Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Cancer Res ; 84(2): 181-183, 2024 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-38055893

RESUMO

Current immunotherapeutic approaches are tailored towards biomarkers and mechanisms presented in the primary tumor microenvironment, yet few are designed against targets that arise in the metastatic site. In a recent Cancer Discovery article, Yofe and colleagues spatially and temporally mapped the evolving breast tumor metastatic environment within the lung at single-cell resolution to identify immunologic cell types and mechanisms underlying the formation of metastatic nodules. The authors identified myeloid cell, monocyte, and neutrophil phenotypes as the most significant differences between the primary tumor and metastatic site. The authors also revealed the existence of a metastasis-specific population of macrophages expressing Trem2 in both human and murine models that exhibit immunosuppressive features. Interestingly, Trem2+ macrophages are enriched at the invasive border of metastatic nodules compared with the metastatic core, suggesting they play an exclusionary role within the site. Characterization of the premetastatic lung in conjunction with pseudotime lineage analysis revealed that Fn1+ monocytes precede metastasis formation and are the likely source of Trem2+ macrophages in developed nodules. This study supports the characterization of metastatic immune microenvironments to identify novel immunotherapeutic targets that may not exist within the primary site but play a significant role in patient outcome.


Assuntos
Neoplasias da Mama , Neoplasias Pulmonares , Humanos , Animais , Camundongos , Feminino , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Pulmão/patologia , Neoplasias Pulmonares/patologia , Macrófagos/metabolismo , Microambiente Tumoral , Glicoproteínas de Membrana , Receptores Imunológicos
2.
Methods Mol Biol ; 2270: 125-147, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33479897

RESUMO

Emerging research suggests that IL-35-producing regulatory B cells accumulate in patients and mouse models of pancreatic cancer, one of the most lethal cancers, characterized by late diagnosis, high mortality, and morbidity. Identification of IL-35-producing B cells can be challenging due to the heterodimeric nature of IL-35 and diversity of cell surface markers that define regulatory B-cell subsets across spectrum of diseases. In this chapter, we describe the methods for the isolation of splenic and tumor-infiltrating murine regulatory B cells and subsequent detection of IL-35 by RT-qPCR and intracellular staining, as well as detection of circulating IL-35 by ELISA. We also describe methods for the detection of IL-35-producing human B cells by flow cytometry, RT-qPCR, and immunofluorescence in the context of pancreatic cancer. This chapter should facilitate the study of regulatory IL-35+ B cells in cancer, autoimmunity, and inflammation.


Assuntos
Linfócitos B/citologia , Citometria de Fluxo/métodos , Interleucinas/análise , Animais , Subpopulações de Linfócitos B/citologia , Subpopulações de Linfócitos B/imunologia , Linfócitos B/imunologia , Linfócitos B Reguladores/imunologia , Citocinas/metabolismo , Ensaio de Imunoadsorção Enzimática/métodos , Humanos , Interleucina-10/imunologia , Interleucinas/sangue , Camundongos , RNA Mensageiro/análise , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos
3.
Immunol Rev ; 299(1): 74-92, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33368346

RESUMO

Tumorigenesis proceeds through discrete steps where acquisition of genetic lesions and changes in the surrounding microenvironment combine to drive unrestricted neoplastic proliferation and metastasis. The ability of tumor-infiltrating immune cells to promote tumor growth via the provision of signals that enable tumor cell survival and proliferation as well as contribute to immune suppression is an active area of research. Recent efforts have provided us with mechanistic insights into how B cells can positively and negatively regulate immune responses. Negative regulation of immune responses in cancer can be mediated by regulatory B cells and is often a result of increased production of cytokines that can directly and indirectly affect anti-tumor immune function and cancer cell growth. Signals that lead to the expansion of regulatory B cells and the spectrum of their functional roles are not well understood and are the subject of active research by many groups. Here, we elaborate broadly on the history of regulatory B cells in cancer and summarize recent studies that have established genetic models for the study of regulatory B cell function and their potential for therapeutic intervention in the setting of solid cancers.


Assuntos
Linfócitos B Reguladores , Neoplasias , Citocinas , Humanos , Imunidade , Microambiente Tumoral
4.
Front Immunol ; 12: 745873, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35046933

RESUMO

B cells can act as potent suppressors of anti-tumor T cell immunity, presenting a mechanism of resistance to immunotherapy. In pancreatic ductal adenocarcinoma, B cells can display a T cell-suppressive or regulatory phenotype centered on the expression of the cytokine Interleukin 35 (IL-35). While B cell-mediated immunosuppression presents a barrier to anti-tumorigenic T cell function, it is not clear how regulatory B cell function could be targeted, and the signals that promote this suppressive phenotype in B cells are not well understood. Here we use a novel IL-35 reporter model to understand which signaling pathways are important for immunosuppressive properties in B cells. In vitro analysis of IL-35 reporter B cells revealed a synergy between the BCR and TLR4 signaling pathways is sufficient to induce IL-35 expression. However, in vivo, B cell receptor activation, as opposed to MyD88 signaling in B cells, is central to B cell-mediated suppression and promotion of pancreatic cancer growth. Further analysis identified protein kinase D2 (PKD2) as being a key downstream regulator of IL-35 expression in B cells. Regulatory B cells with an inactivating mutation in PKD2 failed to produce IL-35 or fully suppress effector T cell function in vitro. Furthermore, inhibition of PKD in B cells decreased tumor growth and promoted effector T cell function upon adoptive transfer into B cell-deficient mice. Collectively, these data provide insight into how regulatory B cell function is promoted in pancreatic cancer and identify potential therapeutic targets to restrain this function.


Assuntos
Linfócitos B Reguladores/imunologia , Carcinoma Ductal Pancreático/imunologia , Neoplasias Pancreáticas/imunologia , Proteína Quinase D2/imunologia , Receptores de Antígenos de Linfócitos B/imunologia , Animais , Interleucinas/biossíntese , Interleucinas/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/imunologia
5.
Sci Rep ; 10(1): 7390, 2020 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-32355248

RESUMO

Pancreatic cancer is an aggressive malignancy, often diagnosed at metastatic stages. Several studies have implicated systemic factors, such as extracellular vesicle release and myeloid cell expansion, in the establishment of pre-metastatic niches in cancer. The Rab27a GTPase is overexpressed in advanced cancers, can regulate vesicle trafficking, and has been previously linked to non-cell autonomous control of tumor growth and metastasis, however, the role of Rab27a itself in the metastatic propensity of pancreatic cancer is not well understood. Here, we have established a model to study how Rab27a directs formation of the pre-metastatic niche. Loss of Rab27a in pancreatic cancer cells did not decrease tumor growth in vivo, but resulted in altered systemic myeloid cell expansion, both in the primary tumors and at the distant organ sites. In metastasis assays, loss of Rab27a expression in tumor cells injected into circulation compromised efficient outgrowth of metastatic lesions. However, Rab27a knockdown cells had an unexpected advantage at initial steps of metastatic seeding, suggesting that Rab27a may alter cell-autonomous invasive properties of the tumor cells. Gene expression analysis of gene expression revealed that downregulation of Rab27a increased expression of genes involved in epithelial-to-mesenchymal transition pathways, consistent with our findings that primary tumors arising from Rab27a knockdown cells were more invasive. Overall, these data reveal that Rab27a can play divergent roles in regulating pro-metastatic propensity of pancreatic cancer cells: by generating pro-metastatic environment at the distant organ sites, and by suppressing invasive properties of the cancer cells.


Assuntos
Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/biossíntese , Neoplasias Pancreáticas/enzimologia , Proteínas rab27 de Ligação ao GTP/biossíntese , Animais , Linhagem Celular Tumoral , Feminino , Masculino , Camundongos , Metástase Neoplásica , Proteínas de Neoplasias/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Proteínas rab27 de Ligação ao GTP/genética
6.
Cancer Immunol Res ; 8(3): 292-308, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32024640

RESUMO

Pancreatic ductal adenocarcinoma (PDA) is an aggressive malignancy characterized by a paucity of tumor-proximal CD8+ T cells and resistance to immunotherapeutic interventions. Cancer-associated mechanisms that elicit CD8+ T-cell exclusion and resistance to immunotherapy are not well-known. Here, using a Kras- and p53-driven model of PDA, we describe a mechanism of action for the protumorigenic cytokine IL35 through STAT3 activation in CD8+ T cells. Distinct from its action on CD4+ T cells, IL35 signaling in gp130+CD8+ T cells activated the transcription factor STAT3, which antagonized intratumoral infiltration and effector function of CD8+ T cells via suppression of CXCR3, CCR5, and IFNγ expression. Inhibition of STAT3 signaling in tumor-educated CD8+ T cells improved PDA growth control upon adoptive transfer to tumor-bearing mice. We showed that activation of STAT3 in CD8+ T cells was driven by B cell- but not regulatory T cell-specific production of IL35. We also demonstrated that B cell-specific deletion of IL35 facilitated CD8+ T-cell activation independently of effector or regulatory CD4+ T cells and was sufficient to phenocopy therapeutic anti-IL35 blockade in overcoming resistance to anti-PD-1 immunotherapy. Finally, we identified a circulating IL35+ B-cell subset in patients with PDA and demonstrated that the presence of IL35+ cells predicted increased occurrence of phosphorylated (p)Stat3+CXCR3-CD8+ T cells in tumors and inversely correlated with a cytotoxic T-cell signature in patients. Together, these data identified B cell-mediated IL35/gp130/STAT3 signaling as an important direct link to CD8+ T-cell exclusion and immunotherapy resistance in PDA.


Assuntos
Linfócitos B/imunologia , Carcinoma Ductal Pancreático/imunologia , Interleucinas/imunologia , Neoplasias Pancreáticas/imunologia , Fator de Transcrição STAT3/imunologia , Animais , Apoptose/imunologia , Linfócitos T CD8-Positivos/imunologia , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Carcinoma Ductal Pancreático/terapia , Estudos de Casos e Controles , Proliferação de Células/fisiologia , Humanos , Imunoterapia Adotiva/métodos , Interleucinas/genética , Ativação Linfocitária , Linfócitos do Interstício Tumoral/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/terapia , Receptores CCR5/genética , Receptores CCR5/imunologia , Receptores CXCR3/genética , Receptores CXCR3/imunologia , Fator de Transcrição STAT3/genética , Transdução de Sinais/imunologia , Linfócitos T Reguladores/imunologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Nat Biotechnol ; 38(4): 448-459, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32015548

RESUMO

Cytokines that stimulate T cell proliferation, such as interleukin (IL)-15, have been explored as a means of boosting the antitumor activity of chimeric antigen receptor (CAR) T cells. However, constitutive cytokine signaling in T cells and activation of bystander cells may cause toxicity. IL-23 is a two-subunit cytokine known to promote proliferation of memory T cells and T helper type 17 cells. We found that, upon T cell antigen receptor (TCR) stimulation, T cells upregulated the IL-23 receptor and the IL-23α p19 subunit, but not the p40 subunit. We engineered expression of the p40 subunit in T cells (p40-Td cells) and obtained selective proliferative activity in activated T cells via autocrine IL-23 signaling. In comparison to CAR T cells, p40-Td CAR T cells showed improved antitumor capacity in vitro, with increased granzyme B and decreased PD-1 expression. In two xenograft and two syngeneic solid tumor mouse models, p40-Td CAR T cells showed superior efficacy in comparison to CAR T cells and attenuated side effects in comparison to CAR T cells expressing IL-18 or IL-15.


Assuntos
Imunoterapia Adotiva/métodos , Interleucina-23/metabolismo , Neoplasias/terapia , Linfócitos T/imunologia , Animais , Hipóxia Celular/genética , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Subunidade p40 da Interleucina-12/genética , Subunidade p40 da Interleucina-12/metabolismo , Interleucina-23/genética , Ativação Linfocitária , Camundongos , Neoplasias/imunologia , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/metabolismo , Receptores de Interleucina/genética , Receptores de Interleucina/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/genética , Linfócitos T/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Cytokine ; 125: 154817, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31472403

RESUMO

B cells are important modulators of immune responses both in autoimmunity and cancer. We have previously shown that B regulatory (Breg) cells promote pancreatic cancer via production of IL35, a heterodimeric cytokine comprised of the subunits p35 (Il12a) and Ebi3. However, it is not known how production of IL35 is regulated in vivo in the context of cancer-associated inflammation. To begin addressing this question, we have generated a knock-in mouse model, Il12aGFP, where an IRES-emGFP gene was inserted within the 3' UTR of the Il12a locus. EmGFP signal in B cells from the Il12aGFP mice correlated with expression of p35 mRNA and protein. Using this model, we observed that in addition to Bregs, expression of GFP (p35) is upregulated in several other B cell subtypes in response to cancer. We assessed the expression of the other IL35 subunit, Ebi3, using a published tdTomato reporter model. We determined that Ebi3 expression was more tightly regulated in vivo and in vitro, suggesting that stimuli affecting Ebi3 upregulation are more likely to result in production of full IL35 heterodimer. We were also able to detect GFP and Tomato signal in myeloid & T cell lineages suggesting that these reporter models could also be used for tracking IL12-, IL27- and IL35-producing cells. Furthermore, using primary B cells isolated from reporter mice, we identified BCR, CD40 and TLR pathways as potential drivers of IL35 expression. These findings highlight the importance of pancreatic cancer-associated inflammatory processes as drivers of cytokine expression and provide a tool to dissect both disease-associated regulation of IL12- and IL35-competent lineage cells as well as establish assays for pharmacological targeting of individual subunits of heterodimeric IL12 family cytokines.


Assuntos
Linfócitos B Reguladores/imunologia , Subunidade p35 da Interleucina-12/metabolismo , Interleucinas/metabolismo , Antígenos de Histocompatibilidade Menor/metabolismo , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/metabolismo , Receptores de Citocinas/metabolismo , Animais , Antígenos CD40/metabolismo , Linhagem Celular Tumoral , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/imunologia , Inflamação/imunologia , Inflamação/metabolismo , Subunidade p35 da Interleucina-12/genética , Interleucinas/genética , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas c-bcr/metabolismo , Receptores Toll-Like/metabolismo , Regulação para Cima
9.
Cancer Cell ; 35(2): 221-237.e8, 2019 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-30753824

RESUMO

The high expression across multiple tumor types and restricted expression in normal tissues make B7-H3 an attractive target for immunotherapy. We generated chimeric antigen receptor (CAR) T cells targeting B7-H3 (B7-H3.CAR-Ts) and found that B7-H3.CAR-Ts controlled the growth of pancreatic ductal adenocarcinoma, ovarian cancer and neuroblastoma in vitro and in orthotopic and metastatic xenograft mouse models, which included patient-derived xenograft. We also found that 4-1BB co-stimulation promotes lower PD-1 expression in B7-H3.CAR-Ts, and superior antitumor activity when targeting tumor cells that constitutively expressed PD-L1. We took advantage of the cross-reactivity of the B7-H3.CAR with murine B7-H3, and found that B7-H3.CAR-Ts significantly controlled tumor growth in a syngeneic tumor model without evident toxicity. These findings support the clinical development of B7-H3.CAR-Ts.


Assuntos
Antígenos B7/imunologia , Carcinoma Ductal Pancreático/terapia , Imunoterapia Adotiva/métodos , Neuroblastoma/terapia , Neoplasias Ovarianas/terapia , Neoplasias Pancreáticas/terapia , Receptores de Antígenos Quiméricos/imunologia , Animais , Antígenos B7/genética , Antígeno B7-H1/imunologia , Antígenos CD28/imunologia , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/imunologia , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Técnicas de Cocultura , Feminino , Humanos , Imunoterapia Adotiva/efeitos adversos , Masculino , Camundongos Endogâmicos C57BL , Neuroblastoma/genética , Neuroblastoma/imunologia , Neuroblastoma/patologia , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/patologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/patologia , Receptores de Antígenos Quiméricos/genética , Transdução de Sinais , Carga Tumoral , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Cancer Immunol Res ; 6(9): 1014-1024, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29980536

RESUMO

Although successes in cancer immunotherapy have generated considerable excitement, this form of treatment has been largely ineffective in patients with pancreatic ductal adenocarcinoma (PDA). Mechanisms that contribute to the poor antitumor immune response in PDA are not well understood. Here, we demonstrated that cytokine IL35 is a major immunosuppressive driver in PDA and potentiates tumor growth via the suppression of endogenous antitumor T-cell responses. The growth of pancreatic tumors in mice deficient for IL35 was significantly reduced. An analysis of tumor-infiltrating immune cells revealed a role for IL35 in the expansion of regulatory T cells and the suppression of CD4+ effector T cells. We also detected a robust increase in both the infiltration and activation of cytotoxic CD8+ T cells, suggesting that targeting IL35 may be an effective strategy to convert PDA from an immunologically "cold" to "hot" tumor. Although PDA is typically resistant to anti-PD-1 immunotherapy, we demonstrated robust synergistic reduction in tumor growth when IL35 deficiency was combined with anti-PD-1 treatment. These findings provide new insight into the function of IL35 in the pathogenesis of pancreatic cancer and underscore the potential significance of IL35 as a therapeutic target for use in combination immunotherapy approaches in this deadly malignancy. Cancer Immunol Res; 6(9); 1014-24. ©2018 AACR.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Carcinoma Ductal Pancreático/imunologia , Interleucinas/imunologia , Neoplasias Pancreáticas/imunologia , Linfócitos T Reguladores/imunologia , Animais , Carcinoma Ductal Pancreático/patologia , Feminino , Imunoterapia , Interleucinas/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Pancreáticas/patologia , Receptor de Morte Celular Programada 1/imunologia
12.
Appl Opt ; 42(30): 6179-83, 2003 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-14594082

RESUMO

Analyses of iron ore slurries in industrial and laboratory environments showed that many physical and geometric parameters affect the stability and reproducibility of the response to laser-induced breakdown spectroscopy. A thorough reexamination of the sampling strategy led to a revised sampling layout that ensures a true representative sampling of the slurry and significantly improves the sensitivity and repeatability of the sampling method. An examination of the characteristics of slurries revealed that the mean particulate size and the concentration of solids in a slurry influence the measurement of silica, whereas the magnetite content is responsible for a matrix effect. On-line monitoring of iron ore slurries should be practicable, provided that these variables are controlled within a few percent or that some means of correction is implemented.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA