Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Oncoimmunology ; 12(1): 2201147, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37089449

RESUMO

The clinical successes of immune checkpoint blockade (ICB) in advanced cancer patients have recently spurred the clinical implementation of ICB in the neoadjuvant and perioperative setting. However, how neoadjuvant ICB therapy affects the systemic immune landscape and metastatic spread remains to be established. Tumors promote both local and systemic expansion of regulatory T cells (Tregs), which are key orchestrators of tumor-induced immunosuppression, contributing to immune evasion, tumor progression and metastasis. Tregs express inhibitory immune checkpoint molecules and thus may be unintended targets for ICB therapy counteracting its efficacy. Using ICB-refractory models of spontaneous primary and metastatic breast cancer that recapitulate the poor ICB response of breast cancer patients, we observed that combined anti-PD-1 and anti-CTLA-4 therapy inadvertently promotes proliferation and activation of Tregs in the tumor, tumor-draining lymph node and circulation. Also in breast cancer patients, Treg levels were elevated upon ICB. Depletion of Tregs during neoadjuvant ICB in tumor-bearing mice not only reshaped the intratumoral immune landscape into a state favorable for ICB response but also induced profound and persistent alterations in systemic immunity, characterized by elevated CD8+ T cells and NK cells and durable T cell activation that was maintained after treatment cessation. While depletion of Tregs in combination with neoadjuvant ICB did not inhibit primary tumor growth, it prolonged metastasis-related survival driven predominantly by CD8+ T cells. This study demonstrates that neoadjuvant ICB therapy of breast cancer can be empowered by simultaneous targeting of Tregs, extending metastasis-related survival, independent of a primary tumor response.


Assuntos
Neoplasias da Mama , Ativação Linfocitária , Linfócitos T Reguladores , Humanos , Neoplasias da Mama/imunologia , Neoplasias da Mama/terapia , Linfócitos T Reguladores/imunologia , Terapia Neoadjuvante , Inibidores de Checkpoint Imunológico/uso terapêutico , Células Matadoras Naturais/imunologia , Células Mieloides/imunologia , Metástase Neoplásica , Animais , Camundongos , Linfócitos T CD8-Positivos/imunologia
2.
Blood Adv ; 7(11): 2388-2400, 2023 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-36920782

RESUMO

Cancer enhances the risk of venous thromboembolism, but a hypercoagulant microenvironment also promotes cancer progression. Although anticoagulants have been suggested as a potential anticancer treatment, clinical studies on the effect of such modalities on cancer progression have not yet been successful for unknown reasons. In normal physiology, complex formation between the subendothelial-expressed tissue factor (TF) and the blood-borne liver-derived factor VII (FVII) results in induction of the extrinsic coagulation cascade and intracellular signaling via protease-activated receptors (PARs). In cancer, TF is overexpressed and linked to poor prognosis. Here, we report that increased levels of FVII are also observed in breast cancer specimens and are associated with tumor progression and metastasis to the liver. In breast cancer cell lines, tumor-expressed FVII drives changes reminiscent of epithelial-to-mesenchymal transition (EMT), tumor cell invasion, and expression of the prometastatic genes, SNAI2 and SOX9. In vivo, tumor-expressed FVII enhanced tumor growth and liver metastasis. Surprisingly, liver-derived FVII appeared to inhibit metastasis. Finally, tumor-expressed FVII-induced prometastatic gene expression independent of TF but required a functional endothelial protein C receptor, whereas recombinant activated FVII acting via the canonical TF:PAR2 pathway inhibited prometastatic gene expression. Here, we propose that tumor-expressed FVII and liver-derived FVII have opposing effects on EMT and metastasis.


Assuntos
Neoplasias da Mama , Humanos , Feminino , Neoplasias da Mama/genética , Transdução de Sinais , Tromboplastina/genética , Tromboplastina/metabolismo , Microambiente Tumoral
3.
Cells ; 11(15)2022 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-35954190

RESUMO

Neutrophils are not only crucial immune cells for the neutralization of pathogens during infections, but they are also key players in tissue repair and cancer. Several methods are available to investigate the in vivo role of neutrophils in these conditions, including the depletion of neutrophils with neutralizing antibodies against Ly6G, or the blockade of neutrophil recruitment with CXCR2 inhibitors. A limited number of transgenic mouse models were generated that rely on the disruption of genes important for neutrophil development or on the injection of diphtheria toxin to induce neutrophil ablation. However, these methods have various limitations, including a lack of neutrophil specificity, a lack of long-term efficacy, or a lack of the ability to conditionally deplete neutrophils. Therefore, we generated a transgenic mouse model for the inducible and reversible ablation of neutrophils using the ATTAC (Apoptosis Through Targeted Activation of Caspase 8) approach. With the ATTAC strategy, which relies on the expression of the caspase 8-FKBP fusion protein, apoptosis is induced upon administration of a chemical dimerizer (FK506 analogue) that facilitates the dimerization and activation of caspase 8. In order to achieve specific neutrophil depletion, we cloned the ATTAC construct under the human migration inhibitory factor-related protein 8 (hMRP8) promotor. The newly generated hMRP8-ATTAC mice expressed high levels of the transgene in neutrophils, and, as a consequence, dimerizer injection induced an efficient reduction of neutrophil levels in all the organs analyzed under homeostatic conditions. In situations with extensive pressure on the bone marrow to mobilize neutrophils, for instance in the context of cancer, effective neutrophil depletion in this model requires further optimization. In conclusion, we here describe the generation and characterization of a new transgenic model for conditional neutrophil ablation and highlight the need to improve the ATTAC strategy for the depletion of large numbers of rapidly generated short-lived cells, such as neutrophils.


Assuntos
Neoplasias , Neutrófilos , Animais , Caspase 8/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Neoplasias/metabolismo , Infiltração de Neutrófilos , Neutrófilos/metabolismo
4.
Cell Rep ; 38(9): 110447, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35235800

RESUMO

Breast cancer is accompanied by systemic immunosuppression, which facilitates metastasis formation, but how this shapes organotropism of metastasis is poorly understood. Here, we investigate the impact of mammary tumorigenesis on regulatory T cells (Tregs) in distant organs and how this affects multi-organ metastatic disease. Using a preclinical mouse mammary tumor model that recapitulates human metastatic breast cancer, we observe systemic accumulation of activated, highly immunosuppressive Tregs during primary tumor growth. Tumor-educated Tregs show tissue-specific transcriptional rewiring in response to mammary tumorigenesis. This has functional consequences for organotropism of metastasis, as Treg depletion reduces metastasis to tumor-draining lymph nodes, but not to lungs. Mechanistically, we find that Tregs control natural killer (NK) cell activation in lymph nodes, thereby facilitating lymph node metastasis. In line, an increased Treg/NK cell ratio is observed in sentinel lymph nodes of breast cancer patients compared with healthy controls. This study highlights that immune regulation of metastatic disease is highly organ dependent.


Assuntos
Neoplasias da Mama , Animais , Neoplasias da Mama/patologia , Carcinogênese/patologia , Feminino , Humanos , Células Matadoras Naturais/patologia , Linfonodos , Metástase Linfática/patologia , Camundongos
5.
Semin Immunol ; 57: 101546, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34887163

RESUMO

Neutrophils are multifaceted innate immune cells that play a significant role in the progression of cancer by exerting both pro- and anti-tumorigenic functions. The crosstalk between cancer cells and neutrophils is complex and emerging evidence is pointing at cancer cell-intrinsic programs regulating neutrophil abundance, phenotype and function. Cancer cell-derived soluble mediators are key players in modulating the interaction with neutrophils. Here, we review how intrinsic features of cancer cells, including cancer cell genetics, epigenetics, signaling, and metabolism, manipulate neutrophil behavior and how to target these processes to impact cancer progression. A molecular understanding of cancer cell-intrinsic properties that shape the crosstalk with neutrophils will provide novel therapeutic strategies for personalized immunomodulation in cancer patients.


Assuntos
Neoplasias , Neutrófilos , Carcinogênese , Humanos , Imunomodulação , Transdução de Sinais , Microambiente Tumoral
6.
Methods Enzymol ; 632: 133-154, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32000893

RESUMO

The progression of cancer is strongly influenced by the crosstalk between cancer cells and immune cells. Immune cells can have both pro- and anti-tumor functions depending on the signals present in the environment. A significant proportion of the immune compartment of most solid tumors consists of tumor-associated macrophages. Although their abundance has been associated with poor prognosis in many solid tumor types, the molecular mechanisms by which cancer cells influence macrophage phenotype and function are largely unknown. In this chapter, we provide a detailed description of in vitro assays to study the impact of cancer cells on macrophages. We provide protocols to obtain macrophages from murine bone marrow and human peripheral blood, and to expose these macrophages to cancer cell-derived secreted molecules using conditioned medium from cancer cells. We describe several assays to assess cancer cell-induced polarization of macrophages. This experimental set-up can be utilized to gain molecular insights into how cancer cells influence macrophages.


Assuntos
Ativação de Macrófagos , Macrófagos/imunologia , Neoplasias/imunologia , Animais , Antígenos CD/análise , Antígenos CD/imunologia , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Diferenciação Celular , Separação Celular/métodos , Citometria de Fluxo/métodos , Humanos , Macrófagos/citologia , Microambiente Tumoral , Macrófagos Associados a Tumor/citologia , Macrófagos Associados a Tumor/imunologia
7.
Nature ; 572(7770): 538-542, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31367040

RESUMO

Cancer-associated systemic inflammation is strongly linked to poor disease outcome in patients with cancer1,2. For most human epithelial tumour types, high systemic neutrophil-to-lymphocyte ratios are associated with poor overall survival3, and experimental studies have demonstrated a causal relationship between neutrophils and metastasis4,5. However, the cancer-cell-intrinsic mechanisms that dictate the substantial heterogeneity in systemic neutrophilic inflammation between tumour-bearing hosts are largely unresolved. Here, using a panel of 16 distinct genetically engineered mouse models for breast cancer, we uncover a role for cancer-cell-intrinsic p53 as a key regulator of pro-metastatic neutrophils. Mechanistically, loss of p53 in cancer cells induced the secretion of WNT ligands that stimulate tumour-associated macrophages to produce IL-1ß, thus driving systemic inflammation. Pharmacological and genetic blockade of WNT secretion in p53-null cancer cells reverses macrophage production of IL-1ß and subsequent neutrophilic inflammation, resulting in reduced metastasis formation. Collectively, we demonstrate a mechanistic link between the loss of p53 in cancer cells, secretion of WNT ligands and systemic neutrophilia that potentiates metastatic progression. These insights illustrate the importance of the genetic makeup of breast tumours in dictating pro-metastatic systemic inflammation, and set the stage for personalized immune intervention strategies for patients with cancer.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Inflamação/genética , Inflamação/patologia , Metástase Neoplásica/patologia , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética , Proteínas Wnt/metabolismo , Animais , Neoplasias da Mama/complicações , Modelos Animais de Doenças , Feminino , Inflamação/complicações , Inflamação/imunologia , Interleucina-1beta/imunologia , Interleucina-1beta/metabolismo , Camundongos , Neutrófilos/imunologia
8.
Medicine (Baltimore) ; 95(18): e3457, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27149444

RESUMO

Recently, treatment with MEK inhibitors has been shown to be an effective treatment option for metastatic melanoma. Treatment efficacy is dependent on inhibition of MAPK-related melanoma proliferation. However, targeting of MEK can be accompanied by a time-dependent and reversible serous retinopathy of unknown origin.We analyzed the molecular mechanism by which the MEK inhibitor binimetinib may lead to retinopathy, using neuroretina and cell models of retinal pigment epithelium (RPE).Binimetinib inhibited the MAPK pathway while discontinuation of treatment resulted in reactivation. However, cell proliferation was not inhibited correspondingly during binimetinib treatment of ARPE19 cells. Remarkably, post-mitotic neuroretinal tissue displayed a strong MAPK activation that was lost after binimetinib treatment.We propose that binimetinib-associated retinopathy is correlated with inhibition of the MAPK pathway in multiple retinal components. Retinal cells are able to regain the activation after binimetinib treatment, mimicking the reversibility of the retinopathy. As most retinal cells are nonregenerating, other mechanisms than stimulation of proliferation must be involved.


Assuntos
Benzimidazóis , MAP Quinase Quinase Quinases/antagonistas & inibidores , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Melanoma/tratamento farmacológico , Doenças Retinianas , Epitélio Pigmentado da Retina , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Benzimidazóis/administração & dosagem , Benzimidazóis/efeitos adversos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Humanos , Melanoma/patologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Metástase Neoplásica , Doenças Retinianas/induzido quimicamente , Doenças Retinianas/metabolismo , Epitélio Pigmentado da Retina/efeitos dos fármacos , Epitélio Pigmentado da Retina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA