GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
Filter
  • American Association for Cancer Research (AACR)  (6)
  • 1
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 7, No. 7 ( 2019-07-01), p. 1091-1105
    Abstract: Clear-cell renal cell carcinoma (ccRCC) possesses an unmet medical need, particularly at the metastatic stage, when surgery is ineffective. Complement is a key factor in tissue inflammation, favoring cancer progression through the production of complement component 5a (C5a). However, the activation pathways that generate C5a in tumors remain obscure. By data mining, we identified ccRCC as a cancer type expressing concomitantly high expression of the components that are part of the classical complement pathway. To understand how the complement cascade is activated in ccRCC and impacts patients' clinical outcome, primary tumors from three patient cohorts (n = 106, 154, and 43), ccRCC cell lines, and tumor models in complement-deficient mice were used. High densities of cells producing classical complement pathway components C1q and C4 and the presence of C4 activation fragment deposits in primary tumors correlated with poor prognosis. The in situ orchestrated production of C1q by tumor-associated macrophages (TAM) and C1r, C1s, C4, and C3 by tumor cells associated with IgG deposits, led to C1 complex assembly, and complement activation. Accordingly, mice deficient in C1q, C4, or C3 displayed decreased tumor growth. However, the ccRCC tumors infiltrated with high densities of C1q-producing TAMs exhibited an immunosuppressed microenvironment, characterized by high expression of immune checkpoints (i.e., PD-1, Lag-3, PD-L1, and PD-L2). Our data have identified the classical complement pathway as a key inflammatory mechanism activated by the cooperation between tumor cells and TAMs, favoring cancer progression, and highlight potential therapeutic targets to restore an efficient immune reaction to cancer.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Epidemiology, Biomarkers & Prevention, American Association for Cancer Research (AACR), Vol. 26, No. 6 ( 2017-06-01), p. 905-913
    Abstract: Background: Night-shift work (NSW) has been suggested as a possible cause of breast cancer, and its association with mammographic density (MD), one of the strongest risk factors for breast cancer, has been scarcely addressed. This study examined NSW and MD in Spanish women. Methods: The study covered 2,752 women aged 45–68 years recruited in 2007–2008 in 7 population-based public breast cancer screening centers, which included 243 women who had performed NSW for at least one year. Occupational data and information on potential confounders were collected by personal interview. Two trained radiologist estimated the percentage of MD assisted by a validated semiautomatic computer tool (DM-scan). Multivariable mixed linear regression models with random screening center–specific intercepts were fitted using log-transformed percentage of MD as the dependent variable and adjusting by known confounding variables. Results: Having ever worked in NSW was not associated with MD [{e^\beta }$:0.96; 95% confidence interval (CI), 0.86–1.06]. However, the adjusted geometric mean of the percentage of MD in women with NSW for more than 15 years was 25% higher than that of those without NSW history (MD & gt;15 years:20.7% vs. MDnever:16.5%;{e^\beta }$:1.25; 95% CI,1.01–1.54). This association was mainly observed in postmenopausal participants ({e^\beta }$:1.28; 95% CI, 1.00–1.64). Among NSW-exposed women, those with ≤2 night-shifts per week had higher MD than those with 5 to 7 nightshifts per week ({e^\beta }$:1.42; 95% CI, 1.10–1.84). Conclusions: Performing NSW was associated with higher MD only in women with more than 15 years of cumulated exposure. These findings warrant replication in futures studies. Impact: Our findings suggest that MD could play a role in the pathway between long-term NSW and breast cancer. Cancer Epidemiol Biomarkers Prev; 26(6); 905–13. ©2017 AACR.
    Type of Medium: Online Resource
    ISSN: 1055-9965 , 1538-7755
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    detail.hit.zdb_id: 2036781-8
    detail.hit.zdb_id: 1153420-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 23, No. 15 ( 2017-08-01), p. 4416-4428
    Abstract: Purpose: The efficacy of PD-1 checkpoint blockade as adjuvant therapy in localized clear cell renal cell carcinoma (ccRCC) is currently unknown. The identification of tumor microenvironment (TME) prognostic biomarkers in this setting may help define which patients could benefit from checkpoint blockade and uncover new therapeutic targets. Experimental Design: We performed multiparametric flow cytometric immunophenotypic analysis of T cells isolated from tumor tissue [tumor-infiltrating lymphocytes (TIL)], adjacent non-malignant renal tissue [renal-infiltrating lymphocytes (RIL)] , and peripheral blood lymphocytes (PBL), in a cohort of patients (n = 40) with localized ccRCC. Immunophenotypic data were integrated with prognostic and histopathologic variables, T-cell receptor (TCR) repertoire analysis of sorted CD8+PD-1+ TILs, tumor mRNA expression, and digital quantitative immunohistochemistry. Results: On the basis of TIL phenotypic characterization, we identified three dominant immune profiles in localized ccRCC: (i) immune-regulated, characterized by polyclonal/poorly cytotoxic CD8+PD-1+Tim-3+Lag-3+ TILs and CD4+ICOS+ cells with a Treg phenotype (CD25+CD127−Foxp3+/Helios+GITR+), that developed in inflamed tumors with prominent infiltrations by dysfunctional dendritic cells and high PD-L1 expression; (ii) immune-activated, enriched in oligoclonal/cytotoxic CD8+PD-1+Tim-3+ TILs, that represented 22% of the tumors; and (iii) immune-silent, enriched in TILs exhibiting RIL-like phenotype, that represented 56% of patients in the cohort. Only immune-regulated tumors displayed aggressive histologic features, high risk of disease progression in the year following nephrectomy, and a CD8+PD-1+Tim-3+ and CD4+ICOS+ PBL phenotypic signature. Conclusions: In localized ccRCC, the infiltration with CD8+PD-1+Tim-3+Lag-3+ exhausted TILs and ICOS+ Treg identifies the patients with deleterious prognosis who could benefit from adjuvant therapy with TME-modulating agents and checkpoint blockade. This work also provides PBL phenotypic markers that could allow their identification. Clin Cancer Res; 23(15); 4416–28. ©2017 AACR.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 4, No. 1_Supplement ( 2016-01-01), p. A085-A085
    Abstract: This study aimed to decipher the potential mechanisms linking increased CD8+ T cell infiltration with an adverse clinical outcome in ccRCC. Clear cell Renal Cell Carcinoma (ccRCC) appears to be one of the most sensitive tumors to checkpoint blockade therapies. However, contrary to most cancers, some studies suggest that high densities of infiltrating CD8+ T cells correlate with patients' poor prognosis. In this study we determined: 1) the prognosis associated with the expression of immune checkpoints and its coordination with dendritic cell (DC) and CD8+ cell infiltration, and 2) the major phenotypic traits of CD8+ tumor infiltrating lymphocytes (CD8+ TILs). The prognosis associated with CD8+ and dendritic cell (DC) infiltrations, in addition to the expression of immune checkpoints were investigated in a cohort of 135 primary ccRCC by quantitative immunohistochemistry (IHC). We found that the densities of CD8+, PD-1+ and LAG-3+ cells were closely inter-correlated, and independently associated with decreased PFS and OS. In addition, patients whose tumors presented both high densities of PD-1+ infiltrating cells and PD-L1+ and/or L2+ tumor cells ( & gt;5% positive cells), displayed the worst clinical outcome [9% 5-year disease-free survival (DFS) vs. 64% in the other group, P & lt;0.0001]. These parameters were modulated by the presence, phenotype and localization of DC in the tumour microenvironment. High densities of immature DC isolated in the tumour stroma (DC-Lamp+/CD83dim/HLA-DRdim) were associated with high expression of immune checkpoints and patients' poor clinical outcome. In contrast, the presence of mature DC (DC-Lamp+/CD83bright/HLA-DRbright) within Tertiary Lymphoid Structures (TLS) (TLS-DC) identified, among the tumours with high CD8+ TIL densities, those with low expression of immune checkpoints and prolonged survival (73% DFS at 5-years compared to 10%, P=0.001). Analysis of gene expression data from the TCGA (n=495) demonstrated an association between the over-expression of CD8 TIL-related genes (CD8A, TBX21, IRF1, GZMB and IFNG) and patients' poor clinical outcome, in addition to a strong correlation between the expression of CD8A, PD-1, LAG-3, PD-L2 and IFNG genes in ccRCC. These results suggest that in the absence of an immune response orchestrated by TLS-DC, the CD8+ T cells in ccRCC display an inhibited phenotype, associated with more advanced tumor stages and poor clinical outcome. To functionally characterize the CD8+ T cell infiltrates in ccRCC, we investigated the major phenotypic traits of freshly isolated lymphocytes from 21 primary tumors obtained by partial or radical nephrectomy. Non-supervised analyses of the CD8+ TIL phenotype revealed two different types of tumors: The first group or “inhibited group” (8/21) was characterized by the over-expression of inhibitory molecules (PD-1 and TIM-3) and activation molecules (CD69 and CD38), and the expansion of the effector memory cell subpopulation (CCR7-CD45RA-) in the CD8+ TIL; the second group (14/21) was characterized by the expansion of the CD8+EMRA subpopulation (CCR7-CD45RA+) in addition to a reduced expression of activation or inhibitory markers. Preliminary data suggest that the “inhibited group” of tumors is characterized by more advanced tumor stages, higher Fuhrman grades and increased CD8+ cell densities (as determined by IHC), and thus shares major characteristics with tumors associated with patients' worst clinical outcome in the retrospective cohort. The functional orientation and cytotoxic potential of TIL are under investigation in both groups of tumors to further characterize the exhaustion and/or suppressive features of CD8+ T cells associated with poor clinical outcome in primary ccRCC. In summary, we demonstrated that the infiltration with CD8+ TILs in ccRCC is accompanied by the enhanced expression of immune checkpoints in a subgroup of tumors displaying more aggressive features and advanced tumors stages. This immune profile defines a poor prognosis group of patients that should be suitable to receive immune checkpoint inhibitors. Citation Format: Nicolas A. Giraldo, Becht Etienne, Laetitia Lacroix, Yann Vano, Rafael Sanchez-Salas, Pierre Validire, Virginie Verkarre, Arnaud Mejean, Marie-Caroline Dieu-Nosjean, Stephane Oudard, Xavier Cathelineau, Wolf H. Fridman, Catherine Sautès-Fridman. Orchestration and prognostic significance of immune checkpoints in the microenvironment of primary clear cell renal cell cancer. [abstract]. In: Proceedings of the CRI-CIMT-EATI-AACR Inaugural International Cancer Immunotherapy Conference: Translating Science into Survival; September 16-19, 2015; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2016;4(1 Suppl):Abstract nr A085.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 9, No. 8 ( 2021-08-01), p. 891-908
    Abstract: The complement system plays a complex role in cancer. In clear cell renal cell carcinoma (ccRCC), local production of complement proteins drives tumor progression, but the mechanisms by which they do this are poorly understood. We found that complement activation, as reflected by high plasma C4d or as C4d deposits at the tumor site, was associated with poor prognosis in two cohorts of patients with ccRCC. High expression of the C4-activating enzyme C1s by tumor cells was associated with poor prognosis in three cohorts. Multivariate Cox analysis revealed that the prognostic value of C1s was independent from complement deposits, suggesting the possibility of complement cascade–unrelated, protumoral functions for C1s. Silencing of C1s in cancer cell lines resulted in decreased proliferation and viability of the cells and in increased activation of T cells in in vitro cocultures. Tumors expressing high levels of C1s showed high infiltration of macrophages and T cells. Modification of the tumor cell phenotype and T-cell activation were independent of extracellular C1s levels, suggesting that C1s was acting in an intracellular, noncanonical manner. In conclusion, our data point to C1s playing a dual role in promoting ccRCC progression by triggering complement activation and by modulating the tumor cell phenotype and tumor microenvironment in a complement cascade–independent, noncanonical manner. Overexpression of C1s by tumor cells could be a new escape mechanism to promote tumor progression. See related Spotlight by Magrini and Garlanda, p. 855. See article by Daugan et al., p. 909 (40).
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 9, No. 8 ( 2021-08-01), p. 909-925
    Abstract: The complement system is a powerful and druggable innate immune component of the tumor microenvironment. Nevertheless, it is challenging to elucidate the exact mechanisms by which complement affects tumor growth. In this study, we examined the processes by which the master complement regulator factor H (FH) affects clear cell renal cell carcinoma (ccRCC) and lung cancer, two cancers in which complement overactivation predicts poor prognosis. FH was present in two distinct cellular compartments: the membranous (mb-FH) and intracellular (int-FH) compartments. Int-FH resided in lysosomes and colocalized with C3. In ccRCC and lung adenocarcinoma, FH exerted protumoral action through an intracellular, noncanonical mechanism. FH silencing in ccRCC cell lines resulted in decreased proliferation, due to cell-cycle arrest and increased mortality, and this was associated with increased p53 phosphorylation and NFκB translocation to the nucleus. Moreover, the migration of the FH-silenced cells was reduced, likely due to altered morphology. These effects were cell type–specific because no modifications occurred upon CFH silencing in other FH-expressing cells tested: tubular cells (from which ccRCC originates), endothelial cells (human umbilical vein endothelial cells), and squamous cell lung cancer cells. Consistent with this, in ccRCC and lung adenocarcinoma, but not in lung squamous cell carcinoma, int-FH conferred poor prognosis in patient cohorts. Mb-FH performed its canonical function of complement regulation but had no impact on tumor cell phenotype or patient survival. The discovery of intracellular functions for FH redefines the role of the protein in tumor progression and its use as a prognostic biomarker or potential therapeutic target. See article by Daugan et al., p. 891 (36).
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...