103 research outputs found

    Patterns of antibody responses to nonviral cancer antigens in head and neck squamous cell carcinoma patients differ by human papillomavirus status

    Get PDF
    There have been hints that nonviral cancer antigens are differentially expressed in human papillomavirus (HPV)-positive and HPV-negative head and neck squamous cell carcinoma (HNSCC). Antibody responses (AR) to cancer antigens may be used to indirectly determine cancer antigen expression in the tumor using a noninvasive and tissue-saving liquid biopsy. Here, we set out to characterize AR to a panel of nonviral cancer antigens in HPV-positive and HPV-negative HNSCC patients. A fluorescent microbead multiplex serology to 29 cancer antigens (16 cancer-testis antigens, 5 cancer-retina antigens and 8 oncogenes) and 29 HPV-antigens was performed in 382 HNSCC patients from five independent cohorts (153 HPV-positive and 209 HPV-negative). AR to any of the cancer antigens were found in 272/382 patients (72%). The ten most frequent AR were CT47, cTAGE5a, c-myc, LAGE-1, MAGE-A1, -A3, -A4, NY-ESO-1, SpanX-a1 and p53. AR to MAGE-A3, MAGE-A9 and p53 were found at significantly different prevalences by HPV status. An analysis of AR mean fluorescent intensity values uncovered remarkably different AR clusters by HPV status. To identify optimal antigen selections covering a maximum of patients with ā‰¤10 AR, multiobjective optimization revealed distinct antigen selections by HPV status. We identified that AR to nonviral antigens differ by HPV status indicating differential antigen expression. Multiplex serology may be used to characterize antigen expression using serum or plasma as a tissue-sparing liquid biopsy. Cancer antigen panels should address the distinct antigen repertoire of HPV-positive and HPV-negative HNSCC

    Heterogeneous expression of predictive biomarkers PD-L1 and TIGIT in non-mucinous lung adenocarcinoma and corresponding lymph node metastasis: A challenge for clinical biomarker testing

    No full text
    The use of immune checkpoint inhibitors (ICI) targeting the PD-L1:PD1 interaction revolutionized tumor treatment by re-activating the anti-tumoral capacity of the immune system. Assessment of tumor mutational burden, microsatellite instability, or expression of the surface marker PD-L1 have been used to predict individual response to ICI therapy. However, the predicted response does not always correspond to the actual therapy outcome. We hypothesize that tumor heterogeneity might be a major cause of this inconsistency. In this respect we recently demonstrated that PD-L1 shows heterogenous expression in the different growth patterns of non-small cell lung cancer (NSCLC) - lepidic, acinar, papillary, micropapillary and solid. Furthermore, additional inhibitory receptors, like T cell immunoglobulin and ITIM domain (TIGIT), appear to be heterogeneously expressed and affect the outcome of anti-PD-L1 treatment. Given this heterogeneity in the primary tumor, we set out to analyze the situation in corresponding lymph node metastases, since these are often used to obtain biopsy material for tumor diagnosis, staging and molecular analysis. Again, we observed heterogeneous expression of PD-1, PD-L1, TIGIT, Nectin-2 and PVR in relation to different regions and growth pattern distribution that varied between the primary tumor and their metastases. Together, our study underscores the complex situation regarding the heterogeneity of NSCLC samples and suggest that the analysis of a small biopsy from lymph node metastases may not be sufficient to ensure a reliable prediction of ICI therapy success

    GSK3Ī² modulates NF-ĪŗB activation and RelB degradation through site-specific phosphorylation of BCL10

    No full text
    Abstract Glycogen synthase kinase 3Ī² (GSK3Ī²) is a ubiquitously expressed serine/threonine kinase involved in the regulation of various cellular functions, such as energy homoeostasis, cell growth and developmental processes. More recently, GSK3Ī² has been identified as a part of a protein complex involved in the regulation of the CARMA1-BCL10-MALT1 complex (CBM complex) formation, which is a key signalling event upon antigen receptor engagement of B and T cells, required for the activation of the NF-ĪŗB and JNK pathways. However, conflicting reports have been published regarding the role of GSK3Ī² for the activation of the NF-ĪŗB signalling pathways. Therefore, we aimed to determine the impact of GSK3Ī² on the NF-ĪŗB signalling induced upon T cell activation. Blocking GSK3Ī² by either pharmacologic inhibitors (SB216763 and SB415286) or by RNAi caused a reduced proteolysis of the MALT1 targets CYLD1, BCL10 and RelB as well as diminished IĪŗBĪ± degradation, NF-ĪŗB DNA binding and NF-ĪŗB activity. This negative effect on NF-ĪŗB appears to be due to a diminished CBM complex formation caused by a reduced BCL10 phosphorylation. Taken together, we provide here evidence for a novel regulatory mechanism by which GSK3Ī² affects NF-ĪŗB signalling in activated T cells

    Octamer-dependent transcription in T cells is mediated by NFAT and NFāˆ’ĪŗBNF-\kappa B

    Get PDF
    The transcriptional co-activator BOB.1/OBF.1 was originally identified in B cells and is constitutively expressed throughout B cell development. BOB.1/OBF.1 associates with the transcription factors Oct1 and Oct2, thereby enhancing octamer-dependent transcription. In contrast, in T cells, BOB.1/OBF.1 expression is inducible by treatment of cells with PMA/Ionomycin or by antigen receptor engagement, indicating a marked difference in the regulation of BOB.1/OBF.1 expression in B versus T cells. The molecular mechanisms underlying the differential expression of BOB.1/OBF.1 in T and B cells remain largely unknown. Therefore, the present study focuses on mechanisms controlling the transcriptional regulation of BOB.1/OBF.1 and Oct2 in T cells. We show that both calcineurin- and NFāˆ’ĪŗBNF-\kappa B-inhibitors efficiently attenuate the expression of BOB.1/OBF.1 and Oct2 in T cells. In silico analyses of the BOB.1/OBF.1 promoter revealed the presence of previously unappreciated combined NFAT/NFāˆ’ĪŗBNF-\kappa B sites. An array of genetic and biochemical analyses illustrates the involvement of the Ca2+Ca^{2+}/calmodulin-dependent phosphatase calcineurin as well as NFAT and NFāˆ’ĪŗBNF-\kappa B transcription factors in the transcriptional regulation of octamer-dependent transcription in T cells. Conclusively, impaired expression of BOB.1/OBF.1 and Oct2 and therefore a hampered octamer-dependent transcription may participate in T cell-mediated immunodeficiency caused by the deletion of NFAT or NFāˆ’ĪŗBNF-\kappa B transcription factors

    IĪŗB Kinases Modulate the Activity of the Androgen Receptor in Prostate Carcinoma Cell Lines12

    Get PDF
    Enhanced nuclear localization of nuclear factor ĪŗB (NF-ĪŗB) in prostate cancer (PCa) samples and constitutive NF-ĪŗB signaling in a class of PCa cell lines with low androgen receptor (AR) expression (PC3 and DU-145) imply an important role of the IĪŗB kinase (IKK)/NF-ĪŗB system in PCa. However, most PCa and PCa cell lines depend on the activity of the AR, and the role of NF-ĪŗB in these AR-expressing PCa remains unclear. Here, we demonstrate that inhibition of NF-ĪŗB signaling by the IKK inhibitor BMS345541 reduced proliferation and increased apoptosis in AR-expressing PCa cell lines. Furthermore, AR activity and target gene expression were distinctively reduced, whereas AR protein levels remained unaltered on BMS345541 treatment. Similar effects were observed particularly after small interfering RNA (siRNA)-mediated knockdown of IKK1, but not by siRNA-mediated suppression of IKK2. Moreover, IKK1 overexpression augmented 5Ī±-dihydrotestosterone-induced nuclear AR translocation, whereas nuclear AR was reduced by IKK1 knockdown or BMS345541. However, because IKK1 also enhances the activity of a chronically nuclear AR mutant, modulation of the subcellular distribution seems not to be the only mechanism by which IKK1 enhances AR activity. Finally, reduced in vivo AR phosphorylation after BMS345541 treatment and in vitro AR phosphorylation by IKK1 or IKK2 imply that AR constitutes a novel IKK target. Taken together, our data identify IKK1 as a potentially target structure for future therapeutic intervention in PCa

    The Ca2+-dependent Phosphatase Calcineurin Controls the Formation of the Carma1-Bcl10-Malt1 Complex during T Cell Receptor-induced NF-ĪŗB Activation*

    No full text
    T cell receptor (TCR) ligation induces increased diacylglycerol and Ca2+ levels in T cells, and both secondary messengers are crucial for TCR-induced nuclear factor of activated T cells (NF-AT) and NF-ĪŗB signaling pathways. One prominent calcium-dependent enzyme involved in the regulation of NF-AT and NF-ĪŗB signaling pathways is the protein phosphatase calcineurin. However, in contrast to NF-AT, which is directly dephosphorylated by calcineurin, the molecular basis of the calcium-calcineurin dependence of the TCR-induced NF-ĪŗB activity remains largely unknown. Here, we demonstrate that calcineurin regulates TCR-induced NF-ĪŗB activity by controlling the formation of a protein complex composed of Carma1, Bcl10, and Malt1 (CBM complex). For instance, increased calcium levels induced by ionomycin or thapsigargin augmented the phorbol 12-myristate 13-acetate-induced formation of the CBM complex and activation of NF-ĪŗB, whereas removal of calcium by the calcium chelator EGTA-acetoxymethyl ester (AM) attenuated both processes. Furthermore, inhibition of the calcium-dependent phosphatase calcineurin with the immunosuppressive agent cyclosporin A (CsA) or FK506 as well as siRNA-mediated knockdown of calcineurin A strongly affected the PMA + ionomycin- or anti-CD3 + CD28-induced CBM complex assembly. Mechanistically, the positive effect of calcineurin on the CBM complex formation seems to be linked to a dephosphorylation of Bcl10. For instance, Bcl10 was found to be hyperphosphorylated in Jurkat T cells upon treatment with CsA or EGTA-AM, and calcineurin dephosphorylated Bcl10 in vivo and in vitro. Furthermore, we show here that calcineurin A interacts with the CBM complex. In summary, the evidence provided here argues for a previously unanticipated role of calcineurin in CBM complex formation as a molecular basis of the inhibitory function of CsA or FK506 on TCR-induced NF-ĪŗB activity

    Antibody responses to cancer antigens identify patients with a poor prognosis among HPV-positive and HPV-negative head and neck squamous cell carcinoma patients

    Full text link
    Purpose: The identification of high-risk patients within Human Papillomavirus (HPV) positive and negative head and neck squamous cell carcinoma patients is needed for improved treatment and surveillance strategies. In this study, we set out to discover Antibody responses (AR) with prognostic impact in head and neck squamous cell carcinoma (HNSCC) stratified by HPV-status. Experimental Design: A fluorescent bead-based multiplex serology assay to 29 cancer antigens (16 cancer-testis antigens, 5 cancer-retina antigens, 8 oncogenes) and 29 HPV-antigens was performed in samples of 362 HNSCC patients from five independent cohorts (153 HPV-positive, 209 HPV-negative). A multivariable cox proportional hazard model with bootstrapping (M=1000) was used for validation of prognostic antibody responses. Results: AR to any of the cancer antigens were found in 257/362 patients (71%). In HPV-negative patients, antibody responses to to c-myc, MAGE-A1, -A4 and Rhodopsin E2 (combined as ARhigh risk) were significantly associated with shorter overall survival. In HPV-positive patients antibody responses to IMP-1 were discovered as a negative prognostic factor. ARhigh risk (HR=1.76) and antibody responses to IMP-1 (HR=3.28) were confirmed as independent markers for a poor prognosis in a multivariable Cox proportional hazard model with bootstrapping (M=1000). Conclusion: We identified AR to cancer antigens that associate with a dismal prognosis in HNSCC patients beyond HPV-positive-status. ARhigh risk may be used to detect HPV-negative patients with an extraordinarily bad prognosis. Most importantly, AR to IMP-1 may serve as a marker for a subgroup of HPV-positive patients that present with a poor prognosis similar to that in HPV-negative patients

    Trail (TNF-related apoptosis-inducing ligand) induces an inflammatory response in human adipocytes

    No full text
    Abstract High serum concentrations of TNF-related apoptosis-inducing ligand (TRAIL), a member of the tumor necrosis factor protein family, are found in patients with increased BMI and serum lipid levels. In a model of murine obesity, both the expression of TRAIL and its receptor (TRAIL-R) is elevated in adipose tissue. Accordingly, TRAIL has been proposed as an important mediator of adipose tissue inflammation and obesity-associated diseases. The aim of this study was to investigate if TRAIL regulates inflammatory processes at the level of the adipocyte. Using human Simpson-Golabi-Behmel syndrome (SGBS) cells as a model system, we found that TRAIL induces an inflammatory response in both preadipocytes and adipocytes. It stimulates the expression of interleukin 6 (IL-6), interleukin 8 (IL-8) as well as the chemokines monocyte chemoattractant protein-1 (MCP-1) and chemokine C-C motif ligand 20 (CCL-20) in a time- and dose-dependent manner. By using small molecule inhibitors, we found that both the NFĪŗB and the ERK1/2 pathway are crucial for mediating the effect of TRAIL. Taken together, we identified a novel pro-inflammatory function of TRAIL in human adipocytes. Our findings suggest that targeting the TRAIL/TRAIL-R system might be a useful strategy to tackle obesity-associated adipose tissue inflammation

    Patterns of antibody responses to nonviral cancer antigens in head and neck squamous cell carcinoma patients differ by human papillomavirus status

    Get PDF
    There have been hints that nonviral cancer antigens are differentially expressed in human papillomavirus (HPV)ā€positive and HPVā€negative head and neck squamous cell carcinoma (HNSCC). Antibody responses (AR) to cancer antigens may be used to indirectly determine cancer antigen expression in the tumor using a noninvasive and tissueā€saving liquid biopsy. Here, we set out to characterize AR to a panel of nonviral cancer antigens in HPVā€positive and HPVā€negative HNSCC patients. A fluorescent microbead multiplex serology to 29 cancer antigens (16 cancerā€testis antigens, 5 cancerā€retina antigens and 8 oncogenes) and 29 HPVā€antigens was performed in 382 HNSCC patients from five independent cohorts (153 HPVā€positive and 209 HPVā€negative). AR to any of the cancer antigens were found in 272/382 patients (72%). The ten most frequent AR were CT47, cTAGE5a, cā€myc, LAGEā€1, MAGEā€A1, ā€A3, ā€A4, NYā€ESOā€1, SpanXā€a1 and p53. AR to MAGEā€A3, MAGEā€A9 and p53 were found at significantly different prevalences by HPV status. An analysis of AR mean fluorescent intensity values uncovered remarkably different AR clusters by HPV status. To identify optimal antigen selections covering a maximum of patients with ā‰¤10 AR, multiobjective optimization revealed distinct antigen selections by HPV status. We identified that AR to nonviral antigens differ by HPV status indicating differential antigen expression. Multiplex serology may be used to characterize antigen expression using serum or plasma as a tissueā€sparing liquid biopsy. Cancer antigen panels should address the distinct antigen repertoire of HPVā€positive and HPVā€negative HNSCC
    • ā€¦
    corecore