Skip to main content
Erschienen in: Breast Cancer Research 1/2012

Open Access 01.02.2012 | Research article

Alterations in tumor necrosis factor signaling pathways are associated with cytotoxicity and resistance to taxanes: a study in isogenic resistant tumor cells

verfasst von: Jason A Sprowl, Kerry Reed, Stephen R Armstrong, Carita Lanner, Baoqing Guo, Irina Kalatskaya, Lincoln Stein, Stacey L Hembruff, Adam Tam, Amadeo M Parissenti

Erschienen in: Breast Cancer Research | Ausgabe 1/2012

insite
INHALT
download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Introduction

The taxanes paclitaxel and docetaxel are widely used in the treatment of breast, ovarian, and other cancers. Although their cytotoxicity has been attributed to cell-cycle arrest through stabilization of microtubules, the mechanisms by which tumor cells die remains unclear. Paclitaxel has been shown to induce soluble tumor necrosis factor alpha (sTNF-α) production in macrophages, but the involvement of TNF production in taxane cytotoxicity or resistance in tumor cells has not been established. Our study aimed to correlate alterations in the TNF pathway with taxane cytotoxicity and the acquisition of taxane resistance.

Methods

MCF-7 cells or isogenic drug-resistant variants (developed by selection for surviving cells in increasing concentrations of paclitaxel or docetaxel) were assessed for sTNF-α production in the absence or presence of taxanes by enzyme-linked immunosorbent assay (ELISA) and for sensitivity to docetaxel or sTNF-α by using a clonogenic assay (in the absence or presence of TNFR1 or TNFR2 neutralizing antibodies). Nuclear factor (NF)-κB activity was also measured with ELISA, whereas gene-expression changes associated with docetaxel resistance in MCF-7 and A2780 cells were determined with microarray analysis and quantitative reverse transcription polymerase chain reaction (RTqPCR).

Results

MCF-7 and A2780 cells increased production of sTNF-α in the presence of taxanes, whereas docetaxel-resistant variants of MCF-7 produced high levels of sTNF-α, although only within a particular drug-concentration threshold (between 3 and 45 nM). Increased production of sTNF-α was NF-κB dependent and correlated with decreased sensitivity to sTNF-α, decreased levels of TNFR1, and increased survival through TNFR2 and NF-κB activation. The NF-κB inhibitor SN-50 reestablished sensitivity to docetaxel in docetaxel-resistant MCF-7 cells. Gene-expression analysis of wild-type and docetaxel-resistant MCF-7, MDA-MB-231, and A2780 cells identified changes in the expression of TNF-α-related genes consistent with reduced TNF-induced cytotoxicity and activation of NF-κB survival pathways.

Conclusions

We report for the first time that taxanes can promote dose-dependent sTNF-α production in tumor cells at clinically relevant concentrations, which can contribute to their cytotoxicity. Defects in the TNF cytotoxicity pathway or activation of TNF-dependent NF-κB survival genes may, in contrast, contribute to taxane resistance in tumor cells. These findings may be of strong clinical significance.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​bcr3083) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

JAS performed the majority of the experiments associated with this investigation and wrote the manuscript. KR performed ELISA and RTqPCR experiments and assisted in preparing the manuscript. SA and CL generated the A2780DXL cell line and assisted with microarray experiments. BG conducted the microarray experiments and data analysis and deposited microarray data in the Gene Expression Omnibus database. JAS, SLH, and AT performed the clonogenic assays. IK and LS performed the FI network analyses. AMP participated in the conception and design of the study, assisted in the interpretation of the data, reviewed the manuscript, and made final manuscript revisions. All authors read and approved the final manuscript.
Abkürzungen
A2780DXL
docetaxel-resistant A2780 cells (405 nM)
ANOVA
analysis of variance
DISC
death-inducing signaling complex
ELISA
enzyme-linked immunosorbent assay
MCF-7CC cells
co-cultured control MCF-7 cells propagated in the absence of drug
MCF-7TAX-1
first MCF-7 cell-line selection with paclitaxel
MCF-7TAX-2
second MCF-7 cell-line selection with paclitaxel
MCF-7TXT8
docetaxel-resistant MCF-7 cells selected to dose 8 (1.11 nM)
MCF-7TXT9
docetaxel-resistant MCF-7 cells selected to dose 9 (3.33 nM)
MCF-7TXT10
docetaxel-resistant MCF-7 cells selected to dose 10 (5.00 nM)
MCF-7TXT11
docetaxel-resistant MCF-7 cells selected to dose 11 (15 nM)
MCF-7TXT12
docetaxel-resistant MCF-7 cells selected to dose 11 (45 nM)
mTNF-α
soluble tumor necrosis factor alpha
RTqPCR
reverse transcriptase quantitative polymerase chain reaction
NF-kB
nuclear factor-kappaB
sTNFα
soluble tumor necrosis factor alpha
TNF
tumor necrosis factor
TNF-α
tumor necrosis factor alpha
TNFR1
tumor necrosis factor receptor 1
TNFR2
tumor necrosis factor receptor 2.

Introduction

Taxanes are a family of chemotherapy drugs used to treat various human cancer types [16]. The most common family members include paclitaxel and docetaxel, which block microtubule depolymerization, inducing cell-cycle arrest at mitosis and multinucleation of tumor cells [7, 8]. Taxanes also reduce tumor angiogenesis and cell migration, while stimulating the immune system against neoplasms [911].
Another mechanism for taxane cytotoxicity may involve tumor-necrosis factor (TNF)-α production, because paclitaxel has been shown to augment TNF-α levels in murine macrophages [12]. TNF-α is a membrane-integrated cytokine (mTNF-α) generally produced in activated macrophages and monocytes [13], which can be released from cells in a soluble form (sTNF-α) by the action of the metalloproteinase ADAM-17 [14]. The release of sTNF-α from cells can then induce cell death or a cell-survival response, depending on the receptor to which it binds: TNFR1 or TNFR2, respectively (reviewed in [15]. Although mTNF-α binds with equal affinity to both receptors, sTNF-α preferentially binds to TNFR1, which has a death-effector domain that induces caspase-8 cleavage and apoptosis [16, 17]. Unlike TNFR1, TNFR2 does not contain a death domain. Limited reports suggest that TNFR2 activation promotes cell death, although the mechanism for this is poorly understood and may require the presence of TNFR1 [18, 19]. Nevertheless, TNFR2 has been shown to induce NF-κB activity and cell survival [20].
Many mechanisms associated with resistance to taxanes have been identified in vitro, such as overexpression of the drug-efflux pump Abcb1, β-tubulin gene mutations, or overexpression of β-tubulin (type III); however, their clinical relevance remains unclear [21, 22]. Multiple mechanisms of taxane resistance likely occur simultaneously in cells [23]. To restore tumor sensitivity to taxanes appreciably in cancer patients, all clinically relevant mechanisms of docetaxel resistance must be identified.
To better understand the various pathways associated with taxane resistance, our laboratory selected MCF-7 breast tumor cells for survival in increasing concentrations (doses) of paclitaxel (MCF-7TAX-1 cells) [24] or docetaxel (MCF-7TXT cells) [23]. Increased Abcb1 expression and decreased taxane uptake occurred on acquisition of taxane resistance in these cells, but a pan-ABC transporter inhibitor that restored taxane uptake had no or only a partial effect on drug sensitivity in these cells [23]. Therefore, additional mechanisms must contribute to taxane resistance, and these cell lines may serve as an attractive tool for assessing the possible role of TNF-α and other pathways in taxane cytotoxicity or resistance.
This study reveals for the first time that docetaxel concentrations of 3 nM or greater induce tumor necrosis factor (TNF) expression in MCF-7 cells, and that acquisition of docetaxel resistance can be temporally correlated with elevations in cellular TNF-α levels resistance to TNF-α cytotoxicity, degradation of TNFR1, and promotion of TNFR2-induced survival pathways through the activation of NF-κB. In further support of the role of TNF in taxane cytotoxicity and resistance, we also report that both paclitaxel and docetaxel can induce TNF-α expression in A2780 ovarian carcinoma cells. In addition, we report the consistent alteration in networks of TNF-related genes on acquisition of docetaxel resistance in breast and ovarian tumor cells.

Materials and methods

Cell culture and maintenance

MCF-7 cells from the American Tissue Culture Collection (catalog number HTB-22) were cultured or selected for survival in increasing doses of docetaxel or paclitaxel, as previously described [23, 24]. The initial concentrations of docetaxel and paclitaxel used to begin selection (dose 1) were 0.51 and 0.56 nM, respectively. Cells selected to docetaxel concentrations of 1.11 nM (dose 8, MCF-7TXT8), 3.33 nM (dose 9, MCF-7TXT9), 5.00 nM (dose 10, MCF-7TXT10), 15 nM (dose 11, MCF-7TXT11), and 45 nM (dose 12, MCF-7TXT12) were used in this study. Numbers in subscripts of cell-line names refer to the maximal docetaxel dose level to which the cells were exposed. The paclitaxel-resistant cell line used in this study was selected in an identical manner to a final concentration of 6.64 nM paclitaxel (MCF-7TAX-1 cells; hyphenated number indicates the first cell-line selection, not drug dose). MCF-7 cells were also "selected" in the absence of taxanes to passage numbers similar to those of drug-selected cells to control for genotypic or phenotypic changes associated with long-term culture ("co-cultured control" MCF-7CC cells). A2780 ovarian carcinoma cells from the European Collection of Cell Cultures were also selected for resistance to docetaxel in an identical manner (A2780DXL cells), including the creation of "co-cultured control" A2780CC cells (Armstrong et al., unpublished data).

Measurement of sTNF-α and sTNFR1 in cell-culture media

Concentrated proteins from the medium of 2 million MCF-7CC, MCF-7TXT, or A2780 cells (grown in culture in the absence or presence of various concentrations of paclitaxel or docetaxel) were assessed for levels of sTNF-α or sTNFR1 by using ELISA kits from R&D Systems, following the manufacturer's instructions.

Clonogenic assays

Cellular sensitivity to TNF-α or docetaxel was assessed by using a clonogenic assay, as described previously [24]. Docetaxel resistance factors for the cell lines were determined by dividing the median inhibitory concentration (IC50) for docetaxel in the taxane-resistant cell lines by the IC50 for MCF-7CC cells. In some experiments, cells were exposed to 1 μg/ml cycloheximide, TNFR1 or TNFR2 neutralizing antibodies from R&D Systems (both at 5 μg/ml), or a peptide from Calbiochem Laboratories (La Jolla, CA), which potently blocks NF-κB function by inhibiting translocation of the NF-κB complex into the nucleus [25] (SN-50, 7 μg/ml). A control peptide at the same concentration (SN-50 M) was used in the latter experiments to assess the specificity of NF-κB inhibition.

Immunoblotting analysis

MCF-7CC, MCF-7TXT, and MCF-7TAX-1 cells were incubated in the absence or presence of 20 ng/ml TNF-α for 24 hours. Cells were extracted in RIPA buffer, and 100 μg of extract proteins assessed for the expression of specific proteins by using standard immunoblotting procedures, as previously described [24]. Antibodies used in these experiments included TNFR1-, TNFR2-, and IκB-specific antibodies from Cell Signaling Technology (Danvers, MA) and a mouse-derived GAPDH antibody from Santa Cruz Laboratories. Densitometric quantitation of bands generated by the IκB antibody was performed by using AlphaEaseFC software (Alpha Innotech, San Leandro, CA). Band intensity was normalized relative to GAPDH band intensity.

Quantification of TNFR1 and TNF-α transcript levels by RTqPCR

The levels of TNFR1 and TNF-α transcripts in MCF-7CC and MCF-7TXT10 cells were assessed as described previously [26] by using the following primers: TNFR1: forward, 5'-ACTGCCTCAGCTGCTCCAAAT-3'; reverse, 5'-CCGGTCCACTGTGCAAGAA-3', TNF-α: forward, 5'-TCTTCTCGAACCCCGAGTGA-3', reverse, 5'-GGAGCTGCCCCT-CAGCTT-3'; and S28: forward, 5'-TCCATCATCCGCAATGTAAAAG-3', reverse, 5'-GCTTCTGCGTCTGACTCCAAA-3'.

Measurement of NF-κB activity

MCF-7CC and MCF-7TXT cells were cultured in the presence or absence of 50 nM docetaxel for 24 hours. The activity of the NF-κB p65 and p50 subunits in 10 μg of nuclear extracts was assessed as outlined in the TransAM NF-κB Family ELISA kit (Active Motif, Carlsbad, CA). Readings at 450 nm were normalized to the sum of all readings on the plate to compare across triplicate experiments.

Identification of changes in gene expression associated with the acquisition of docetaxel resistance

Agilent 4 × 44 k human genome oligonucleotide arrays were used to profile differences in gene expression between MCF-7TXT and MCF-7cc cells at selection dose 10 and between docetaxel-resistant and wild-type A2780 ovarian carcinoma cells (A2780DXL and A2780 cells, respectively) at the maximally tolerated dose by using MIAME standards [27]. RNA was isolated from each cell line by using RNeasy Mini kits (Qiagen, Mississauga, ON), and 500 ng of each RNA preparation was labeled and amplified by using Agilent Quick Amp labeling kits. The labeling and array hybridization procedures were performed as per the manufacturer's protocol for a two-color microarray experiment.

Identification of differences in gene expression associated with docetaxel resistance

The hybridized microarrays were scanned by using Agilent scanners and feature extraction software (version 10_7_3_1), and differentially expressed genes associated with the acquisition of docetaxel resistance were identified by using Partek Genomic Suite (Partek, Inc., St. Louis, MO). The background-corrected intensity values were used for analysis. A three-way ANOVA was performed to identify significant changes in gene expression by using the Method of Moments [28]. Genes with greater than twofold differences in gene expression were selected with a false discovery rate of either 0.05 or 0.01 [29]. The data from these array experiments were deposited in the National Centre for Biotechnology Information Gene Expression Omnibus database (accession number GSE26129) [30].

Network-based analysis of gene expression

To determine whether the previously described changes in gene expression associated with acquisition of docetaxel resistance in breast or ovarian tumor cells may reflect changes in the function of specific biochemical pathways in these cells, the genes identified as being associated with docetaxel resistance were subjected to functional-interaction (FI) network analysis [31]. In brief, the FI network covers ~50% of the human proteome representing more than 200,000 functional interactions. Pairwise shortest paths among genes of interest in the FI network were calculated and hierarchically clustered (based on the average-linkage method). Clusters were then selected containing more than 90% of altered genes. To calculate a P value for the average shortest path, we performed a 1,000-fold permutation test by randomly selecting the same number of genes from the biggest connected network component. A minimum spanning tree algorithm was used to find linkers that connected all genes of interest in one subnetwork [32]. We used the Markov Cluster Algorithm (MCL) [33] with inflation of 1.6 for network clustering. Only the biggest clusters with numbers of proteins not less than 2% of the total network were taken into account. All network diagrams were drawn by using Cytoscape [34]. The functional enrichment analysis for pathways was based on a binominal test. A false discovery rate was calculated based on 1,000 permutations on all genes in the FI network. This network-based analysis was also applied to another dataset that documents differences in gene expression between docetaxel-resistant and parental MDA-MB-231 breast cancer cell lines (Gene Expression Omnibus (GEO) accession number GSE28784).

Confirmation of microarray-based changes in gene expression by reverse transcription quantitative polymerase chain reaction

A number of the TNF-α-related genes in these networks were further assessed for expression in wild-type and docetaxel-resistant MCF-7 and A2780 cells with reverse transcription quantitative polymerase chain reaction (RTqPCR) by using the primers depicted in Table 1 and the method described earlier.
Table 1
Primers selected for confirmation of changes in the expression of tumor necrosis factor-α-related genes by reverse transcription quantitative polymerase chain reaction
Gene
Forward primer
Reverse primer
S28
5'TCC ATC ATC CGC AAT GTA AAG-3'
5'-GCT TCT CGC TCT GAC TCC AAA-3'
TNFAIP3
5'-GAC CAT GGC ACA ACT CAT CTC A-3'
5'-GTT AGC TTC ATC CAA CTT TGC GGC ATT G-3'
TNFSF10
5'-CGT GTA CTT TAC CAA CGA GCT GA-3'
5'-ACG GAG TTG CCA CTT GAC TTG-3'
TNFSF13
5'-ACT CTC AGT TGC CCT CTG GTT G-3'
5'-GGA ACT CTG CTC CGG GAG ACT C-3'
TNFSF14
5'-TTT GCT CCA CAG TTG GCC TAA TC-3'
5'-CAA TGA CTG TGG CCT CAC CTT C-3'
TLR1
5'-GGT ACC AGG CCC TCT TCC TCG TTA G-3'
5'-TAG GAA CGT GGA TGA GAC CG TTT TT-3'
TLR6
5'-GCA AAA ACC CTT CAC CTT GTT TTT C-3'
5'-CCA AGT CGT TTC TAT GTG GTT GAG G-3'
BIRC3
5'-TGT TGG GAA TCT GGA GAT GA-3'
5'-CGG ATG AAC TCC TGT CCT TT-3'
TNFR1
5'-ACT GCC TCA GCT GCT CCA AAT-3'
5'-CCG GTC CAC TGT GCA AGA A-3'
TNFα
5'-TCT TCT CGA ACC CCG AGT GA-3'
5'-GGA GCT GCC CCT CAG CTT-3'
Reaction conditions were as described in Materials and methods, and expression was assessed relative to that of S28, the internal reference gene.

Results

Docetaxel increases sTNF-α production in MCF-7CC and A2780 cells

MCF-7CC and A2780CC cells secreted low levels of sTNF-α (1.69 × 10-18 ± 0.40 × 10-18 g/cell and 3.02 × 10-18 ± 0.28 × 10-18 g/cell, respectively). These levels were not significantly changed when cells were treated with 0.1 to 1 nM docetaxel. In contrast, media extracted from MCF-7CC cells treated with ≥ 3 nM docetaxel produced significantly elevated levels of sTNF-α (Figure 1a). A2780 cells produced even greater amounts of TNF-α in response to docetaxel (Figure 1a). Interestingly, the taxane paclitaxel (at concentrations ≥ 15 nM) induced even higher levels of sTNF-α production than docetaxel in A2780 cells (Figure 1b). Given the stronger induction of TNF-α by docetaxel in A2780 cells, we then assessed whether upstream mechanisms responsible for TNF-α induction in A2780 cells were similar to those of macrophages. Comparable to the induction of TNF-α expression by lipopolysaccharides in macrophages [35], we observed that TNF-α induction by docetaxel in A2780 cells was dependent on NF-κB, because an inhibitor of this transcription factor (SN-50) significantly reduced the induction of TNF-α by docetaxel (Figure 1c). The basal amount of sTNF-α production and the magnitude of docetaxel-induced sTNF-α production varied between experiments (compare Figures 1a and 1c for 45 nM docetaxel). Nevertheless, the sTNF-α levels were consistently and substantially higher in cells treated with taxanes. The extent of TNF-α induction by the taxanes appeared to decrease at higher docetaxel concentrations, possibly because of other deleterious effects of these agents on cells at the higher doses.

Selection of MCF-7 cells in increasing concentrations of docetaxel results in acquisition of progressive docetaxel resistance above a threshold dose

Increasing exposure of MCF-7 cells to docetaxel up to a con{centration of 1.1 nM (dose 8, MCF-7TXT8 cells) did not affect docetaxel sensitivity (Figure 2). However, selection to 3.33 nM docetaxel (dose 9, MCF-7TXT9 cells) resulted in an 11.4-fold resistance to docetaxel. Above this threshold, resistance factors increased to 16.6, 32.8, and 184 for cells selected to final docetaxel concentrations of 5 nM (dose 10, MCF-7TXT10 cells), 15 nM (dose 11, MCF-7TXT11 cells), and 45 nM (dose 12, MCF-7TXT12 cells), respectively. Interestingly, MCF-7TXT cells exhibited an even greater cross-resistance to paclitaxel, with resistance factors of 148 and 251 at selection doses 11 and 12, respectively [23]. The resistance factor for MCF-7 cells selected for resistance to paclitaxel at the maximally tolerated dose (MCF-7TAX-1 cells) was 42. These cells also exhibited strong cross-resistance to docetaxel (46-fold) [24]. In contrast, ovarian A2780 cells could be selected for resistance to considerably higher concentrations of docetaxel. A2780DXL cells at their maximally tolerated dose (405 nM) exhibited ~4,000-fold resistance to docetaxel (Armstrong et al., unpublished data).

Effects of docetaxel on sTNFα in MCF-7CC and MCF-7TXT cell lines

MCF-7CC and MCF-7TXT8 cells secreted low amounts of TNF-α (11.5 × 10-18 ± 0.4 × 10-18 g/cell and 5.5 × 10-18 ± 1.4 × 10-18 g/cell, respectively). When these cell lines were exposed to 50 nM docetaxel, no significant difference in sTNF-α secretion was observed (Figure 3a). In contrast, untreated MCF-7TXT9 and MCF-7TXT10 cells secreted 31.8-fold and 18.2-fold higher levels of sTNF-α than did MCF-7CC cells (P < 0.0001), and addition of 50 nM docetaxel increased sTNF-α production a further 1.62-fold and 1.27-fold, respectively (P < 0.01). sTNF-α levels returned to basal levels in MCF-7TXT11 and MCF-7TXT12 cells, even after treatment with 50 nM docetaxel. No differences in sTNF-α levels were observed between MCF-7CC and MCF-7TAX-1 cells, in the presence or absence of docetaxel (data not shown). TNF-α transcript levels in MCF-7TXT10 cells (relative to S28 expression) were 198.5 ± 30.5 higher than the levels of this transcript in MCF-7CC cells (Figure 3b), suggesting that elevated secretion of sTNF-α is likely due to dramatically increased expression of TNF-α transcripts and protein.

MCF-7TXT and MCF-7TAX-1 cells are resistant to TNF-α-induced cytotoxicity

TNF-α (10 ng/ml) reduced colony formation in a clonogenic assay by 79.8% ± 6.0% and 66.6% ± 1.7% for MCF-7CC and MCF-7TXT8 cells, respectively (P < 0.0001) (Figure 4a). In contrast, MCF-7TXT9, MCF-7TXT10, MCF-7TXT11, and MCF-7TAX-1 cells all had similar levels of colony formation in the absence or presence of 10 ng/ml TNF-α, indicating substantial TNF-α resistance. TNF actually increased colony formation in MCF-7TXT12 cells, possibly because of a high level of activation of growth and survival pathways in these cells at the highest selection dose, some of which are TNF-α dependent (see Discussion). The cell lines were also cultured in the presence of varying concentrations of TNF-α. Colony formation was very strongly reduced in MCF-7CC cells in the presence of 50 or 100 ng/ml TNF-α (P < 0.0001) (Figure 4b). Reductions in colony formation were much smaller for MCF-7TXT10 cells treated with 50 ng/ml or 100 ng/ml TNF-α, again indicating resistance to TNF-α cytotoxicity in docetaxel-resistant cells. MCF-7TAX-1 cells treated with 10 ng/ml TNF-α formed similar numbers of colonies as did untreated cells, suggesting that these cells were also resistant to TNF-α. However, TNF-α concentrations of 50 or 100 ng/ml induced strong reductions in colony formation relative to MCF-7TXT10 cells, suggesting greater resistance to TNF-α in the former cell line than in the latter.

TNFR1 protein levels (but not transcript levels) are reduced on acquisition of docetaxel resistance in MCF-7 cells

Unlike TNFR2, the levels of TNFR1 protein (as measured in immunoblotting experiments) decreased on acquisition of docetaxel resistance at dose 9 (MCF-7TXT9 cells) and remained low in MCF-7TXT10 and MCF-7TXT12 cells (Figure 5a). Interestingly, RTqPCR analysis revealed no significant differences in TNFR1 transcript expression between these cell lines (Figure 5b). Similar soluble TNFR1 (sTNFR1) levels were observed in MCF-7CC and MCF-7TXT8 cells (Figure 5c), although levels decreased in MCF-7TXT9 and MCF-7TXT10 cells (P < 0.001). sTNFR1 levels then returned to those of MCF-7TXT8 cells as docetaxel-selective pressure was increased.

Induction of docetaxel resistance in MCF-7 cells through application of a TNFR1 neutralizing antibody

Significant differences in colony formation were observed between TNFR1 neutralizing antibody-treated MCF-7CC cells and untreated cells when incubated with 1.23 nM (P < 0.0001), 0.41 nM (P = 0.0002), 0.14 nM (P = 0.0006), and 0.046 nM (P < 0.0001) docetaxel (Figure 5d). Nonlinear regression curve-fitting programs revealed that MCF-7CC cells incubated with the TNFR1 neutralizing antibody were about 2.25-fold more resistant to docetaxel than were untreated cells, consistent with a role for the TNF-α pathway in docetaxel cytotoxicity.

Activation of NF-κB on acquisition of docetaxel resistance

Unlike MCF-7TAX-1 cells, MCF-7TXT10 cells had 35% lower IκB levels than did MCF-7CC cells (P = 0.03) (Figure 6a). Measurement of NF-κB binding in nuclear extracts from MCF-7 and MCF-7TXT8 cells revealed low binding of NF-κB p65 and p50 subunits to the NF-κB transcription factor binding site (Figures 6b and 6c). In contrast, nuclear extracts from MCF-7TXT9 and MCF-7TXT10 cells exhibited more than threefold higher levels of subunit binding to the NF-κB sequence compared with equivalent extracts from MCF-7CC cells (P < 0.05). This binding was reduced as cells were exposed to higher docetaxel selection doses. Interestingly, 50 nM docetaxel induced even higher levels of p65 and p50 subunit binding in MCF-7CC and MCF-7TXT cells, except when docetaxel selection doses were more than 15 nM (doses 11 and 12).

Promotion of TNF-α cytotoxicity in MCF-7TXT10 cells by cycloheximide or a TNFR2 neutralizing antibody

As previously observed, exposure of MCF-7CC cells to 10 ng/ml TNF-α strongly decreased colony formation in a clonogenic assay, whereas MCF-7TXT10 cells exhibited significant resistance to TNF-α (Figure 7a). The addition of the protein-synthesis inhibitor cycloheximide, 5 μg/ml, restored the ability of TNF-α to be cytotoxic to MCF-7TXT10 cells, while having only a small additional effect on TNF-α cytotoxicity in MCF-7CC cells. These observations suggested that a protein, possibly NF-κB, is critical for maintaining resistance to TNF-α.
To test this hypothesis, and because NF-κB is activated on TNF-α binding to TNFR2, resulting in enhanced expression of survival genes [20], we theorized that docetaxel cytotoxicity might be increased in MCF-7TXT10 cells on addition of a TNFR2-neutralizing antibody or an inhibitor of NF-κB function. Supporting this conjecture, we observed a greater reduction in colony formation for TNFR2-neutralizing antibody-treated cells than untreated cells when treated with 41.2 nM (P = 0.0007), 13.7 nM (P = 0.005), 4.5 nM (P = 0.006), or 1.7 nM (P = 0.01) docetaxel (Figure 7b). Nonlinear regression curve-fitting for three independent experiments revealed that the TNFR2 neutralizing antibody rendered MCF-7TXT10 cells 2.13-fold more sensitive to docetaxel than were untreated cells. Moreover, as shown in Figure 7c the peptide SN-50, which contains the nuclear localization signal of NF-κB and thus blocks the transcription factor translocation to the nucleus [25], increased docetaxel cytotoxicity to an even greater degree in MCF-7TXT10 cells (7.1-fold). In contrast, a control peptide (SN-50 M), in which critical basic amino acids within the nuclear localization signal are replaced with uncharged amino acids, had no effect on docetaxel sensitivity (Figure 7c).

Network-based analysis of genes associated with the acquisition of docetaxel resistance

Assessment of microarray data by using an FI network approach (see Materials and methods) revealed 2,235 genes that were differently expressed between parental and docetaxel-resistant MCF-7 breast cancer cell lines (fold-change > 2.0 and FDR ≤ 0.05). Of these, 834 (37.3%) were in the FI network, and hierarchic clustering reduced this to a set of 753 of the most interconnected candidates. This gene set was then used for further analyses. The average shortest-distance calculation showed that genes in this set were linked together much more tightly than would be expected by chance alone (P < 0.001), indicating that these differentially expressed genes occupy a small corner of the large FI network space. A subnetwork was built from these 753 genes by adding the minimum number of linker genes required to form fully connected networks involving these genes. The resulting networks consisted of 938 genes, 185 of which were linkers. A Markov clustering algorithm was then used to identify clusters of proteins (coded by the genes) that are highly interconnected with each other and less connected to the outside world. This algorithm identified 14 clusters consisting of more than 20 genes, including a cluster of 22 TNF-associated genes and eight linkers (Figure 8a).
We used an identical approach to identify clusters of differentially expressed genes between wild-type and docetaxel-resistant A2780 ovarian carcinoma cells. Of 955 genes that were differentially expressed between the two cell lines, a network of 11 TNF-related genes and three linkers was identified (Figure 8b). When the same approach was used to identify networks of genes differentially expressed between docetaxel-sensitive and docetaxel-resistant MDA-MB-231 cells (data obtained from GEO, accession number GSE28784), a cluster of 22 TNF-related genes and three linkers was identified (see Figure 8c).

Confirmation of changes in the expression of TNF-α-dependent genes by RTqPCR

The expression of a selected number of genes within the previously identified TNF-α signaling networks was quantitatively assessed with RTqPCR. As shown in Figure 9, a generally strong concordance was noted between changes in gene expression identified by microarray analysis and those determined by RTqPCR (12 of 14 gene-expression changes assessed). Six TNF-α-dependent genes were confirmed to have altered expression on selection of MCF-7 cells for resistance to docetaxel, including TNFSF13, TNFSF10, TLR6, TNFAIP3, TNFSF14, and BIRC3 (the latter two genes being upregulated 30-fold and 21-fold, respectively). Three of these genes were also upregulated in A2780DXL cells (BIRC3, TLR6, and TNFSF10, which increased expression almost 300-fold).

Discussion

Although taxanes are known to inhibit cell division by preventing microtubule depolymerization and inducing multinucleation [8, 36], it is unclear whether these are their sole mechanisms of tumor cell growth arrest/death in vitro and in vivo. Paclitaxel has been shown to increase sTNF-α release from murine macrophages [12, 37], although the levels used in those studies would be unachievable in patients, and docetaxel had no effect on TNF-α expression in the same study. In our study, we showed for the first time that docetaxel (at concentrations between 3 and 45 nM) can stimulate TNF-α production and sTNF-α release from both breast and ovarian tumor cells. Such concentrations are clearly in the range of plasma levels of docetaxel in breast cancer patients after docetaxel infusion (10 to 75 nM) [38] and are likely sufficiently high to induce TNF expression in even poorly vascularized tumors. This newly identified TNF-dependent mechanism of docetaxel action may also account for its reported immunomodulatory activity [11, 39]. In addition, we show in this article that paclitaxel treatment (at 5 and 15 nM concentrations) can dramatically increase sTNF-α release from ovarian tumor cells.
Our study also illustrates that the acquisition of docetaxel resistance in breast tumor cells temporally correlates with increased production and release of sTNF-α from cells, despite the ability of sTNF-α to be cytotoxic to cells [40]. However, the onset of docetaxel resistance in MCF-7 cells (at docetaxel selection doses ≥ 3.33 nM) also correlated with strongly reduced levels of TNFR1, which would block the ability of TNF-α to induce cell death. Although the mechanism responsible for TNFR1 reduction remains undefined, neither changes in TNFR1 transcript levels nor increased levels of sTNFR1 in the media were found, suggesting that the receptor was not shed from cells by the ADAM-17 protease [14]. In fact, MCF-7TXT9 and MCF-7TXT10 cells exhibited decreased levels of sTNFR1 in the medium in which it was grown. It is possible that increased levels of sTNFα produced by these cells bound to sTNFR1 in the medium, preventing its detection by the TNFR1 antibody. Taken together, our findings suggest that downregulation of TNFR1 occurs posttranscriptionally, because of either reduced translation of the TNFR1 transcript or increased TNFR1 proteolysis.
A recent study [41] found that TNF-α or paclitaxel induced NF-κB activity in C2C12 myotubes. However, paclitaxel did not induce increased TNF-α production, and inhibition of TNFR1 blocked TNF-α-induced NF-κB activation but did not abolish paclitaxel-induced NF-κB activity [41]. It is important to note that, in these studies, TNF-α levels were assessed only 4 hours after treatment with paclitaxel (10 nM to 10 μM).
Whereas docetaxel selection doses between 3 and 5 nM resulted in highly elevated sTNF-α production, higher selection doses (≥ 15 nM) did not. This was despite the ability of the drug to induce TNF-α production in wild-type cells over a large concentration range (Figure 1). This may be explained by the increased expression of the Abcb1 drug transporter and reduced docetaxel uptake that we observed in MCF-7TXT11 and MCF-7TXT12 cells. Expression was maximal at the highest selection doses (≥ 15 nM) [42]. We propose that docetaxel accumulates at sufficient concentrations to induce production of sTNF-α in MCF-7TXT9 and MCF-7TXT10 cells. However, at or above 15 nM docetaxel, MCF-7TXT cells exhibit reduced drug uptake, such that docetaxel accumulation is insufficient to stimulate TNF-α production.
The mechanism for resistance to taxanes and TNF-α in MCF-7TAX-1 cells appears to differ from that of MCF-7TXT cells. TNFR1 levels were equivalent in MCF-7TAX-1 and MCF-7CC cells (data not shown), and IκB levels were also unchanged during selection for paclitaxel resistance (Figure 6). Because only cells exposed to the maximally tolerated dose of paclitaxel were retained during selection of MCF-7TAX-1 cells, it is likely that cells selected at lower doses could have exhibited elevated production of TNF-α and TNF-α-mediated NF-κB activation. However, survival by circumventing the TNF-α ability to stimulate TNFR1-induced cytotoxicity must lie downstream of the receptor. MCF-7TAX-1 cells are also high expressors of Abcb1 [24]. Interestingly, another paclitaxel-resistant MCF-7 cell line (MCF-7TAX-2 cells) [23] retained sensitivity to TNF-α (data not shown), suggesting that defects in the TNF-α pathway are not critical for taxane resistance in vitro. Nevertheless, considering that three of the four taxane-resistant cell lines exhibited alterations in TNF-α signaling and that docetaxel has been shown to increase sTNF-α levels in both breast and ovarian tumor cells, it appears that we have identified a common but unknown mechanism of taxane cytotoxicity and resistance that warrants further study for its potential clinical relevance.
To provide further support for a general involvement of the TNF-α pathway in docetaxel cytotoxicity and in the induction of docetaxel resistance, we also showed in this study that selection of breast and ovarian tumor cells for resistance to docetaxel results in changes in the expression of networks of genes related to TNF-α signaling (Figure 8 and Table 2). Quite strikingly, the vast majority of the upregulated genes depicted in Table 2 code for proteins that are TNF-ligand family members, TNF-receptor family members, TNF receptor-associated proteins, TNF-dependent activators of NF-κB, or proteins that help promote degradation of the inhibitor of NF-κB (IκB). Other upregulated genes are TNF-dependent inhibitors of apoptosis. Downregulated genes code for proteins that inhibit the activation of NF-kB or promote apoptosis. The net effect of the changes in gene expression would thus be to promote the ability of TNF to augment NF-κB-dependent cell survival, while blocking its ability to induce tumor cell death via activation of TNFR1.
Table 2
Tumor-necrosis factor α (TNFα)- and nuclear factor (NF)-κB-related genes associated with the acquisition of docetaxel resistance in MCF-7 breast tumor cells, MDA-MB-231 breast tumor cells, and A2780 ovarian carcinoma cells
Gene
Direction and magnitude of change in expressiona
Role of gene product
Refs.
Changes in the expression of TNFα- or NF-κB-related genes in MCF-7TXT10 cells relative to MCF-7CC10 cells
TNF
Increased 12.6, 1.43
Binds to TNFR1 to promote cell death and to TNFR2 to stimulate expression of NF-κB-dependent survival genes
[54, 55]
TNFSF10
Increased 6.88
TNF-like ligand that binds to DcR2 to activate NF-κB-dependent survival genes; also called TRAIL
[56]
TNFRSF19
Increased 16.5, 1.64
Member of the TNF-receptor family that binds to lymphotoxin alpha, activates NF-κB-mediated transcription
[57]
TNFRSF14
Increased, 4.24
TNF-receptor family member, interacts with members of the TNFR-associated factor (TRAF) family and activates NF-κB and AP-1
[58]
TNFSF13
Increased, 3.07
Binds to TNFRSF13B to promote NF-κB activation; also called APRIL
[59]
BIRC3
Increased, 52.2
Member of a family of proteins that inhibits apoptosis by binding to TNFR-associated factors TRAF1 and TRAF2
[60]
TLR2
Increased, 5.19
Stimulates NF-κB activation
[61]
TRIM38
Increased, 4.22, 7.28
Removes Lys63-linked ubiquitin chains from TRAF2 and TRAF6, negatively regulating NF-κB activity
[62]
TNFRSF6B
Decreased, -2.02
Member of the TNF-receptor superfamily, which, on binding of TNF, inhibits cell proliferation and induces apoptosis
[63]
TRAF3IP2
Decreased, -2.14
Associates with and activates I-κB kinase, leading to the liberation of NF-κB from its complex with I-κB
[64]
Changes in the expression of TNFα- or NF-κB-related genes in A2780 DXL cells relative to A2780 cells (MTD)
TNFSF13
Increased, 2.10
Binds to TNFRSF13B to promote NF-κB activation; also called APRIL
[59]
TNFRSF11B
Increased, 2.01
Decoy receptor for the TNF-related apoptosis-inducing ligand TRAIL; confers resistance to TNF- and TRAIL-induced apoptosis
[65]
CARD14
Increased, 2.19, 1.31
Associates with guanylate kinase members that interact with BCL10 and activate NF-κB
[66]
IRAK4
Increased, 3.70, 3.86
Required for the optimal transduction of IL-1-induced signals, including the activation of IRAK-1, NF-κB, and JNK
[67]
TLR1
Increased, 2.79
Acts via MYD88 and TRAF6 to stimulate NF-κB activation, cytokine secretion, and the inflammatory response
[68]
TLR6
Increased, 2.26, 1.57
Also acts via MYD88 and TRAF6 to stimulate NF-κB activation, cytokine secretion, and the inflammatory response
[68]
BIRC3
Increased, 8.83
Member of a family of proteins that inhibits apoptosis by binding to TNFR-associated factors TRAF1 and TRAF2
[60]
RFFL
Increased, 2.15, 2.13
Endosome-associated ubiquitin ligase for RIP and regulates TNF-induced NF-κB activation
[69]
TRAF3IP2
Increased, 2.16
Associates with and activates I-κB kinase, leading to the liberation of NF-κB from its complex with I-κB
[64]
SMPD3
Decreased, -1.59, -2.03
Translocates to the plasma membrane in response to TNF-α in a time- and dose-dependent manner
[70]
SOX9
Decreased, -30.8
Protein whose expression and activity is negatively regulated by TNF-dependent NF-κB activation
[71]
Changes in the expression of TNFα- or NF-κB-related genes in docetaxel-resistant MB-231 cells relative to A2780 cells
CAST
Increased, 2.44
Inhibits degradation of NF-κB to prolong NF-κB activation
[72]
BCL10
Increased, 3.48
Potent activator of NF-κB activity
[73]
RPS6KA3
Increased, 5.95
Phosphorylates I-κB and activates NF-κB in response to TNF
[74]
MAP2K5
Increased, 3.54
A survival protein highly expressed in MCF-7 breast tumor cells resistant to etoposide and TNF-α
[75]
MKNK2
Increased, 1.76
Promotes TNF-α biosynthesis at the posttranscriptional levels
[76]
[75]
TNFRSF10B
Decreased, -1.67
Also known as TRAIL receptor 2; stimulates apoptosis via FADD
[77]
FADD
Decreased, -2.00
A component of the caspase 8-activating complex induced by TNF-α binding to TNFR1
[78]
CRADD
Decreased, -2.43
An adaptor protein that promotes TNFα-induced apoptosis through interaction with the TNFR1-interacting protein RIP
[79]
TRAF1
Decreased, -2.39
Negatively regulates the ability of TNFR2 to promote cell proliferation and NF-κB activation in T cells
[80]
MKNK1
Decreased, -2.00
Promotes TNF-α-mediated mRNA degradation
[81]
Full-genome oligo-based microarray experiments were performed comparing differences in gene expression between wild-type and docetaxel-resistant MCF-7 cell lines. Differentially expressed genes were then classified into various functional interaction networks, as described in Materials and methods. Information on the identities and roles of genes associated with TNF-α signaling are presented in tabular form, with particular emphasis on the products of genes that are known to play a role in the induction of NF-κB-dependent survival genes or in the inhibition of apoptosis. a Numbers represent the fold increase (positive numbers) or decrease (negative numbers) in expression of the gene as revealed by one or more oligo probes on the Agilent 44K human microarrays.
The findings of our study may have significant clinical relevance. A presentation at the 26th annual meeting of the European Association of Urology in 2011 [43] revealed that serum levels of proinflammatory cytokines, including TNF-α, increased 2 days after administration of docetaxel to patients with castration-resistant prostate cancer. Interestingly, these changes in cytokine expression correlated with the induction of apoptosis and with clinical response. In addition, a study presented recently at the American Association for Cancer Research, 101st Annual Meeting [44], revealed that, in patients with serous epithelial ovarian carcinoma, pretreatment tumor expression of various genes within the TNF-α and NF-κB signaling networks could be used to distinguish between responders and nonresponders to paclitaxel/carboplatin chemotherapy. It also was shown in a small study involving patients with locally advanced breast cancer that tumor levels of nuclear (activated) NF-κB could be used to distinguish between responders and nonresponders to neoadjuvant anthracycline- and/or taxane-based chemotherapy regimens [45]. These and other studies strongly support the likely clinical significance of the findings. For example, because TNF-α has been shown to reduce tumor vascularization in mice through its effects on TNFR1-expressing endothelial cells [46], the reported ability of docetaxel to affect tumor angiogenesis [10] may be through an ability of the drug to promote sTNFα-mediated decreases in tumor vascularization. Moreover, one of the well-established dose-limiting toxicities associated with docetaxel chemotherapy in breast cancer patients is fatigue [47], and high TNF levels have been shown to correlate with fatigue onset in cancer patients [48]. Given our findings of docetaxel-induced TNF-α production, perhaps these two phenomena are linked.
Finally, a previous clinical study used a TNF-decoy receptor (entanercept) to permit patients to tolerate higher doses of docetaxel without significant toxicity [49]; however, given our findings, it is not surprising that these blockers would create a greater tolerance to docetaxel, unfortunately at the likely expense of lesser anti-tumor efficacy. Our findings further question the utility of administering docetaxel to cancer patients on TNF-α blockers for treatment of co-morbid inflammatory diseases.

Conclusions

Our study provides evidence for the first time that taxanes can induce sTNF-α expression in two tumor cell lines of different tissue origin. Although this would promote the cytotoxicity of docetaxel, continued exposure to the drug appears to result in a downregulation of TNF-α-mediated cytotoxicity, while promoting TNF-α-dependent activation of NF-kB-dependent cell-survival pathways and the inhibition of apoptosis. In addition, although drug-resistance studies often involve selection of cells to maximally tolerated drug doses [50, 51], our study illustrates the critical role that the drug-selection dose may play on the mechanisms by which tumor cells acquire chemotherapy resistance. At lower doses of taxanes (3 to 5 nM), TNF-α-mediated activation of NF-κB-dependent cell-survival pathways appears to be an important mechanism of taxane resistance, whereas at selection doses ≥ 15 nM docetaxel, the drug induces overexpression of Abcb1, resulting in reduced accumulation of docetaxel into cells and a consequent reduction in docetaxel-stimulated TNF-α production. Since the concentration of docetaxel within patient tumors is typically lower than that present in the vasculature, perhaps the pathways associated with resistance to lower concentrations of docetaxel are of greater clinical relevance. This may explain why Abcb1 inhibitors have had little ability to reverse resistance to taxanes in cancer patients [52, 53]. In addition, given that some cancer patients with inflammatory diseases may be taking TNF blockers, this newly identified role for TNF in docetaxel cytotoxicity may be of particular importance, suggesting that these blockers may compromise the efficacy of docetaxel chemotherapy.

Acknowledgements

These studies were supported by a grant (to A.M.P.) from the Ontario Institute for Cancer Research (02-MAY-0159) and by core support funds from the Northern Cancer Research Foundation.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

JAS performed the majority of the experiments associated with this investigation and wrote the manuscript. KR performed ELISA and RTqPCR experiments and assisted in preparing the manuscript. SA and CL generated the A2780DXL cell line and assisted with microarray experiments. BG conducted the microarray experiments and data analysis and deposited microarray data in the Gene Expression Omnibus database. JAS, SLH, and AT performed the clonogenic assays. IK and LS performed the FI network analyses. AMP participated in the conception and design of the study, assisted in the interpretation of the data, reviewed the manuscript, and made final manuscript revisions. All authors read and approved the final manuscript.
insite
INHALT
download
DOWNLOAD
print
DRUCKEN
Literatur
1.
Zurück zum Zitat Sjostrom J, Blomqvist C, Mouridsen H, Pluzanska A, Ottosson-Lonn S, Bengtsson NO, Ostenstad B, Mjaaland I, Palm-Sjovall M, Wist E, Valvere V, Anderson H, Bergh J: Docetaxel compared with sequential methotrexate and 5-fluorouracil in patients with advanced breast cancer after anthracycline failure: a randomised phase III study with crossover on progression by the Scandinavian Breast Group. Eur J Cancer. 1999, 35: 1194-1201. 10.1016/S0959-8049(99)00122-7.CrossRefPubMed Sjostrom J, Blomqvist C, Mouridsen H, Pluzanska A, Ottosson-Lonn S, Bengtsson NO, Ostenstad B, Mjaaland I, Palm-Sjovall M, Wist E, Valvere V, Anderson H, Bergh J: Docetaxel compared with sequential methotrexate and 5-fluorouracil in patients with advanced breast cancer after anthracycline failure: a randomised phase III study with crossover on progression by the Scandinavian Breast Group. Eur J Cancer. 1999, 35: 1194-1201. 10.1016/S0959-8049(99)00122-7.CrossRefPubMed
2.
Zurück zum Zitat Shepherd FA, Dancey J, Ramlau R, Mattson K, Gralla R, O'Rourke M, Levitan N, Gressot L, Vincent M, Burkes R, Coughlin S, Kim Y, Berille J: Prospective randomized trial of docetaxel versus best supportive care in patients with non-small-cell lung cancer previously treated with platinum-based chemotherapy. J Clin Oncol. 2000, 18: 2095-2103.PubMed Shepherd FA, Dancey J, Ramlau R, Mattson K, Gralla R, O'Rourke M, Levitan N, Gressot L, Vincent M, Burkes R, Coughlin S, Kim Y, Berille J: Prospective randomized trial of docetaxel versus best supportive care in patients with non-small-cell lung cancer previously treated with platinum-based chemotherapy. J Clin Oncol. 2000, 18: 2095-2103.PubMed
3.
Zurück zum Zitat Tannock IF, de WR, Berry WR, Horti J, Pluzanska A, Chi KN, Oudard S, Theodore C, James ND, Turesson I, Rosenthal MA, Eisenberger MA: Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. N Engl J Med. 2004, 351: 1502-1512. 10.1056/NEJMoa040720.CrossRefPubMed Tannock IF, de WR, Berry WR, Horti J, Pluzanska A, Chi KN, Oudard S, Theodore C, James ND, Turesson I, Rosenthal MA, Eisenberger MA: Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. N Engl J Med. 2004, 351: 1502-1512. 10.1056/NEJMoa040720.CrossRefPubMed
4.
Zurück zum Zitat Ajani JA, Fodor MB, Tjulandin SA, Moiseyenko VM, Chao Y, Cabral FS, Majlis A, Assadourian S, Van CE: Phase II multi-institutional randomized trial of docetaxel plus cisplatin with or without fluorouracil in patients with untreated, advanced gastric, or gastroesophageal adenocarcinoma. J Clin Oncol. 2005, 23: 5660-5667. 10.1200/JCO.2005.17.376.CrossRefPubMed Ajani JA, Fodor MB, Tjulandin SA, Moiseyenko VM, Chao Y, Cabral FS, Majlis A, Assadourian S, Van CE: Phase II multi-institutional randomized trial of docetaxel plus cisplatin with or without fluorouracil in patients with untreated, advanced gastric, or gastroesophageal adenocarcinoma. J Clin Oncol. 2005, 23: 5660-5667. 10.1200/JCO.2005.17.376.CrossRefPubMed
5.
Zurück zum Zitat Posner MR: Docetaxel in squamous cell cancer of the head and neck. Anticancer Drugs. 2001, 12 (Suppl 1): S21-S24.PubMed Posner MR: Docetaxel in squamous cell cancer of the head and neck. Anticancer Drugs. 2001, 12 (Suppl 1): S21-S24.PubMed
6.
Zurück zum Zitat Iaffaioli RV, Tortoriello A, Santangelo M, Turitto G, Libutti M, Benassai G, Frattolillo A, Ciccarelli PD, De RP, Crovella F, Carbone I, Barbarisi A: Phase I dose escalation study of gemcitabine and paclitaxel plus colony-stimulating factors in previously treated patients with advanced breast and ovarian cancer. Clin Oncol (R Coll Radiol). 2000, 12: 251-255. Iaffaioli RV, Tortoriello A, Santangelo M, Turitto G, Libutti M, Benassai G, Frattolillo A, Ciccarelli PD, De RP, Crovella F, Carbone I, Barbarisi A: Phase I dose escalation study of gemcitabine and paclitaxel plus colony-stimulating factors in previously treated patients with advanced breast and ovarian cancer. Clin Oncol (R Coll Radiol). 2000, 12: 251-255.
7.
Zurück zum Zitat Ringel I, Horwitz SB: Studies with RP 56976 (taxotere): a semisynthetic analogue of taxol. J Natl Cancer Inst. 1991, 83: 288-291. 10.1093/jnci/83.4.288.CrossRefPubMed Ringel I, Horwitz SB: Studies with RP 56976 (taxotere): a semisynthetic analogue of taxol. J Natl Cancer Inst. 1991, 83: 288-291. 10.1093/jnci/83.4.288.CrossRefPubMed
8.
Zurück zum Zitat Chazard M, Pellae-Cosset B, Garet F, Soares JA, Lucidi B, Lavail Y, Lenaz L: Taxol (paclitaxel), first molecule of a new class of cytotoxic agents: taxanes. Bull Cancer. 1994, 81: 173-181.PubMed Chazard M, Pellae-Cosset B, Garet F, Soares JA, Lucidi B, Lavail Y, Lenaz L: Taxol (paclitaxel), first molecule of a new class of cytotoxic agents: taxanes. Bull Cancer. 1994, 81: 173-181.PubMed
9.
Zurück zum Zitat Haldar S, Basu A, Croce CM: Bcl2 is the guardian of microtubule integrity. Cancer Res. 1997, 57: 229-233.PubMed Haldar S, Basu A, Croce CM: Bcl2 is the guardian of microtubule integrity. Cancer Res. 1997, 57: 229-233.PubMed
10.
Zurück zum Zitat Sweeney CJ, Miller KD, Sissons SE, Nozaki S, Heilman DK, Shen J, Sledge GW: The antiangiogenic property of docetaxel is synergistic with a recombinant humanized monoclonal antibody against vascular endothelial growth factor or 2-methoxyestradiol but antagonized by endothelial growth factors. Cancer Res. 2001, 61: 3369-3372.PubMed Sweeney CJ, Miller KD, Sissons SE, Nozaki S, Heilman DK, Shen J, Sledge GW: The antiangiogenic property of docetaxel is synergistic with a recombinant humanized monoclonal antibody against vascular endothelial growth factor or 2-methoxyestradiol but antagonized by endothelial growth factors. Cancer Res. 2001, 61: 3369-3372.PubMed
11.
Zurück zum Zitat Tong AW, Seamour B, Lawson JM, Ordonez G, Vukelja S, Hyman W, Richards D, Stein L, Maples PB, Nemunaitis J: Cellular immune profile of patients with advanced cancer before and after taxane treatment. Am J Clin Oncol. 2000, 23: 463-472. 10.1097/00000421-200010000-00007.CrossRefPubMed Tong AW, Seamour B, Lawson JM, Ordonez G, Vukelja S, Hyman W, Richards D, Stein L, Maples PB, Nemunaitis J: Cellular immune profile of patients with advanced cancer before and after taxane treatment. Am J Clin Oncol. 2000, 23: 463-472. 10.1097/00000421-200010000-00007.CrossRefPubMed
12.
Zurück zum Zitat Bogdan C, Ding A: Taxol, a microtubule-stabilizing antineoplastic agent, induces expression of tumor necrosis factor alpha and interleukin-1 in macrophages. J Leukoc Biol. 1992, 52: 119-121.PubMed Bogdan C, Ding A: Taxol, a microtubule-stabilizing antineoplastic agent, induces expression of tumor necrosis factor alpha and interleukin-1 in macrophages. J Leukoc Biol. 1992, 52: 119-121.PubMed
13.
Zurück zum Zitat Meloni F: Tumor necrosis factor alpha. Biological aspects. G Ital Chemioter. 1989, 36: 29-37.PubMed Meloni F: Tumor necrosis factor alpha. Biological aspects. G Ital Chemioter. 1989, 36: 29-37.PubMed
14.
Zurück zum Zitat Black RA, Rauch CT, Kozlosky CJ, Peschon JJ, Slack JL, Wolfson MF, Castner BJ, Stocking KL, Reddy P, Srinivasan S, Nelson N, Boiani N, Schooley KA, Gerhart M, Davis R, Fitzner JN, Johnson RS, Paxton RJ, March CJ, Cerretti DP: A metalloproteinase disintegrin that releases tumour-necrosis factor-alpha from cells. Nature. 1997, 385: 729-733. 10.1038/385729a0.CrossRefPubMed Black RA, Rauch CT, Kozlosky CJ, Peschon JJ, Slack JL, Wolfson MF, Castner BJ, Stocking KL, Reddy P, Srinivasan S, Nelson N, Boiani N, Schooley KA, Gerhart M, Davis R, Fitzner JN, Johnson RS, Paxton RJ, March CJ, Cerretti DP: A metalloproteinase disintegrin that releases tumour-necrosis factor-alpha from cells. Nature. 1997, 385: 729-733. 10.1038/385729a0.CrossRefPubMed
16.
Zurück zum Zitat Grell M, Zimmermann G, Hulser D, Pfizenmaier K, Scheurich P: TNF receptors TR60 and TR80 can mediate apoptosis via induction of distinct signal pathways. J Immunol. 1994, 153: 1963-1972.PubMed Grell M, Zimmermann G, Hulser D, Pfizenmaier K, Scheurich P: TNF receptors TR60 and TR80 can mediate apoptosis via induction of distinct signal pathways. J Immunol. 1994, 153: 1963-1972.PubMed
17.
Zurück zum Zitat Hsu H, Xiong J, Goeddel DV: The TNF receptor 1-associated protein TRADD signals cell death and NF-kappa B activation. Cell. 1995, 81: 495-504. 10.1016/0092-8674(95)90070-5.CrossRefPubMed Hsu H, Xiong J, Goeddel DV: The TNF receptor 1-associated protein TRADD signals cell death and NF-kappa B activation. Cell. 1995, 81: 495-504. 10.1016/0092-8674(95)90070-5.CrossRefPubMed
18.
Zurück zum Zitat Declercq W, Denecker G, Fiers W, Vandenabeele P: Cooperation of both TNF receptors in inducing apoptosis: involvement of the TNF receptor-associated factor binding domain of the TNF receptor 75. J Immunol. 1998, 161: 390-399.PubMed Declercq W, Denecker G, Fiers W, Vandenabeele P: Cooperation of both TNF receptors in inducing apoptosis: involvement of the TNF receptor-associated factor binding domain of the TNF receptor 75. J Immunol. 1998, 161: 390-399.PubMed
19.
Zurück zum Zitat Tartaglia LA, Ayres TM, Wong GH, Goeddel DV: A novel domain within the 55 kd TNF receptor signals cell death. Cell. 1993, 74: 845-853. 10.1016/0092-8674(93)90464-2.CrossRefPubMed Tartaglia LA, Ayres TM, Wong GH, Goeddel DV: A novel domain within the 55 kd TNF receptor signals cell death. Cell. 1993, 74: 845-853. 10.1016/0092-8674(93)90464-2.CrossRefPubMed
20.
Zurück zum Zitat Rothe M, Wong SC, Henzel WJ, Goeddel DV: A novel family of putative signal transducers associated with the cytoplasmic domain of the 75 kDa tumor necrosis factor receptor. Cell. 1994, 78: 681-692. 10.1016/0092-8674(94)90532-0.CrossRefPubMed Rothe M, Wong SC, Henzel WJ, Goeddel DV: A novel family of putative signal transducers associated with the cytoplasmic domain of the 75 kDa tumor necrosis factor receptor. Cell. 1994, 78: 681-692. 10.1016/0092-8674(94)90532-0.CrossRefPubMed
21.
Zurück zum Zitat Monzo M, Rosell R, Sanchez JJ, Lee JS, O'Brate A, Gonzalez-Larriba JL, Alberola V, Lorenzo JC, Nunez L, Ro JY, Martin C: Paclitaxel resistance in non-small-cell lung cancer associated with beta-tubulin gene mutations. J Clin Oncol. 1999, 17: 1786-1793.PubMed Monzo M, Rosell R, Sanchez JJ, Lee JS, O'Brate A, Gonzalez-Larriba JL, Alberola V, Lorenzo JC, Nunez L, Ro JY, Martin C: Paclitaxel resistance in non-small-cell lung cancer associated with beta-tubulin gene mutations. J Clin Oncol. 1999, 17: 1786-1793.PubMed
22.
Zurück zum Zitat Mozzetti S, Ferlini C, Concolino P, Filippetti F, Raspaglio G, Prislei S, Gallo D, Martinelli E, Ranelletti FO, Ferrandina G, Scambia G: Class III beta-tubulin overexpression is a prominent mechanism of paclitaxel resistance in ovarian cancer patients. Clin Cancer Res. 2005, 11: 298-305.PubMed Mozzetti S, Ferlini C, Concolino P, Filippetti F, Raspaglio G, Prislei S, Gallo D, Martinelli E, Ranelletti FO, Ferrandina G, Scambia G: Class III beta-tubulin overexpression is a prominent mechanism of paclitaxel resistance in ovarian cancer patients. Clin Cancer Res. 2005, 11: 298-305.PubMed
23.
Zurück zum Zitat Hembruff SL, Laberge ML, Villeneuve DJ, Guo B, Veitch Z, Cecchetto M, Parissenti AM: Role of drug transporters and drug accumulation in the temporal acquisition of drug resistance. BMC Cancer. 2008, 8: 318-10.1186/1471-2407-8-318.CrossRefPubMedPubMedCentral Hembruff SL, Laberge ML, Villeneuve DJ, Guo B, Veitch Z, Cecchetto M, Parissenti AM: Role of drug transporters and drug accumulation in the temporal acquisition of drug resistance. BMC Cancer. 2008, 8: 318-10.1186/1471-2407-8-318.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Guo B, Villeneuve DJ, Hembruff SL, Kirwan AF, Blais DE, Bonin M, Parissenti AM: Cross-resistance studies of isogenic drug-resistant breast tumor cell lines support recent clinical evidence suggesting that sensitivity to paclitaxel may be strongly compromised by prior doxorubicin exposure. Breast Cancer Res Treat. 2004, 85: 31-51. 10.1023/B:BREA.0000021046.29834.12.CrossRefPubMed Guo B, Villeneuve DJ, Hembruff SL, Kirwan AF, Blais DE, Bonin M, Parissenti AM: Cross-resistance studies of isogenic drug-resistant breast tumor cell lines support recent clinical evidence suggesting that sensitivity to paclitaxel may be strongly compromised by prior doxorubicin exposure. Breast Cancer Res Treat. 2004, 85: 31-51. 10.1023/B:BREA.0000021046.29834.12.CrossRefPubMed
25.
Zurück zum Zitat Lin YZ, Yao SY, Veach RA, Torgerson TR, Hawiger J: Inhibition of nuclear translocation of transcription factor NF-kappa B by a synthetic peptide containing a cell membrane-permeable motif and nuclear localization sequence. J Biol Chem. 1995, 270: 14255-14258. 10.1074/jbc.270.24.14255.CrossRefPubMed Lin YZ, Yao SY, Veach RA, Torgerson TR, Hawiger J: Inhibition of nuclear translocation of transcription factor NF-kappa B by a synthetic peptide containing a cell membrane-permeable motif and nuclear localization sequence. J Biol Chem. 1995, 270: 14255-14258. 10.1074/jbc.270.24.14255.CrossRefPubMed
26.
Zurück zum Zitat Hembruff SL, Villeneuve DJ, Parissenti AM: The optimization of quantitative reverse transcription PCR for verification of cDNA microarray data. Anal Biochem. 2005, 345: 237-249. 10.1016/j.ab.2005.07.014.CrossRefPubMed Hembruff SL, Villeneuve DJ, Parissenti AM: The optimization of quantitative reverse transcription PCR for verification of cDNA microarray data. Anal Biochem. 2005, 345: 237-249. 10.1016/j.ab.2005.07.014.CrossRefPubMed
27.
Zurück zum Zitat Brazma A, Hingamp P, Quackenbush J, Sherlock G, Spellman P, Stoeckert C, Aach J, Ansorge W, Ball CA, Causton HC, Gaasterland T, Glenisson P, Holstege FC, Kim IF, Markowitz V, Matese JC, Parkinson H, Robinson A, Sarkans U, Schulze-Kremer S, Stewart J, Taylor R, Vilo J, Vingron M: Minimum information about a microarray experiment (MIAME)-toward standards for microarray data. Nat Genet. 2001, 29: 365-371. 10.1038/ng1201-365.CrossRefPubMed Brazma A, Hingamp P, Quackenbush J, Sherlock G, Spellman P, Stoeckert C, Aach J, Ansorge W, Ball CA, Causton HC, Gaasterland T, Glenisson P, Holstege FC, Kim IF, Markowitz V, Matese JC, Parkinson H, Robinson A, Sarkans U, Schulze-Kremer S, Stewart J, Taylor R, Vilo J, Vingron M: Minimum information about a microarray experiment (MIAME)-toward standards for microarray data. Nat Genet. 2001, 29: 365-371. 10.1038/ng1201-365.CrossRefPubMed
28.
Zurück zum Zitat Eisenhart C: The assumptions underlying the analysis of variance. Biometrics. 1947, 3: 1-21. 10.2307/3001534.CrossRefPubMed Eisenhart C: The assumptions underlying the analysis of variance. Biometrics. 1947, 3: 1-21. 10.2307/3001534.CrossRefPubMed
29.
Zurück zum Zitat Tamhane AC, Dunlop DD: Statistics and Data Analysis from Elementary to Intermediate. 2000, Englewood Cliffs, NJ:Prentice Hall, 473-4. 1 Tamhane AC, Dunlop DD: Statistics and Data Analysis from Elementary to Intermediate. 2000, Englewood Cliffs, NJ:Prentice Hall, 473-4. 1
31.
Zurück zum Zitat Wu G, Feng X, Stein L: A human functional protein interaction network and its application to cancer data analysis. Genome Biol. 2010, 11: R53-10.1186/gb-2010-11-5-r53.CrossRefPubMedPubMedCentral Wu G, Feng X, Stein L: A human functional protein interaction network and its application to cancer data analysis. Genome Biol. 2010, 11: R53-10.1186/gb-2010-11-5-r53.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Gross JL, Yellen J: Graph Theory and Its Applications. 1998, Boca Raton, FL:, CRC Press, 1 Gross JL, Yellen J: Graph Theory and Its Applications. 1998, Boca Raton, FL:, CRC Press, 1
33.
Zurück zum Zitat Enright AJ, Van DS, Ouzounis CA: An efficient algorithm for large-scale detection of protein families. Nucleic Acids Res. 2002, 30: 1575-1584. 10.1093/nar/30.7.1575.CrossRefPubMedPubMedCentral Enright AJ, Van DS, Ouzounis CA: An efficient algorithm for large-scale detection of protein families. Nucleic Acids Res. 2002, 30: 1575-1584. 10.1093/nar/30.7.1575.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, Ideker T: Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003, 13: 2498-2504. 10.1101/gr.1239303.CrossRefPubMedPubMedCentral Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, Ideker T: Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003, 13: 2498-2504. 10.1101/gr.1239303.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Udalova IA, Knight JC, Vidal V, Nedospasov SA, Kwiatkowski D: Complex NF-kappaB interactions at the distal tumor necrosis factor promoter region in human monocytes. J Biol Chem. 1998, 273: 21178-21186. 10.1074/jbc.273.33.21178.CrossRefPubMed Udalova IA, Knight JC, Vidal V, Nedospasov SA, Kwiatkowski D: Complex NF-kappaB interactions at the distal tumor necrosis factor promoter region in human monocytes. J Biol Chem. 1998, 273: 21178-21186. 10.1074/jbc.273.33.21178.CrossRefPubMed
36.
Zurück zum Zitat Moos PJ, Fitzpatrick FA: Taxanes propagate apoptosis via two cell populations with distinctive cytological and molecular traits. Cell Growth Differ. 1998, 9: 687-697.PubMed Moos PJ, Fitzpatrick FA: Taxanes propagate apoptosis via two cell populations with distinctive cytological and molecular traits. Cell Growth Differ. 1998, 9: 687-697.PubMed
37.
Zurück zum Zitat Manthey CL, Qureshi N, Stutz PL, Vogel SN: Lipopolysaccharide antagonists block taxol-induced signaling in murine macrophages. J Exp Med. 1993, 178: 695-702. 10.1084/jem.178.2.695.CrossRefPubMed Manthey CL, Qureshi N, Stutz PL, Vogel SN: Lipopolysaccharide antagonists block taxol-induced signaling in murine macrophages. J Exp Med. 1993, 178: 695-702. 10.1084/jem.178.2.695.CrossRefPubMed
38.
Zurück zum Zitat Clarke SJ, Rivory LP: Clinical pharmacokinetics of docetaxel. Clin Pharmacokinet. 1999, 36: 99-114. 10.2165/00003088-199936020-00002.CrossRefPubMed Clarke SJ, Rivory LP: Clinical pharmacokinetics of docetaxel. Clin Pharmacokinet. 1999, 36: 99-114. 10.2165/00003088-199936020-00002.CrossRefPubMed
39.
Zurück zum Zitat Lee LF, Hellendall RP, Wang Y, Haskill JS, Mukaida N, Matsushima K, Ting JP: IL-8 reduced tumorigenicity of human ovarian cancer in vivo due to neutrophil infiltration. J Immunol. 2000, 164: 2769-2775.CrossRefPubMed Lee LF, Hellendall RP, Wang Y, Haskill JS, Mukaida N, Matsushima K, Ting JP: IL-8 reduced tumorigenicity of human ovarian cancer in vivo due to neutrophil infiltration. J Immunol. 2000, 164: 2769-2775.CrossRefPubMed
40.
Zurück zum Zitat Cai Z, Capoulade C, Moyret-Lalle C, mor-Gueret M, Feunteun J, Larsen AK, Paillerets BB, Chouaib S: Resistance of MCF7 human breast carcinoma cells to TNF-induced cell death is associated with loss of p53 function. Oncogene. 1997, 15: 2817-2826. 10.1038/sj.onc.1201445.CrossRefPubMed Cai Z, Capoulade C, Moyret-Lalle C, mor-Gueret M, Feunteun J, Larsen AK, Paillerets BB, Chouaib S: Resistance of MCF7 human breast carcinoma cells to TNF-induced cell death is associated with loss of p53 function. Oncogene. 1997, 15: 2817-2826. 10.1038/sj.onc.1201445.CrossRefPubMed
41.
Zurück zum Zitat Jackman RW, Rhoads MG, Cornwell E, Kandarian SC: Microtubule-mediated NF-kappaB activation in the TNF-alpha signaling pathway. Exp Cell Res. 2009, 315: 3242-3249. 10.1016/j.yexcr.2009.08.020.CrossRefPubMedPubMedCentral Jackman RW, Rhoads MG, Cornwell E, Kandarian SC: Microtubule-mediated NF-kappaB activation in the TNF-alpha signaling pathway. Exp Cell Res. 2009, 315: 3242-3249. 10.1016/j.yexcr.2009.08.020.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Reed K, Hembruff SL, Laberge ML, Villeneuve DJ, Cote GB, Parissenti AM: Hypermethylation of the ABCB1 downstream gene promoter accompanies ABCB1 gene amplification and increased expression in docetaxel-resistant MCF-7 breast tumor cells. Epigenetics. 2008, 3: 270-280. 10.4161/epi.3.5.6868.CrossRefPubMed Reed K, Hembruff SL, Laberge ML, Villeneuve DJ, Cote GB, Parissenti AM: Hypermethylation of the ABCB1 downstream gene promoter accompanies ABCB1 gene amplification and increased expression in docetaxel-resistant MCF-7 breast tumor cells. Epigenetics. 2008, 3: 270-280. 10.4161/epi.3.5.6868.CrossRefPubMed
43.
Zurück zum Zitat Evans CP: Serum levels of pro-inflammatory cytokines immediately increase two days after application of docetaxel in patients with castration-resistant prostate cancer and correlate with apoptosis and clinical response. 26th Annual Meeting of the Suropean Association of Urology, Vienna, Austria. 2011 Evans CP: Serum levels of pro-inflammatory cytokines immediately increase two days after application of docetaxel in patients with castration-resistant prostate cancer and correlate with apoptosis and clinical response. 26th Annual Meeting of the Suropean Association of Urology, Vienna, Austria. 2011
44.
Zurück zum Zitat Koti M, Vidal R, Nuin P, Haslehurst A, Weberpals J, Childs T, Bryson P, Feilloter HE, Squire J, Park PC: Identification of biomarkers of chemoresistance in serous epithelial ovarian cancer using integrative molecular profiling. American Association for Cancer Research 102nd Annual Meeting. 2011, Orlando, FL, , Abstract 11-A-6026-AACR Koti M, Vidal R, Nuin P, Haslehurst A, Weberpals J, Childs T, Bryson P, Feilloter HE, Squire J, Park PC: Identification of biomarkers of chemoresistance in serous epithelial ovarian cancer using integrative molecular profiling. American Association for Cancer Research 102nd Annual Meeting. 2011, Orlando, FL, , Abstract 11-A-6026-AACR
45.
Zurück zum Zitat Montagut C, Tusquets I, Ferrer B, Corominas JM, Bellosillo B, Campas C, Suarez M, Fabregat X, Campo E, Gascon P, Serrano S, Fernandez PL, Rovira A, Albanell J: Activation of nuclear factor-kappa B is linked to resistance to neoadjuvant chemotherapy in breast cancer patients. Endocr Relat Cancer. 2006, 13: 607-616. 10.1677/erc.1.01171.CrossRefPubMed Montagut C, Tusquets I, Ferrer B, Corominas JM, Bellosillo B, Campas C, Suarez M, Fabregat X, Campo E, Gascon P, Serrano S, Fernandez PL, Rovira A, Albanell J: Activation of nuclear factor-kappa B is linked to resistance to neoadjuvant chemotherapy in breast cancer patients. Endocr Relat Cancer. 2006, 13: 607-616. 10.1677/erc.1.01171.CrossRefPubMed
46.
Zurück zum Zitat Stoelcker B, Ruhland B, Hehlgans T, Bluethmann H, Luther T, Mannel DN: Tumor necrosis factor induces tumor necrosis via tumor necrosis factor receptor type 1-expressing endothelial cells of the tumor vasculature. Am J Pathol. 2000, 156: 1171-1176. 10.1016/S0002-9440(10)64986-3.CrossRefPubMedPubMedCentral Stoelcker B, Ruhland B, Hehlgans T, Bluethmann H, Luther T, Mannel DN: Tumor necrosis factor induces tumor necrosis via tumor necrosis factor receptor type 1-expressing endothelial cells of the tumor vasculature. Am J Pathol. 2000, 156: 1171-1176. 10.1016/S0002-9440(10)64986-3.CrossRefPubMedPubMedCentral
47.
Zurück zum Zitat Schroder CP, de ML, Westermann AM, Smit WM, Creemers GJ, de GH, Stouthard JM, van DG, Erjavec Z, van BA, Vader W, Willemse PH: Weekly docetaxel in metastatic breast cancer patients: No superior benefits compared to three-weekly docetaxel. Eur J Cancer. 2011, 47: 1355-62. 10.1016/j.ejca.2010.12.018.CrossRefPubMed Schroder CP, de ML, Westermann AM, Smit WM, Creemers GJ, de GH, Stouthard JM, van DG, Erjavec Z, van BA, Vader W, Willemse PH: Weekly docetaxel in metastatic breast cancer patients: No superior benefits compared to three-weekly docetaxel. Eur J Cancer. 2011, 47: 1355-62. 10.1016/j.ejca.2010.12.018.CrossRefPubMed
48.
Zurück zum Zitat Aouizerat BE, Dodd M, Lee K, West C, Paul SM, Cooper BA, Wara W, Swift P, Dunn LB, Miaskowski C: Preliminary evidence of a genetic association between tumor necrosis factor alpha and the severity of sleep disturbance and morning fatigue. Biol Res Nurs. 2009, 11: 27-41. 10.1177/1099800409333871.CrossRefPubMed Aouizerat BE, Dodd M, Lee K, West C, Paul SM, Cooper BA, Wara W, Swift P, Dunn LB, Miaskowski C: Preliminary evidence of a genetic association between tumor necrosis factor alpha and the severity of sleep disturbance and morning fatigue. Biol Res Nurs. 2009, 11: 27-41. 10.1177/1099800409333871.CrossRefPubMed
49.
Zurück zum Zitat Monk JP, Phillips G, Waite R, Kuhn J, Schaaf LJ, Otterson GA, Guttridge D, Rhoades C, Shah M, Criswell T, Caligiuri MA, Villalona-Calero MA: Assessment of tumor necrosis factor alpha blockade as an intervention to improve tolerability of dose-intensive chemotherapy in cancer patients. J Clin Oncol. 2006, 24: 1852-1859. 10.1200/JCO.2005.04.2838.CrossRefPubMed Monk JP, Phillips G, Waite R, Kuhn J, Schaaf LJ, Otterson GA, Guttridge D, Rhoades C, Shah M, Criswell T, Caligiuri MA, Villalona-Calero MA: Assessment of tumor necrosis factor alpha blockade as an intervention to improve tolerability of dose-intensive chemotherapy in cancer patients. J Clin Oncol. 2006, 24: 1852-1859. 10.1200/JCO.2005.04.2838.CrossRefPubMed
50.
Zurück zum Zitat Yoo GH, Piechocki MP, Ensley JF, Nguyen T, Oliver J, Meng H, Kewson D, Shibuya TY, Lonardo F, Tainsky MA: Docetaxel induced gene expression patterns in head and neck squamous cell carcinoma using cDNA microarray and PowerBlot. Clin Cancer Res. 2002, 8: 3910-3921.PubMed Yoo GH, Piechocki MP, Ensley JF, Nguyen T, Oliver J, Meng H, Kewson D, Shibuya TY, Lonardo F, Tainsky MA: Docetaxel induced gene expression patterns in head and neck squamous cell carcinoma using cDNA microarray and PowerBlot. Clin Cancer Res. 2002, 8: 3910-3921.PubMed
51.
Zurück zum Zitat Duan Z, Lamendola DE, Duan Y, Yusuf RZ, Seiden MV: Description of paclitaxel resistance-associated genes in ovarian and breast cancer cell lines. Cancer Chemother Pharmacol. 2005, 55: 277-285. 10.1007/s00280-004-0878-y.CrossRefPubMed Duan Z, Lamendola DE, Duan Y, Yusuf RZ, Seiden MV: Description of paclitaxel resistance-associated genes in ovarian and breast cancer cell lines. Cancer Chemother Pharmacol. 2005, 55: 277-285. 10.1007/s00280-004-0878-y.CrossRefPubMed
52.
Zurück zum Zitat Pusztai L, Wagner P, Ibrahim N, Rivera E, Theriault R, Booser D, Symmans FW, Wong F, Blumenschein G, Fleming DR, Rouzier R, Boniface G, Hortobagyi GN: Phase II study of tariquidar, a selective P-glycoprotein inhibitor, in patients with chemotherapy-resistant, advanced breast carcinoma. Cancer. 2005, 104: 682-691. 10.1002/cncr.21227.CrossRefPubMed Pusztai L, Wagner P, Ibrahim N, Rivera E, Theriault R, Booser D, Symmans FW, Wong F, Blumenschein G, Fleming DR, Rouzier R, Boniface G, Hortobagyi GN: Phase II study of tariquidar, a selective P-glycoprotein inhibitor, in patients with chemotherapy-resistant, advanced breast carcinoma. Cancer. 2005, 104: 682-691. 10.1002/cncr.21227.CrossRefPubMed
53.
Zurück zum Zitat van der Holt B, Lowenberg B, Burnett AK, Knauf WU, Shepherd J, Piccaluga PP, Ossenkoppele GJ, Verhoef GE, Ferrant A, Crump M, Selleslag D, Theobald M, Fey MF, Vellenga E, Dugan M, Sonneveld P: The value of the MDR1 reversal agent PSC-833 in addition to daunorubicin and cytarabine in the treatment of elderly patients with previously untreated acute myeloid leukemia (AML), in relation to MDR1 status at diagnosis. Blood. 2005, 106: 2646-2654. 10.1182/blood-2005-04-1395.CrossRefPubMed van der Holt B, Lowenberg B, Burnett AK, Knauf WU, Shepherd J, Piccaluga PP, Ossenkoppele GJ, Verhoef GE, Ferrant A, Crump M, Selleslag D, Theobald M, Fey MF, Vellenga E, Dugan M, Sonneveld P: The value of the MDR1 reversal agent PSC-833 in addition to daunorubicin and cytarabine in the treatment of elderly patients with previously untreated acute myeloid leukemia (AML), in relation to MDR1 status at diagnosis. Blood. 2005, 106: 2646-2654. 10.1182/blood-2005-04-1395.CrossRefPubMed
54.
Zurück zum Zitat Gupta S: A decision between life and death during TNF-alpha-induced signaling. J Clin Immunol. 2002, 22: 185-194. 10.1023/A:1016089607548.CrossRefPubMed Gupta S: A decision between life and death during TNF-alpha-induced signaling. J Clin Immunol. 2002, 22: 185-194. 10.1023/A:1016089607548.CrossRefPubMed
55.
Zurück zum Zitat Karin M, Gallagher E: TNFR signaling: ubiquitin-conjugated TRAFfic signals control stop-and-go for MAPK signaling complexes. Immunol Rev. 2009, 228: 225-240. 10.1111/j.1600-065X.2008.00755.x.CrossRefPubMed Karin M, Gallagher E: TNFR signaling: ubiquitin-conjugated TRAFfic signals control stop-and-go for MAPK signaling complexes. Immunol Rev. 2009, 228: 225-240. 10.1111/j.1600-065X.2008.00755.x.CrossRefPubMed
56.
Zurück zum Zitat gli-Esposti MA, Dougall WC, Smolak PJ, Waugh JY, Smith CA, Goodwin RG: The novel receptor TRAIL-R4 induces NF-kappaB and protects against TRAIL-mediated apoptosis, yet retains an incomplete death domain. Immunity. 1997, 7: 813-820. 10.1016/S1074-7613(00)80399-4.CrossRef gli-Esposti MA, Dougall WC, Smolak PJ, Waugh JY, Smith CA, Goodwin RG: The novel receptor TRAIL-R4 induces NF-kappaB and protects against TRAIL-mediated apoptosis, yet retains an incomplete death domain. Immunity. 1997, 7: 813-820. 10.1016/S1074-7613(00)80399-4.CrossRef
57.
Zurück zum Zitat Hashimoto T, Schlessinger D, Cui CY: Troy binding to lymphotoxin-alpha activates NF kappa B mediated transcription. Cell Cycle. 2008, 7: 106-111. 10.4161/cc.7.1.5135.CrossRefPubMed Hashimoto T, Schlessinger D, Cui CY: Troy binding to lymphotoxin-alpha activates NF kappa B mediated transcription. Cell Cycle. 2008, 7: 106-111. 10.4161/cc.7.1.5135.CrossRefPubMed
58.
Zurück zum Zitat Marsters SA, Ayres TM, Skubatch M, Gray CL, Rothe M, Ashkenazi A: Herpesvirus entry mediator, a member of the tumor necrosis factor receptor (TNFR) family, interacts with members of the TNFR-associated factor family and activates the transcription factors NF-kappaB and AP-1. J Biol Chem. 1997, 272: 14029-14032. 10.1074/jbc.272.22.14029.CrossRefPubMed Marsters SA, Ayres TM, Skubatch M, Gray CL, Rothe M, Ashkenazi A: Herpesvirus entry mediator, a member of the tumor necrosis factor receptor (TNFR) family, interacts with members of the TNFR-associated factor family and activates the transcription factors NF-kappaB and AP-1. J Biol Chem. 1997, 272: 14029-14032. 10.1074/jbc.272.22.14029.CrossRefPubMed
59.
Zurück zum Zitat Wu Y, Bressette D, Carrell JA, Kaufman T, Feng P, Taylor K, Gan Y, Cho YH, Garcia AD, Gollatz E, Dimke D, LaFleur D, Migone TS, Nardelli B, Wei P, Ruben SM, Ullrich SJ, Olsen HS, Kanakaraj P, Moore PA, Baker KP: Tumor necrosis factor (TNF) receptor superfamily member TACI is a high affinity receptor for TNF family members APRIL and BLyS. J Biol Chem. 2000, 275: 35478-35485. 10.1074/jbc.M005224200.CrossRefPubMed Wu Y, Bressette D, Carrell JA, Kaufman T, Feng P, Taylor K, Gan Y, Cho YH, Garcia AD, Gollatz E, Dimke D, LaFleur D, Migone TS, Nardelli B, Wei P, Ruben SM, Ullrich SJ, Olsen HS, Kanakaraj P, Moore PA, Baker KP: Tumor necrosis factor (TNF) receptor superfamily member TACI is a high affinity receptor for TNF family members APRIL and BLyS. J Biol Chem. 2000, 275: 35478-35485. 10.1074/jbc.M005224200.CrossRefPubMed
60.
Zurück zum Zitat Liston P, Roy N, Tamai K, Lefebvre C, Baird S, Cherton-Horvat G, Farahani R, McLean M, Ikeda JE, MacKenzie A, Korneluk RG: Suppression of apoptosis in mammalian cells by NAIP and a related family of IAP genes. Nature. 1996, 379: 349-353. 10.1038/379349a0.CrossRefPubMed Liston P, Roy N, Tamai K, Lefebvre C, Baird S, Cherton-Horvat G, Farahani R, McLean M, Ikeda JE, MacKenzie A, Korneluk RG: Suppression of apoptosis in mammalian cells by NAIP and a related family of IAP genes. Nature. 1996, 379: 349-353. 10.1038/379349a0.CrossRefPubMed
61.
Zurück zum Zitat Scharf S, Hippenstiel S, Flieger A, Suttorp N, N'guessan PD: Induction of human {beta}-Defensin-2 in pulmonary epithelial cells by Legionella pneumophila: Involvement of TLR2 and TLR5, p38 MAPK, JNK, NF-{kappa}B and AP-1. Am J Physiol Lung Cell Mol Physiol. 2010, 298: L687-L695. 10.1152/ajplung.00365.2009.CrossRefPubMed Scharf S, Hippenstiel S, Flieger A, Suttorp N, N'guessan PD: Induction of human {beta}-Defensin-2 in pulmonary epithelial cells by Legionella pneumophila: Involvement of TLR2 and TLR5, p38 MAPK, JNK, NF-{kappa}B and AP-1. Am J Physiol Lung Cell Mol Physiol. 2010, 298: L687-L695. 10.1152/ajplung.00365.2009.CrossRefPubMed
62.
Zurück zum Zitat Brummelkamp TR, Nijman SM, Dirac AM, Bernards R: Loss of the cylindromatosis tumour suppressor inhibits apoptosis by activating NF-kappaB. Nature. 2003, 424: 797-801. 10.1038/nature01811.CrossRefPubMed Brummelkamp TR, Nijman SM, Dirac AM, Bernards R: Loss of the cylindromatosis tumour suppressor inhibits apoptosis by activating NF-kappaB. Nature. 2003, 424: 797-801. 10.1038/nature01811.CrossRefPubMed
63.
Zurück zum Zitat Chen G, Rong M, Luo D: TNFRSF6B neutralization antibody inhibits proliferation and induces apoptosis in hepatocellular carcinoma cell. Pathol Res Pract. 2010, 206: 631-641. 10.1016/j.prp.2010.05.011.CrossRefPubMed Chen G, Rong M, Luo D: TNFRSF6B neutralization antibody inhibits proliferation and induces apoptosis in hepatocellular carcinoma cell. Pathol Res Pract. 2010, 206: 631-641. 10.1016/j.prp.2010.05.011.CrossRefPubMed
64.
Zurück zum Zitat Li X, Commane M, Nie H, Hua X, Chatterjee-Kishore M, Wald D, Haag M, Stark GR: Act1, an NF-kappa B-activating protein. Proc Natl Acad Sci USA. 2000, 97: 10489-10493.CrossRefPubMedPubMedCentral Li X, Commane M, Nie H, Hua X, Chatterjee-Kishore M, Wald D, Haag M, Stark GR: Act1, an NF-kappa B-activating protein. Proc Natl Acad Sci USA. 2000, 97: 10489-10493.CrossRefPubMedPubMedCentral
65.
Zurück zum Zitat Rachner TD, Benad P, Rauner M, Goettsch C, Singh SK, Schoppet M, Hofbauer LC: Osteoprotegerin production by breast cancer cells is suppressed by dexamethasone and confers resistance against TRAIL-induced apoptosis. J Cell Biochem. 2009, 108: 106-116. 10.1002/jcb.22232.CrossRefPubMed Rachner TD, Benad P, Rauner M, Goettsch C, Singh SK, Schoppet M, Hofbauer LC: Osteoprotegerin production by breast cancer cells is suppressed by dexamethasone and confers resistance against TRAIL-induced apoptosis. J Cell Biochem. 2009, 108: 106-116. 10.1002/jcb.22232.CrossRefPubMed
66.
Zurück zum Zitat Bertin J, Wang L, Guo Y, Jacobson MD, Poyet JL, Srinivasula SM, Merriam S, DiStefano PS, Alnemri ES: CARD11 and CARD14 are novel caspase recruitment domain (CARD)/membrane-associated guanylate kinase (MAGUK) family members that interact with BCL10 and activate NF-kappa B. J Biol Chem. 2001, 276: 11877-11882. 10.1074/jbc.M010512200.CrossRefPubMed Bertin J, Wang L, Guo Y, Jacobson MD, Poyet JL, Srinivasula SM, Merriam S, DiStefano PS, Alnemri ES: CARD11 and CARD14 are novel caspase recruitment domain (CARD)/membrane-associated guanylate kinase (MAGUK) family members that interact with BCL10 and activate NF-kappa B. J Biol Chem. 2001, 276: 11877-11882. 10.1074/jbc.M010512200.CrossRefPubMed
67.
Zurück zum Zitat Lye E, Mirtsos C, Suzuki N, Suzuki S, Yeh WC: The role of interleukin 1 receptor-associated kinase-4 (IRAK-4) kinase activity in IRAK-4-mediated signaling. J Biol Chem. 2004, 279: 40653-40658. 10.1074/jbc.M402666200.CrossRefPubMed Lye E, Mirtsos C, Suzuki N, Suzuki S, Yeh WC: The role of interleukin 1 receptor-associated kinase-4 (IRAK-4) kinase activity in IRAK-4-mediated signaling. J Biol Chem. 2004, 279: 40653-40658. 10.1074/jbc.M402666200.CrossRefPubMed
68.
Zurück zum Zitat Xu Y, Tao X, Shen B, Horng T, Medzhitov R, Manley JL, Tong L: Structural basis for signal transduction by the Toll/interleukin-1 receptor domains. Nature. 2000, 408: 111-115. 10.1038/35040600.CrossRefPubMed Xu Y, Tao X, Shen B, Horng T, Medzhitov R, Manley JL, Tong L: Structural basis for signal transduction by the Toll/interleukin-1 receptor domains. Nature. 2000, 408: 111-115. 10.1038/35040600.CrossRefPubMed
69.
Zurück zum Zitat Liao W, Xiao Q, Tchikov V, Fujita K, Yang W, Wincovitch S, Garfield S, Conze D, El-Deiry WS, Schutze S, Srinivasula SM: CARP-2 is an endosome-associated ubiquitin ligase for RIP and regulates TNF-induced NF-kappaB activation. Curr Biol. 2008, 18: 641-649. 10.1016/j.cub.2008.04.017.CrossRefPubMedPubMedCentral Liao W, Xiao Q, Tchikov V, Fujita K, Yang W, Wincovitch S, Garfield S, Conze D, El-Deiry WS, Schutze S, Srinivasula SM: CARP-2 is an endosome-associated ubiquitin ligase for RIP and regulates TNF-induced NF-kappaB activation. Curr Biol. 2008, 18: 641-649. 10.1016/j.cub.2008.04.017.CrossRefPubMedPubMedCentral
70.
Zurück zum Zitat Clarke CJ, Truong TG, Hannun YA: Role for neutral sphingomyelinase-2 in tumor necrosis factor alpha-stimulated expression of vascular cell adhesion molecule-1 (VCAM) and intercellular adhesion molecule-1 (ICAM) in lung epithelial cells: p38 MAPK is an upstream regulator of nSMase2. J Biol Chem. 2007, 282: 1384-1396.CrossRefPubMed Clarke CJ, Truong TG, Hannun YA: Role for neutral sphingomyelinase-2 in tumor necrosis factor alpha-stimulated expression of vascular cell adhesion molecule-1 (VCAM) and intercellular adhesion molecule-1 (ICAM) in lung epithelial cells: p38 MAPK is an upstream regulator of nSMase2. J Biol Chem. 2007, 282: 1384-1396.CrossRefPubMed
71.
Zurück zum Zitat Rockel JS, Kudirka JC, Guzi AJ, Bernier SM: Regulation of Sox9 activity by crosstalk with nuclear factor-kappaB and retinoic acid receptors. Arthritis Res Ther. 2008, 10: R3-10.1186/ar2349.CrossRefPubMedPubMedCentral Rockel JS, Kudirka JC, Guzi AJ, Bernier SM: Regulation of Sox9 activity by crosstalk with nuclear factor-kappaB and retinoic acid receptors. Arthritis Res Ther. 2008, 10: R3-10.1186/ar2349.CrossRefPubMedPubMedCentral
72.
Zurück zum Zitat Liu TL, Shimada H, Ochiai T, Shiratori T, Lin SE, Kitagawa M, Harigaya K, Maki M, Oka M, Abe T, Takiguchi M, Hiwasa T: Enhancement of chemosensitivity toward peplomycin by calpastatin-stabilized NF-kappaB p65 in esophageal carcinoma cells: possible involvement of Fas/Fas-L synergism. Apoptosis. 2006, 11: 1025-1037. 10.1007/s10495-006-6353-y.CrossRefPubMed Liu TL, Shimada H, Ochiai T, Shiratori T, Lin SE, Kitagawa M, Harigaya K, Maki M, Oka M, Abe T, Takiguchi M, Hiwasa T: Enhancement of chemosensitivity toward peplomycin by calpastatin-stabilized NF-kappaB p65 in esophageal carcinoma cells: possible involvement of Fas/Fas-L synergism. Apoptosis. 2006, 11: 1025-1037. 10.1007/s10495-006-6353-y.CrossRefPubMed
73.
Zurück zum Zitat Willis TG, Jadayel DM, Du MQ, Peng H, Perry AR, bdul-Rauf M, Price H, Karran L, Majekodunmi O, Wlodarska I, Pan L, Crook T, Hamoudi R, Isaacson PG, Dyer MJ: Bcl10 is involved in t(1;14)(p22;q32) of MALT B cell lymphoma and mutated in multiple tumor types. Cell. 1999, 96: 35-45. 10.1016/S0092-8674(00)80957-5.CrossRefPubMed Willis TG, Jadayel DM, Du MQ, Peng H, Perry AR, bdul-Rauf M, Price H, Karran L, Majekodunmi O, Wlodarska I, Pan L, Crook T, Hamoudi R, Isaacson PG, Dyer MJ: Bcl10 is involved in t(1;14)(p22;q32) of MALT B cell lymphoma and mutated in multiple tumor types. Cell. 1999, 96: 35-45. 10.1016/S0092-8674(00)80957-5.CrossRefPubMed
74.
Zurück zum Zitat Peng C, Cho YY, Zhu F, Xu YM, Wen W, Ma WY, Bode AM, Dong Z: RSK2 mediates NF-{kappa}B activity through the phosphorylation of IkappaBalpha in the TNF-R1 pathway. FASEB J. 2010, 24: 3490-3499. 10.1096/fj.09-151290.CrossRefPubMedPubMedCentral Peng C, Cho YY, Zhu F, Xu YM, Wen W, Ma WY, Bode AM, Dong Z: RSK2 mediates NF-{kappa}B activity through the phosphorylation of IkappaBalpha in the TNF-R1 pathway. FASEB J. 2010, 24: 3490-3499. 10.1096/fj.09-151290.CrossRefPubMedPubMedCentral
75.
Zurück zum Zitat Weldon CB, Scandurro AB, Rolfe KW, Clayton JL, Elliott S, Butler NN, Melnik LI, Alam J, McLachlan JA, Jaffe BM, Beckman BS, Burow ME: Identification of mitogen-activated protein kinase kinase as a chemoresistant pathway in MCF-7 cells by using gene expression microarray. Surgery. 2002, 132: 293-301. 10.1067/msy.2002.125389.CrossRefPubMed Weldon CB, Scandurro AB, Rolfe KW, Clayton JL, Elliott S, Butler NN, Melnik LI, Alam J, McLachlan JA, Jaffe BM, Beckman BS, Burow ME: Identification of mitogen-activated protein kinase kinase as a chemoresistant pathway in MCF-7 cells by using gene expression microarray. Surgery. 2002, 132: 293-301. 10.1067/msy.2002.125389.CrossRefPubMed
76.
Zurück zum Zitat Kotlyarov A, Neininger A, Schubert C, Eckert R, Birchmeier C, Volk HD, Gaestel M: MAPKAP kinase 2 is essential for LPS-induced TNF-alpha biosynthesis. Nat Cell Biol. 1999, 1: 94-97. 10.1038/10061.CrossRefPubMed Kotlyarov A, Neininger A, Schubert C, Eckert R, Birchmeier C, Volk HD, Gaestel M: MAPKAP kinase 2 is essential for LPS-induced TNF-alpha biosynthesis. Nat Cell Biol. 1999, 1: 94-97. 10.1038/10061.CrossRefPubMed
77.
Zurück zum Zitat Walczak H, gli-Esposti MA, Johnson RS, Smolak PJ, Waugh JY, Boiani N, Timour MS, Gerhart MJ, Schooley KA, Smith CA, Goodwin RG, Rauch CT: TRAIL-R2: a novel apoptosis-mediating receptor for TRAIL. EMBO J. 1997, 16: 5386-5397. 10.1093/emboj/16.17.5386.CrossRefPubMedPubMedCentral Walczak H, gli-Esposti MA, Johnson RS, Smolak PJ, Waugh JY, Boiani N, Timour MS, Gerhart MJ, Schooley KA, Smith CA, Goodwin RG, Rauch CT: TRAIL-R2: a novel apoptosis-mediating receptor for TRAIL. EMBO J. 1997, 16: 5386-5397. 10.1093/emboj/16.17.5386.CrossRefPubMedPubMedCentral
78.
Zurück zum Zitat Wang L, Du F, Wang X: TNF-alpha induces two distinct caspase-8 activation pathways. Cell. 2008, 133: 693-703. 10.1016/j.cell.2008.03.036.CrossRefPubMed Wang L, Du F, Wang X: TNF-alpha induces two distinct caspase-8 activation pathways. Cell. 2008, 133: 693-703. 10.1016/j.cell.2008.03.036.CrossRefPubMed
79.
Zurück zum Zitat Ahmad M, Srinivasula SM, Wang L, Talanian RV, Litwack G, Fernandes-Alnemri T, Alnemri ES: CRADD, a novel human apoptotic adaptor molecule for caspase-2, and FasL/tumor necrosis factor receptor-interacting protein RIP. Cancer Res. 1997, 57: 615-619.PubMed Ahmad M, Srinivasula SM, Wang L, Talanian RV, Litwack G, Fernandes-Alnemri T, Alnemri ES: CRADD, a novel human apoptotic adaptor molecule for caspase-2, and FasL/tumor necrosis factor receptor-interacting protein RIP. Cancer Res. 1997, 57: 615-619.PubMed
80.
Zurück zum Zitat Tsitsikov EN, Laouini D, Dunn IF, Sannikova TY, Davidson L, Alt FW, Geha RS: TRAF1 is a negative regulator of TNF signaling. enhanced TNF signaling in TRAF1-deficient mice. Immunity. 2001, 15: 647-657. 10.1016/S1074-7613(01)00207-2.CrossRefPubMed Tsitsikov EN, Laouini D, Dunn IF, Sannikova TY, Davidson L, Alt FW, Geha RS: TRAF1 is a negative regulator of TNF signaling. enhanced TNF signaling in TRAF1-deficient mice. Immunity. 2001, 15: 647-657. 10.1016/S1074-7613(01)00207-2.CrossRefPubMed
81.
Zurück zum Zitat Rowlett RM, Chrestensen CA, Nyce M, Harp MG, Pelo JW, Cominelli F, Ernst PB, Pizarro TT, Sturgill TW, Worthington MT: MNK kinases regulate multiple TLR pathways and innate proinflammatory cytokines in macrophages. Am J Physiol Gastrointest Liver Physiol. 2008, 294: G452-G459.CrossRefPubMed Rowlett RM, Chrestensen CA, Nyce M, Harp MG, Pelo JW, Cominelli F, Ernst PB, Pizarro TT, Sturgill TW, Worthington MT: MNK kinases regulate multiple TLR pathways and innate proinflammatory cytokines in macrophages. Am J Physiol Gastrointest Liver Physiol. 2008, 294: G452-G459.CrossRefPubMed
Metadaten
Titel
Alterations in tumor necrosis factor signaling pathways are associated with cytotoxicity and resistance to taxanes: a study in isogenic resistant tumor cells
verfasst von
Jason A Sprowl
Kerry Reed
Stephen R Armstrong
Carita Lanner
Baoqing Guo
Irina Kalatskaya
Lincoln Stein
Stacey L Hembruff
Adam Tam
Amadeo M Parissenti
Publikationsdatum
01.02.2012
Verlag
BioMed Central
Erschienen in
Breast Cancer Research / Ausgabe 1/2012
Elektronische ISSN: 1465-542X
DOI
https://doi.org/10.1186/bcr3083

Weitere Artikel der Ausgabe 1/2012

Breast Cancer Research 1/2012 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.