Expression levels of p18INK4C modify the cellular efficacy of cyclin-dependent kinase inhibitors via regulation of Mcl-1 expression in tumor cell lines

2009 ◽  
Vol 8 (6) ◽  
pp. 1460-1472 ◽  
Author(s):  
Tomohiro Eguchi ◽  
Hiraku Itadani ◽  
Toshiyasu Shimomura ◽  
Nobuhiko Kawanishi ◽  
Hiroshi Hirai ◽  
...  
Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4504-4504
Author(s):  
Suzanne van Dorp ◽  
Samantha Hol ◽  
Victoria Marcu-Malina ◽  
Nicole Thuss ◽  
Henk Lokhorst ◽  
...  

Abstract Abstract 4504 Introduction Currently innate immune cells such as γ9δ2T cells are explored in tumor immunotherapy e.g. by adoptive transfer of in vitro expanded bulk γ9δ2T cells. Objective We speculated that γ9δ2T cells are highly variable in function and specificity due to differences in γ9δ2TCR, NKG2D and KIR expression and that efficacy of adoptively transferred γ9δ2T cells can be increased by transfer of defined subpopulations or clones rather than bulk γ9δ2T cells. Methods A variety of γ9δ2T cell clones, derived from a healthy donor, were tested for expression levels of γ9δ2TCR, NKG2D and KIRs by flow cytometry analysis. The sequence of the γ9δ2TCR of different clones was further analyzed. Reactivity of γ9δ2T cell clones to a panel of tumor and normal cell lines was tested and these functional analyses were correlated to receptor expression levels and compared with bulk γ9δ2T cells of the same donor. Results Functional analyses revealed a high interclonal variability in recognizing leukemia or solid tumor cell lines. Consequently, γ9δ2T cell clones with high anti-tumor reactivity were superior in killing tumor cells when compared to bulk γ9δ2T cells. Different variable regions of γ9δ2-chains and different expression levels of NKG2D and KIRs were detected in multiple clones. No correlations could be found between TCR, NKG2D, and KIR expression on γ9δ2T cell clones and their response to different tumor cell lines when clones expressed different γ9δ2TCRs. However, analysis of γ9δ2T-cell clones with identical γ9δ2TCRs revealed that a clone with higher reactivity against cancer cells expresses higher amounts of NKG2D and lower inhibitory KIRs when compared to a clone with lower reactivity. Conclusion γ9δ2TCR, NKG2D and KIR expression in γ9δ2T cells is highly variable and cannot be directly correlated to an effective anti-tumor response. Only, when T-cells express one defined γ9δ2TCR, a modulating activity of NKG2D and KIRs can be observed. Thus, we conclude that anti-tumor reactivity is fine tuned by all three receptors and thereby we speculate that the γ9δ2TCR defines tumor-specificity and activity is further modulated by NKG2D and KIRs. These results support the application of distinct subpopulations or of genetically engineered γδT cells with defined receptors rather than bulk γ9δ2T cells in the context of anti-tumor immunotherapies. Disclosures: No relevant conflicts of interest to declare.


2008 ◽  
Vol 19 (3) ◽  
pp. 1139-1151 ◽  
Author(s):  
Jian-Ying Chuang ◽  
Yi-Ting Wang ◽  
Shiu-Hwa Yeh ◽  
Yi-Wen Liu ◽  
Wen-Chang Chang ◽  
...  

The transcription factor Sp1 is ubiquitously expressed in different cells and thereby regulates the expression of genes involved in many cellular processes. This study reveals that Sp1 was phosphorylated during the mitotic stage in three epithelial tumor cell lines and one glioma cell line. By using different kinase inhibitors, we found that during mitosis in HeLa cells, the c-Jun NH2-terminal kinase (JNK) 1 was activated that was then required for the phosphorylation of Sp1. In addition, blockade of the Sp1 phosphorylation via inhibition JNK1 activity in mitosis resulted in the ubiquitination and degradation of Sp1. JNK1 phosphorylated Sp1 at Thr278/739. The Sp1 mutated at Thr278/739 was unstable during mitosis, possessing less transcriptional activity for the 12(S)-lipoxygenase expression and exhibiting a decreased cell growth rate compared with wild-type Sp1 in HeLa cells. In N-methyl-N-nitrosourea–induced mammary tumors, JNK1 activation provided a potential relevance with the accumulation of Sp1. Together, our results indicate that JNK1 activation is necessary to phosphorylate Sp1 and to shield Sp1 from the ubiquitin-dependent degradation pathway during mitosis in tumor cell lines.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2469-2469
Author(s):  
Ana Acuña-Villaorduña ◽  
Douglas W. McMillin ◽  
Constantine S. Mitsiades

Abstract Abstract 2469 Introduction: Kinases are integral components of diverse signaling cascades which can mediate microenvironment-derived resistance of tumor cells to apoptosis. We thus hypothesized that tumor cell-microenvironment interactions alter the anti-tumor activity of multi-targeted kinase inhibitors and could contibute to discordant efficacy results in conventional preclinical models which do not account for these interactions vs. in clinical studies. Methods: We evaluated 6 multi-targeted kinase inhibitors initially developed to target IGF1R (XL-228), Akt/p70S6K (EXEL-6075), B-/C-RAF(EXEL-0094), CHK1/2 (XL844), met/VEGFR2(EXEL-6323) and JAK2 (XL019). Compounds were tested (0–20 μM, 48 hrs) against luciferase-expressing human multiple myeloma (MM) (n=8), mantle cell lymphoma (n=2) and breast cancer (n=1) cell lines. For each of these treatments, tumor cells were cultured in isolation or co-cultured with immortalized human bone marrow stromal cells (BMSCs, n=2); lung fibroblasts; hepatocytes; or brain astrocytes. Tumor cell viability was quantified by tumor cell compartment-specific bioluminescence imaging (CS-BLI). For each compound and tumor cell line, the area under the dose-response curve (AUC) was calculated as % of the AUC that corresponds to 100% tumor cell viability at all doses, to provide cumulative measure of drug activity across the range of tested drug concentrations. Differences in % AUCs in presence vs. absence of accessory cells were compared by 2-tailed paired t-test. The average value and skewness of distribution for log(10)IC50 of enzymatic activity for each kinase and inhibitor tested were calculated for quantitative comparison of multi-targeted nature of these compounds. Results: Co-culture of most tumor cell lines with accessory cells triggered resistance for 5 of 6 compounds tested, as evidenced by statistically significant (p<0.001 for all tests) increases in %AUC with vs. without co-culture with accessory cells (average %AUC differences of 4.7%, 23.4%, 6.2%, 9.7% and 10.8% for IGF1R, Akt/p70S6K, B-/C-RAF, CHK1/2; and met/VEGFR2 inhibitors, respectively, with corresponding 95% C.I. of 2.0–7.5%, 18.2–28.6%, 2.0–10.4%, 5.3–14.1%, and 3.9–17.6%). In contrast, co-culture with accessory cells increased the response of most tumor cell lines to JAK2 inhibitor (average AUC decrease of 13.8%, 95%CI 1.9–16.4, p<0.001). Results were consistent and statistically significant when analysis was restricted to MM cell lines only. For some inhibitors, heterogeneous responses of different tumor cells to accessory cells were noted. For example, for met/VEGFR2 and CHK1/2 inhibitors, 58.0% (40/69) and 60.0% (36/60) of experimental conditions, respectively, showed co-culture-induced resistance, while 31.9% (22/69) and 18.0% (11/60) of cocultures, respectively, showed sensitization to these inhibitors. High frequency of sensitization was observed in co-cultures of a bortezomib/Dex-resistant subline of MM1S cells treated with met/VEGFR2 inhibitor (63.6%, 7/11 of co-cultures tested), and in co-cultures with BMSCs treated with CHK1/2 inhibitor (29.6% of co-cultures tested). The degree of sensitization or resistance by accessory cells (%AUC difference with vs. without co-culture) did not correlate with quantitative measures of multi-targeted nature for each inhibitor. Conclusions: Accessory cells from different tissues significantly modulated tumor cell responses to diverse multi-targeted kinase inhibitors. Co-cultures enhanced the anti-tumor activity of JAK2 inhibitor, but decreased the activity of the other compounds tested. These different responses did not correlate with how multitargeted each inhibitor was. The sensitization to the JAK inhibitor may specifically reflect increased dependency on JAK signaling for tumor cells when they interact with accessory cells. Accessory cell-induced resistance to some kinase inhibitors (e.g. IGF1R, Akt, CHK1–2) may account for observed differences in their preclinical vs. clinical efficacy. These results provide a rationale for preclinical testing of compounds against large panels of tumor cell lines and clinically-relevant accessory cell types to help develop therapeutics against tumors localized in specific tissue microenvironmental niches. Disclosures: McMillin: Axios Biosciences: Equity Ownership. Mitsiades:Millennium Pharmaceuticals: Honoraria; Celgene: Honoraria; Novartis Pharmaceuticals: Honoraria; Bristol-Myers Squibb: Honoraria; Merck &Co.: Honoraria; Centocor: Honoraria; Arno Therapeutics: Honoraria; Amgen: Research Funding; AVEO Pharma: Research Funding; OSI: Research Funding; EMD Serono: Research Funding; Sunesis: Research Funding; Johnson & Johnson: Research Funding; PharmaMar: Licensing royalties Other; Axios Biosciences: Uncompensated Role as advisor, Uncompensated Role as advisor Other.


2017 ◽  
Vol 14 (2) ◽  
pp. 2537-2544 ◽  
Author(s):  
Shohei Morishita ◽  
Takehiro Suzuki ◽  
Yuki Niwa ◽  
Naoshi Dohmae ◽  
Siro Simizu

Pharmacology ◽  
2006 ◽  
Vol 77 (1) ◽  
pp. 11-16 ◽  
Author(s):  
Hans Prenen ◽  
Gunther Guetens ◽  
Gert de Boeck ◽  
Maria Debiec-Rychter ◽  
Paul Manley ◽  
...  

2011 ◽  
Vol 2011 ◽  
pp. 1-13 ◽  
Author(s):  
Hua C. Gong ◽  
Sean Wang ◽  
Gary Mayer ◽  
Guoan Chen ◽  
Glen Leesman ◽  
...  

We profiled receptor tyrosine kinase pathway activation and key gene mutations in eight human lung tumor cell lines and 50 human lung tumor tissue samples to define molecular pathways. A panel of eight kinase inhibitors was used to determine whether blocking pathway activation affected the tumor cell growth. The HER1 pathway in HER1 mutant cell lines HCC827 and H1975 were found to be highly activated and sensitive to HER1 inhibition. H1993 is a c-MET amplified cell line showing c-MET and HER1 pathway activation and responsiveness to c-MET inhibitor treatment. IGF-1R pathway activated H358 and A549 cells are sensitive to IGF-1R inhibition. The downstream PI3K inhibitor, BEZ-235, effectively inhibited tumor cell growth in most of the cell lines tested, except the H1993 and H1650 cells, while the MEK inhibitor PD-325901 was effective in blocking the growth of KRAS mutated cell line H1734 but not H358, A549 and H460. Hierarchical clustering of primary tumor samples with the corresponding tumor cell lines based on their pathway signatures revealed similar profiles for HER1, c-MET and IGF-1R pathway activation and predict potential treatment options for the primary tumors based on the tumor cell lines response to the panel of kinase inhibitors.


Sign in / Sign up

Export Citation Format

Share Document