodos , Macrófagos/imunologia , Sistema Fagocitário Mononuclear , Nanopartículas/uso terapêutico , Nanoestruturas/estatística & dados numéricos , Neoplasias/imunologia , Animais , Citotoxicidade Imunológica , Sistemas de Liberação de Medicamentos , Humanos , Nanomedicina , Microambiente Tumoral
10.
Adv Drug Deliv Rev ; 65(15): 2087-97, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23751779

RESUMO

Carbon nanotubes have gained tremendous interest in a wide range of applications due to their unique physical, chemical, and electronic properties. Needless to say, close attention to the potential toxicity of carbon nanotubes is of paramount importance. Numerous studies have linked exposure of carbon nanotubes to the induction of inflammation, a complex protective response to harmful stimuli including pathogens, damaged or dying cells, and other irritants. However, inflammation is a double-edged sword as chronic inflammation can lead to destruction of tissues thus compromising the homeostasis of the organism. Here, we provide an overview of the process of inflammation, the key cells and the soluble mediators involved, and discuss research on carbon nanotubes and inflammation, including recent studies on the activation of the so-called inflammasome complex in macrophages resulting in secretion of pro-inflammatory cytokines. Moreover, recent work has shown that inflammatory cells i.e. neutrophils and eosinophils are capable of enzymatic degradation of carbon nanotubes, with mitigation of the pro-inflammatory and pro-fibrotic effects of nanotubes thus underscoring that inflammation is both good and bad.


Assuntos
Macrófagos/metabolismo , Nanotubos de Carbono/toxicidade , Pneumonia/induzido quimicamente , Animais , Doença Crônica , Citocinas/metabolismo , Eosinófilos/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Neutrófilos/metabolismo , Pneumonia/patologia
11.
Acc Chem Res ; 46(3): 733-42, 2013 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-22720979

RESUMO

Engineered nanomaterials offer numerous and tantalizing opportunities in many sectors of society, including medicine. Needless to say, attention should also be paid to the potential for unexpected hazardous effects of these novel materials. To date, much of the nanotoxicology literature has focused on the assessment of cell viability or cell death using primitive assays for the detection of plasma membrane integrity or mitochondrial function or assessment of cellular morphology. However, when assessing the cytotoxic effects of engineered nanomaterials, researchers need not only to consider whether cells are dead or alive but also to assess which of the numerous, highly specific pathways of cell death might be involved. Moreover, it is important to diagnose cell death based not only on morphological markers but on the assessment and quantification of biochemical alterations specific to each form of cell death. In this Account, we provide a description of the three major forms of programmed cell death in mammalian cells: apoptosis, autophagic cell death, and regulated necrosis, sometimes referred to as necroptosis. Apoptosis can be activated via the extrinsic (death receptor-dependent) or via the intrinsic (mitochondria-dependent) route. Apoptotic cell death may or may not require the activation of cytosolic proteases known as caspases. Autophagy (self-eating) has an important homeostatic role in the cell, mediating the removal of dysfunctional or damaged organelles thereby allowing the recycling of cellular building blocks. However, unrestrained autophagy can kill cells. Studies in recent years have revealed that necrosis that depends on activation of the kinases RIP1 and RIP3 is a major form of programmed cell death with roles in development and immunity. We also discuss recent examples of the impact of engineered nanoparticles on the three different pathways of programmed cell death. For example, acute exposure of cells to carbon nanotubes (CNTs) can induce apoptosis whereas chronic exposure to CNTs may yield an apoptosis-resistant and tumorigenic phenotype in lung epithelial cells. Several reports show that nanoparticles, including polystyrene particles, are routed to the lysosomal compartment and trigger cell death through the destabilization of lysosomal membranes with engagement of the intrinsic apoptosis pathway. In addition, a number of studies have demonstrated that nanomaterials such as CNTs, quantum dots, and gold nanoparticles can affect cellular autophagy. An improved understanding of the complexities of the nanomaterial-induced perturbation of different cell death pathways may allow for a better prediction of the consequences of human exposure.


Assuntos
Nanoestruturas/química , Segurança , Animais , Morte Celular/efeitos dos fármacos , Humanos , Modelos Biológicos , Nanoestruturas/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...