Skip to main content
Lung Cancer: Targets and Therapy logoLink to Lung Cancer: Targets and Therapy
. 2010 Sep 4;1:107–117. doi: 10.2147/LCTT.S12874

Genome-wide gene expression analysis of chemoresistant pulmonary carcinoid cells

Ulrike Olszewski 1, Robert Zeillinger 1, Klaus Geissler 1, Gerhard Hamilton 1,
PMCID: PMC5312470  PMID: 28210111

Abstract

Purpose

Carcinoids are highly chemoresistant tumors associated with a dismal prognosis. This study involved a comparison of the genome-wide gene expression pattern of a chemoresistant and a chemosensitive pulmonary carcinoid cell line to reveal factors that contribute to the resistant phenotype.

Materials and methods

Gene expression of UMC-11 chemoresistant carcinoid cells as assessed by 32 K microarray was compared with H835 chemosensitive carcinoid cells, and the genes that were differentially expressed and expected to be related to chemoresistance were selected.

Results

Drug-resistant UMC-11 cells exhibited increased expression of transcripts known to confer resistance to different cytostatics such as P-glycoprotein, multidrug resistance-associated proteins 2 and 3, effectors of the glutathione detoxification and xenobiotics degradation pathways, and ion transporters including Na+/K+-adenosine triphosphatase. In addition, enhanced transcription of several S100 proteins, capable of suppressing apoptosis, regulation of topoisomerase I (topo I) expression by antisense transcripts from TOPO1 pseudogenes, and alterations of the cytoskeleton seem to contribute to the multidrug-resistant phenotype. A multitude of epidermal growth factor (EGF)-related and neuropeptide growth factors, overexpression of proteases, and appearance of aerobic glycolytic metabolism complement the malignant phenotype of the UMC-11 cells.

Conclusion

The multidrug-resistant phenotype of the UMC-11 pulmonary carcinoid cell line seems to be mediated by classical efflux pumps, drug metabolization or conjugation systems, and, possibly, modulation of apoptotic cell death by S100 proteins and topo I expression by pseudogene transcripts. Autocrine or paracrine stimulation by a host of EGF-related and neuropeptide growth factors, as well as high metastatic potency indicated by increased expression of components of aerobic glycolysis and proteolytic enzymes, may furthermore account for the failure of therapeutic interventions.

Keywords: neuroendocrine tumor, drug resistance, microarray, drug transporter, apoptosis

Introduction

Pulmonary carcinoid tumors are considered low-grade malignant neoplasms of the neuroendocrine cells, originating from the Kulchitsky cells of the bronchial mucosa layer, and represent about 1%–5% of all lung tumors.13 Ninety percent of these tumors termed typical carcinoids are well differentiated and characterized by a small degree of mitosis, pleomorphism, and necrosis.4,5 The remaining 10% of the tumors with increased mitotic activity, nuclear pleomorphism, and cellular irregularity are classified as aggressive atypical carcinoids.6,7 The patients with lung carcinoids can be cured by surgical resection, but a fraction of tumors gives rise to widespread metastasis within several years after primary treatment.8,9 In contrast to the more indolent, well-differentiated neuroendocrine tumors exhibiting metastases in less than 15% of cases and revealing a 5-year survival rate of more than 90%, well-differentiated neuroendocrine carcinomas are more aggressive, develop metastases in 30%–50% of cases, and yield a 5-year survival rate between 40% and 60%.1,2,10 Almost all recurrences of either typical or atypical carcinoids involve distant sites. Finally, patients with malignant carcinoids show a 5-year survival rate of about 20% and a median survival of 2 years in the presence of liver metastases.11 Medical treatment of metastatic disease includes somatostatin (SST) analogs, α-interferons, and chemotherapy.1,12

Basically, chemotherapy can be considered for patients with carcinoids progredient under noncytotoxic therapy, provided that they have significant symptoms and poor prognosis.13,14 Clinical trials demonstrated that the response rates to single-agent chemotherapy limited to approximately 20%, and for multiagent chemotherapy, the response rates are invariably less than 40%. Moreover, these responses are frequently short-lived and rarely translate to prolonged survival. In contrast, responses of rapidly proliferating, poorly differentiated (atypical or small cell-like) carcinoid tumors to chemotherapy are high but the duration of response is extremely short. In these studies, doxorubicin, 5-fluorouracil (5-FU), dacarbazine, streptozotocin, cyclophosphamide, cisplatin, and etoposide were used either as single agents or in combinations, but eventually were not recommended for clinical routine practice due to low response rates.1,3,11,15 The situation is complicated by the fact that these clinical trials are usually small studies with variable criteria for inclusion and study design due to the rarity of these tumors. Little is known about the mechanisms contributing to chemoresistance of carcinoids or other well-differentiated neuroendocrine tumors. Screening of chemosensitivies of UMC-11, H727, and H835 pulmonary carcinoid cell lines in the laboratory revealed the resistance of UMC-11 to 9 of 14 chemotherapeutics, whereas H727 and H835 were resistant to 4 of 14 and 5 of 14 of the drugs, respectively.16 Because doubling times were 20.8 hours for H727, 27.0 hours for UMC-11, and 35.7 hours for H835 cells, the more rapidly proliferating and, therefore, probably more chemosensitive H727 cell line was not used in the following gene expression analysis.16 The aim of this study was to compare the transcriptome of the highly chemoresistant UMC-11 with the sensitive H835 cell line to delineate candidate genes possibly conferring multidrug resistance to pulmonary carcinoids.

Materials and methods

Chemicals and cell lines

Unless otherwise noted, all chemicals and solutions were obtained from Sigma-Aldrich (St. Louis, MO). UMC-11 and H835 pulmonary carcinoid cell lines were purchased from the American Tissue Culture Collection (Manassas, VA). Cells were cultured in RPMI-1640 medium supplemented with 10% fetal bovine serum (Seromed, Berlin, Germany), 4 mM glutamine, and antibiotics. UMC-11 was subcultured by trypsinization (2.5% trypsin in EDTA solution; Boehringer Mannheim, Germany), and H835 cells growing in suspension were maintained by replacement of the medium and dispersed by trypsin treatment.

Cell proliferation assay

Cells were harvested, counted, and distributed into the wells of flat-bottomed 96-well microtiter plates at a density of 1 × 104 cells/well in 100 µL medium. 100 µL of appropriate dilutions of test compounds was added to each well, and the plates were incubated under tissue culture conditions for 4 days. Stock solutions of the compounds were prepared in either 70% ethanol or dimethyl sulfoxide and diluted more than 100-fold for use in chemosensitivity assays. Solvent-control wells were included in all tests. Dose–response curves were obtained by the assessment of cell growth at 2-fold drug dilutions in triplicate and used for the calculation of the half maximal inhibitory concentration (IC50) values. Cell proliferation was quantified using a modified tetrazolium dye (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay (EZ4U; Biomedica, Vienna, Austria).

Genome-wide gene expression analysis

Lysates of 30 × 106 cells (extraction buffer: 4 M guanidine isothiocyanate, 0.5% sodium N-lauroyl sarcosinate, 10 mM EDTA, 5 mM sodium citrate, and 100 µM β-mercaptoethanol; 30 minutes, 4°C) were added to cesium trifluoroacetate and centrifuged (46,000 rpm, 15°C, 20 h). Supernatant containing DNA was removed and RNA was precipitated with ice-cold 96% ethanol. Pellets were washed and, following removal of ethanol, resuspended in sterile water. RNA content was measured photometrically.

Gene expression analysis was performed using the Applied Biosystems (ABI) Human Genome Survey Microarray V2.0 (Applied Biosystems, Foster City, CA) according to the manufacturer’s instructions. Therefore, 2–5 µg mRNA (20–50 µg total RNA) was reversely transcribed (RT) to first-strand cDNA (MyCycler thermocycler; Bio-Rad, Hercules, CA). The RT mixture was labeled on ice and purified according to the manufacturer’s instructions for the Applied Biosystems 1700 RT Labeling Kit v002 (Applied Biosystems). Hybridization of cDNA and microarray analysis (Applied Biosystems 1700) were carried out following the manufacturer’s chemiluminescence detection kit protocol. Data for each cell line (n = 2) were filtered, normalized, and log2-transformed before further processing was done using Microsoft Excel software (Microsoft, Redmond, WA) or Statistical Analysis of Microarray (false discovery rate of 10%; Stanford University, Stanford, CA). ABI 1700 gene identities can be accessed via the Panther classification system (www.pantherdb.org).

Results

We previously reported the following IC50 values (IC50 UMC-11/IC50 H835, mean ± standard deviation) for a range of drugs that were obtained for UMC-11 and H835 cells in chemosensitivity assays: vinblastine (ng/mL), 2 ± 0.35/2 ± 0.3; taxol (ng/mL), 10 ± 4.4/9 ± 3.9; camptothecin (nM), 18 ± 5.0/6.5 ± 4.3; 5-FU (µM), 30 ± 4.6/45 ± 8.7; doxorubicin (nM), 370 ± 72/73 ± 2.4; gemcitabine (nM), 833 ± 103/30.0 ± 20; tamoxifen (µM), 9 ± 3.3/7 ± 2.4; cisplatin (µM), 33 ± 15/33 ± 7.0; oxaliplatin (µM), 5 ± 0.25/6 ± 0.51; carboplatin (µM), 36 ± 7.1/36 ± 9.9; mitomycin (ng/mL), 156 ± 15/35 ± 9.0; streptozotocin (µg/mL), 180 ± 33/60 ± 19; etoposide (µg/mL), 45 ± 12.3/0.25 ± 0.02; and dacarbazine (µg/mL), 4 ± 2.3/16 ± 3.9. Thus, UMC-11 cells were resistant to taxol, camptothecin, 5-FU, doxorubicin, gemcitabine, tamoxifen, cisplatin, streptozotocin, and etoposide (IC50 exceeding peak plasma concentration).16

Genome-wide expression analysis was performed for chemoresistant UMC-11 and chemosensitive H835 pulmonary carcinoid cells, and the genes that were significantly overexpressed more than 4-fold in the former cell line compared with the latter were grouped according to their cellular functions and pathways (Table 1). Genes were furthermore selected in regard to their possible relation with the resistant phenotype of the UMC-11 cells. The low number of genes with lower expression in UMC-11 compared with H835 cells was not included in this analysis because of the absence of transcripts with a connection to chemoresistance. After filtering of the 1,520 transcripts that were overexpressed more than 4-fold in UMC-11 compared with H835 carcinoid cells, 386 were annotated to the corresponding genes, and 102 of these are expected to be involved in proliferation, metastasis, and drug resistance of the carcinoid cells, as listed in Table 1.

Table 1.

Relative mRNA expression of selected genes in UMC-11 and H835 carcinoid cell lines

Gene symbol Gene ID ABI 1700 Ratio of expression UMC-11/H835
Metabolism
LDHB 167986 280
LDH 208956 55
GAPDH 236913 9
HK1 178010 8
CS 200851 6
PDK4 101060 12
Hypoxia
HIG1 127432 12
HIGD1A 139216 14
ARNT2 205730 4
Proteases/protease inhibitors
PRSS1 111365 2,636
PRSS2 109205 5,576
PRSS3 154524 1,861
Serpin A2 192373 11
Serpin E2 210342 13
Serpin B6 113902 5
Serpin A10 101912 18
CTSL 172735 32
MMP13 162909 20
TIMP1 134692 1,208
TIMP3 179538 57
Membrane transporters
ATP1B1 199586 7
ATP2A2 142525 4
ATP2B1 102967 6
ATP2C2 146652 51
FXYD2 146341 84
FXYD3 126166 23
ABCB1 182279 44
ABCC2 115883 26
ABCC3 109767 41
ABCD1 165343 5
ABCG1 210662 4
SLCO1A2 192702 4
SLCO2A1 178284 33
SLC1A6 112712 916
SLC18A1 127370 27
SLC4A2 164313 4
SLC4A11 124838 16
SLC12A7 187225 6
SLC20A2 132288 5
SLC35C1 207403 8
Growth factors
EGFR 136952 64
ERBB3 157084 22
TGFA 180395 17
TGFB1 170749 21
TGFB3 142790 8
EREG 100064 201
AREG 123143 147
Hsp90 148821 4
SPRY2 207231 33
IGFII 129922 814
IGFBP1 202509 42
IGFBP3 104923 20
IGFBP5 170211 6
IGFBP6 150353 23
IGF2BP2 147646 24
IL-8 176899 115
CALCB 180834 6,969
VIP 144138 552
NTS 160562 93
VGF 174736 36
NPW 164302 6
AGT 196523 5
SST 157922 887
FGF12 138024 13
ADCY2 193946 618
STIM2 205035 4
S100 Ca2+-binding proteins
A2 192373 11
A4 155850 170
A6 154326 87
A10 213843 185
A11P 187317 178
P 165541 254
A14 185644 36
A16 150790 432
Cytoskeleton
Keratin I 231137 458
Keratin II 178654 103
KRT7 207298 38
KRT8 111916 14
KRT18 114812 171
KRT19 113888 80
CDH1 130505 300
VIL1 191456 264
CLDN4 156519 21
CLDN7 163619 34
CLDN20 159987 74
TUBB3 203100 10
TMSB4X 141609 4
Sulfur metabolism
GSTT1 163516 4
GSTP1 214807 225
GRX 188218 57
ARSE/D 139887 52
SULF2 116303 43
GSTO2 177000 41
GLRX 188218 57
TST 203712 8
Detoxification
AHR 166278 39
CYP26B1 182081 28
CYP2B6 209472 19
ALDH7 A1 219563 30
AKR7 A3 179819 7
Topoisomerase I
TOP1 179728 7
TOPP1 115448 5
TOPP2 236556 5

In UMC-11 cells, enzymes mainly involved in glucose and energy metabolism that were preferentially expressed included lactate dehydrogenase (LDH/LDHB), glyceraldehyde 3-phosphate dehydrogenase (GAPDH), hexokinase 1 (HK1), citrate synthetase (CS), and pyruvate dehydrogenase kinase 4 (PDK4), indicating differences in glucose uptake and utilization between the 2 cell lines. Increased expression of hypoxia-inducible gene domain family member 1A (HIG1/HIGD1A) and aryl hydrocarbon receptor nuclear translocator 2 (ARNT2) together with the changes in regard to glucose metabolization pointed to the presence of an increased rate of aerobic glycolysis (Warburg effect) in UMC-11 cells. Furthermore, in these cells, a number of proteases and protease inhibitors were highly expressed, including serine proteases 1–3 (PRSS1-3), serine protease inhibitors (SERPINs A2, E2, B6, and A10), cathepsin L (CTSL), matrix metallopeptidase 13 (MMP13, identical to collagenase 3), and the corresponding tissue inhibitors of metalloproteinase 1 and 3 (TIMP1 and 3). Hypoxia is known to select for an aggressive cancer phenotype characterized by an increased capacity to infiltrate tissues.

In addition, UMC-11 cells showed higher expression of a large number of membrane transporters compared with H835 cells, thus comprising Na+/K+-adenosine triphosphatase ([ATPase], ATP1B1), the FXYD regulatory subunit 2 (FXYD2), and ion pumps regulating intracellular Ca2+ concentration (ATP2A2, ATP2B1, and ATP2C2). The subfamily members of ATP-binding cassette (ABC) transporters (ABCB1, C2, and C3) resemble the classical resistance proteins P-glycoprotein (P-gp) and multidrug resistance-associated proteins (MRPs) 2 and 3. Furthermore, the 2 solute carrier organic anion transporters (SLCO1A2 and SLCO2A1), shuttling bile acids, prostaglandins (PGs), and other anions, and a number of solute carriers transporting aspartate/glutamate (SLC1A6), biogenic amines (SLC18A1), anions (SLC4A2), sodium (SLC4A11), potassium chloride (SLC12A7), sodium phosphate (SLC20A2), and guanosine diphosphate-fucose (SLC35C1) were found to be upregulated.

Moreover, a large number of growth factors and receptors were overexpressed in UMC-11 compared with H835 cells, including members of the epidermal growth factor (EGF) family, insulin-like growth factor (IGF) family, neuropeptides, and others. In particular, EGF receptor (EGFR), v-erb-b2 erythroblastic leukemia viral oncogene homolog 3 (ERBB3), transforming growth factors (TGFα, β1, and β2), epiregulin (EREG), and amphiregulin (AREG) constitute a growth regulatory system, with heat shock protein 90 (Hsp90) and sprouty 2 (SPRY2) as modulators. A second independent growth factor pathway was provided by IGF2 and regulatory binding proteins (IGFBP1, 3, 5, 6, and IGF2BP2), supplemented by a number of neuropeptides, namely vasointestinal peptide (VIP), neurotensin (NTS), VGF (VGF nerve growth factor inducible), and neuropeptide W (NPW). Interleukin-8 (IL-8), calcitonin-related polypeptide β (CALCB), angiotensin (AGT), and fibroblast growth factor 12 (FGF12) may in addition be involved in the regulation of cell proliferation, with SST as inhibitor. Increased overexpression of adenylate cyclase (ADCY2) pointed to an important role of this enzyme in signal transduction pathways of the growth stimulators.

S100 proteins regulate intracellular processes, such as cell growth and motility, cell cycle regulation, transcription, and differentiation. Twenty members have been identified so far, and altogether, S100 proteins represent the largest subgroup of the EF-hand Ca2+-binding protein family. A unique feature of these proteins is that individual members are localized in specific cellular compartments from where some are able to relocate upon Ca2+ activation, thus transducing the Ca2+ signal temporally and spacially by interacting with different targets highly specific for each S100 protein. Some of the S100 Ca2+-binding proteins overexpressed in the UMC-11 cells, namely A2, A4, A6, A10, P, A14, and A16, seem to have important functions in Ca2+ homeostasis, growth, and apoptosis. Differences in the expression of cytoskeleton constituents comprised type I keratins (KRT18 and KRT19), type II keratins (KRT7 and KRT8), cadherin 1 (CDH1), villin 1 (VIL1), claudins (CLDN4, 7, and 20), tubulin β3 (TUBB3), thymosin β4, X-linked chromosome (TMSB4X), and stromal interaction molecule 2 (STIM2).

Finally, detoxification of xenobiotics may be mediated by conjugation to sulphur-containing compounds, and the corresponding enzymes comprising glutathione S-transferase θ1 (GSTT1), glutathione S-transferase π1 (GSTP1), glutaredoxin (GRX), arylsulfatase (ARSE/D/SULF2), glutathione S-transferase Ω2 (GSTO2), and thiosulfatase (TST) were found to be overexpressed in UMC-11 cells. Other mechanisms for the metabolization of drugs in UMC-11 cells included the cytochrome P450 (CYP) oxidoreductases (CYP26B and CYP12B6) that are regulated by the consistently overexpressed aryl hydrocarbon receptor (AHR), aldehyde dehydrogenase 7A1 (ALDH7A1), and aldo-keto reductase 7A3 (AKR7A3).

Topoisomerases are important targets of a range of cytotoxic drugs, in particular (topoI) for camptothecin derivatives like irinotecan and topotecan. Although TOP 1 expression was elevated in UMC-11 cells, increased amounts of antisense transcripts stemming from TOPP1 and TOPP2 are expected to downregulate TOP1 to induce drug resistance.

Discussion

Neuroendocrine lung tumors are highly refractory to chemotherapy, but the detailed mechanisms resulting in chemoresistance have not been fully characterized so far.11 In contrast to the typical carcinoids, where multidrug resistance (MDR) is mediated by P-gp but not MRP, the atypical carcinoids exhibit cellular detoxification by P-gp, MRP, and GST-π expression.17 Both Bcl-2 and p53 proteins appear unchanged in carcinoids, suggesting that the apoptotic capacity conferred by these two gene products is not involved in chemoresistance. The chemoresistance of typical carcinoids was questioned, in general, based on their sensitivities observed in in vitro tests using these well-differentiated tumors.13 We recently compared the chemosensitivity profiles of the three carcinoid cell lines UMC-11, H727, and H835 and demonstrated marked chemoresistance to a wide range of cytotoxic drugs in UMC-11 cells in contrast to the other two chemosensitive cell lines.16 In this study, genome-wide gene expression analysis was used to search for candidate genes overexpressed in UMC-11 cells compared with H835 cells, and that would possibly hold the potential to confer chemoresistance.

Expression of hexokinase (HK1), an enzyme that traps glucose inside the cell by catalyzing phosphorylation to glucose 6-phosphate, and thus, maintains the downhill concentration gradient permitting facilitated glucose transport into cells for utilization, was higher in UMC-11 than in H835 cells.18 GAPDH catalyzes the conversion of glyceraldehyde 3-phosphate to D-glycerate 1,3-bisphosphate during the sixth step of glycolysis. LDH/LDHB catalyzes the conversion of L-lactate and nicotinamide adenine dinucleotide (NAD) to pyruvate and NADH in the final step of anaerobic glycolysis. This enzyme normally converts pyruvate, the final product of glycolysis, to lactate under hypoxic conditions. PDK4 is a member of the PDK/BCKDK protein kinase family and encodes a mitochondrial protein with a histidine kinase domain.19 This protein inhibits the PD complex (PDC) by phosphorylating one subunit, which contributes to the regulation of glucose metabolism. The PDC converts pyruvate to acetyl coenzyme A (acetyl-CoA) by decarboxylation. Acetyl-CoA may then enter into the citric acid cycle for cellular respiration, thereby the glycolytic metabolic pathway is linked to the citric acid cycle and energy released in the form of NADH PD. In summary, high expression of these enzymes may indicate a shift of the cellular metabolism to increased uptake and catabolization of glucose.

Tumor cells exhibit aerobic glycolysis frequently (Warburg effect), and according to our results, the increased expression of a number of relevant genes corroborate this metabolic shift. Unlike in H835 cells, HIGD1A is overexpressed in UMC-11 cells. HIG1 was found as a novel gene, with unknown function induced by hypoxia and glucose deprivation in human cervical epithelial cells in vitro.20 Hypoxia-induced transcription of the HIF-target genes HIGD1A, egl nine homolog 1 (EGLN1), Bcl2-interacting-protein 3 (BNIP3), and phosphofructokinase 1 is independent of the CH1 domain of CBP/p300, whereas stanniocalcin 1 and SLC2A1 (glucose transporter 1) were moderately affected by the alteration of this transcriptional coactivator.21 Similarly, EGLN1, HIG1, and prolyl 4-hydroxylase are induced by treatment with nickel.22 In good agreement with HIGD1A expression, abundant transcripts of ARNT2, a member of the basic-helix-loop-helix-Per-Arnt-Sim (bHLH-PAS) superfamily of transcription factors that was observed in UMC-11 cells, heterodimerize with HIF-1 in the nucleus and, thus, promote expression of oxygen-responsive genes. In contrast to HK1, GAPDH is known to be induced by hypoxia or nickel.23,24 Therefore, UMC-11 cells clearly showed signs of aerobic glycolysis and induction of hypoxia-induced genes under aerobic conditions.25

Most cancers exhibit elevated protease levels that contribute to certain aspects of tumor behavior in regard to growth, metastasis, and angiogenesis. Increased expression of the CTSs of the cysteine protease family, such as CTSL, correlates with invasion and migration of highly metastatic B16 melanoma cells.26 Trypsin 1 and 2, serine protease 3 (PRSS1, 2, and 3), and MMP13 were preferentially expressed in UMC-11 but not in H835 cells. Trypsin is implicated in colorectal carcinogenesis and promotes growth and dissemination of various cancers.27 Furthermore, trypsin expression is an indicator of poor prognosis and shorter disease-free survival of colorectal cancer patients. Trypsin seems to act either directly by the digestion of type I collagen or indirectly by the activation of other proteinase cascades like MMPs. Stimulated by trypsin, both MMPs and protease-activated receptor 2 may activate the mitogen-activated protein kinase–extracellular signal-regulated protein kinase (MAPK-ERK) pathway through EGFR. MMPs may play an important role in both transformation from adenoma to carcinoma and initiation of invasion and metastasis. Upregulation of MMP13 at the tumor-bone interface is crucial for tumor-induced osteolysis, which suggests that MMP13 is a potential therapeutic target for breast cancer bone metastasis.28 Tumor cell proteases are under tight regulation by proteinase inhibitors like TIMP2 and 3, both acting on MMP13.29 Moreover, the highly expressed serpins (serine protease inhibitors) A1, A10, E2, and F1 are proteins with similar structures that were first identified as inhibitors.30 SERPINA10 was one of the six novel marker genes for neuroendocrine carcinoma cells comprising paraneoplastic antigen MA2 and testican 1 precursor (SPOCK1) among others.31 Overexpression of these proteases in conjunction with their regulatory inhibitors in UMC-11 cells pointed to a highly metastatic and aggressive phenotype.

The solute carrier, human organic anion transporting polypeptide SLCO1A2 is highly expressed in the intestine, kidney, cholangiocytes, and the blood–brain barrier.32 This localization suggests that the transporter may be crucial for the distribution of clinically important drugs, besides shuttling bile acids. SLCO2A1 transports PGs like PGE2 that was identified as the principal prostanoid promoting cell growth and survival in colorectal tumors by increasing Bcl-2 expression, which promotes cell survival under hypoxic conditions.33 The other solute carriers overexpressed in UMC-11 cells are involved in the maintainance of ion homeostasis (Na+, K+, H+, and others), with no clear relation to chemoresistance.

MDR of tumor cells against a whole group of cytotoxic compounds is mediated by ABC efflux pumps.34 P-gp (ABCB1), encoded by the MDR1 gene, was the first such transporter detected. Determination of MDR1 mRNA in human cancers revealed elevated expression in untreated, intrinsically drug-resistant tumors, including those derived from carcinoid tumors.35 Further studies resulted in the characterization of the MRPs in some drug-selected cancer cell lines.36 UMC-11 cells expressed ABCC2 (MRP2) and ABCC3 (MRP3) at higher levels than the drug-sensitive H835 cell line. MRPs appear to mediate glutathione (GSH) homeostasis and the efflux of oxidized GSH derivatives (eg, glutathione disulfide, S-nitrosoglutathione, and glutathione-metal complexes) and other glutathione S-conjugates.37 Among the many processes, apoptosis, cell proliferation, and differentiation are influenced by GSH transporters. MRP gene expression was detected in almost all lung cancer cell lines, including carcinoids and normal lung tissue.36 Furthermore, ABCG1 and ABCD1 are involved in the transport of lipids. In conclusion, MDR in UMC-11 cells seems to be partially effectuated by the classical ABC drug efflux pumps.

UMC-11 cells exhibited higher expression of ATP1B1 encoding the Na+/K+-ATPase that exchanges three intracellular sodium for two extracellular potassium ions and is upregulated by cyclic adenosine monophosphate (cAMP). It serves as a signal transducer, regulates cell adhesion, and its aberrant expression and activity are implicated in the development and progression of different cancers.38 Inhibition of this pump by cardiac glycosides reduces tumor metastasis, which is possibly due to the sensitization to anoikis.39 Altered ion gradients were also proposed as a potential cause of resistance to chemotherapy.40 Furthermore, inhibition of Na+/K+-ATPase markedly reduces intracellular cisplatin accumulation.41 A hemisynthetic cardenolide shows high anticancer activity in cells expressing diverse forms of MDR, which is either conferred by inherent overexpression of selected drug transporter proteins or induced by a range of chemotherapeutic agents.42 Ion transport by Na+/K+-ATPase is modulated by a regulatory subunit belonging to the FXYD protein family.43 UMC-11 cells showed high levels of FXYD2 and FXYD3 constituting the Na+/K+-ATPase γ-subunit and an ion pump regulator, highly expressed in breast cancers and responsible for cancer cell proliferation, respectively.44 The other ion pumps overexpressed in UMC-11 cells namely ATP2A2, B1, and C2 are responsible for the maintainance of cytoplasmic Ca2+ homeostasis.

Growth factors and their cognate receptors do not confer chemoresistance, but since they determine the proliferation rate, they constitute interesting targets for chemotherapy. The EGF/EGFR and ERBB3 growth factor system, as found overexpressed in UMC-11 cells, comprises a range of receptors and mitogenic factors including TGFα and others. The coexpressed factors TGFβ1 and β2 are involved in the regulation of many physiological processes like cell growth, cell differentiation, and extracellular matrix production among others.45 Moreover, TGFβ may function as tumor suppressor or promotor depending on the type, differentiation state, and physiological characteristics of target cells.46 The ErbB ligand epiregulin, found in UMC-11 cells, is highly expressed in non-small cell lung carcinoma (NSCLC) and correlates with nodal metastasis and a shorter duration of survival.47 The related and coexpressed EGFR/TGFR ligand, amphiregulin, was found to predict NSCLC resistance to gefitinib and to suppress apoptosis by sequestration of BAX in the cytoplasm.48 EGFR and activated EGFR (p-EGFR) were expressed in both well-differentiated gastrointestinal carcinoids and pancreatic endocrine tumors in primary and metastatic specimens with a poor prognosis.49 The role of EGFR was studied in pulmonary typical and atypical carcinoid tumors.50 Analysis showed that approximately half of typical carcinoids and one-third of atypical carcinoids produce EGFR, and all of the tumors exhibit moderate to intense staining for ErbB3 and ErbB4 but lack expression of ErbB2. High expression of the chaperone Hsp90 stabilizes oncogenic kinases and mutant EGFR driving proliferation of lung cancer cells.51 It was recently demonstrated that mutant EGFR is a Hsp90 target. In addition, SPRY1 seems to be operative in the regulation of EGFR signaling.52

Another growth factor system frequently overexpressed in cancers is represented by the IGFs. IGF2 and a number of associated regulatory IGF-binding proteins (IGFBP1, 3, 5, 6, and IGF2BP1/2) were elevated in UMC-11 cells. Expression of IGFBP2 along with low amounts of IGFBP1 was found in all carcinoid samples in association with varying occurrences of IGF1R, IGF2R, and IGFBP6.53,54 CALCB was reported to play an important role in the proliferation of various types of epithelial and endothelial cells.55 In an in vitro study, calcitonin-gene related peptide increased the proliferation of A549 alveolar epithelial cells in a dose-dependent and time-dependent manner.56 IL-8 is a chemokine and angiogenic factor that also functions as an autocrine growth factor in several human cancers. In lung cancer, all NSCLC cell lines produced IL-8, in contrast to low levels of IL-8 in SCLC cell lines despite expression of the IL-8 receptors CXCR1 and CXCR2.57 FGF12 is a FGF-homologous factor, which interacts with the intracellular kinase scaffold protein islet brain-2 and voltage-gated sodium channels.58 UMC-11 cells produce AGT that may influence tumor growth and metastasis in a tissue-specific and tumor-specific manner.59 In contrast, SST released by UMC-11 cells counteracts signaling of many other growth factors that contribute to proliferation, such as prolactin, IGF, TGFα and β, platelet-derived growth factor, EGF, and vascular endothelial growth factor (VEGF).

Because carcinoids resemble neuroendocrine tumors, the high expression of neuropeptides comprising VIP, NTS, VGF, and NPW by UMC-11 cells is expected to contribute significantly to autocrine growth stimulation. VIP causes increased proliferation of human breast and lung cancer cells in vitro.60 It binds to cancer cells with high affinity and increases cAMP and gene expression of c-fos, c-jun, c-myc, and VEGF. SCLC, which is a neuroendocrine cancer, produces and secretes gastrin releasing peptide (GRP), NTS, and adrenomedullin (AM) as autocrine growth factors.61 Most of these neuropeptides are detectable in serum samples of carcinoid patients.62 GRP, NTS, and AM bind to G-protein-coupled receptors triggering increased phosphatidylinositol (PI) turnover or elevation of cAMP in SCLC cells. Addition of GRP, NTS, or AM to SCLC cells causes altered expression of nuclear oncogenes like c-fos and stimulation of growth.63 Analysis of peptides secreted by the large-cell neuroendocrine carcinoma cell line revealed 2 fragments that were demonstrated to be parts of VGF, which is usually expressed in nerve cells or neuroendocrine cells.64 Reverse transcription–polymerase chain reaction of lung cancer cell lines showed that VGF mRNA was expressed only in neuroendocrine carcinoma-derived cells. Neuropeptide B (NPB) and NPW are the endogenous ligands of the 2 Gprotein-coupled receptors GPR7 and GPR8 and activate protein kinase A and protein kinase C (PKC) signaling.65 NPB and NPW were reported to stimulate tyrosine kinase and MAPK activities, mediating the proliferative and antiapoptotic effects of NPB and NPW.

Cell type–specific differences exist among the various signal transduction pathways, comprising cAMP, hydrolysis of PI mobilization of intracellular calcium, and tyrosine phosphorylation, and, moreover, different receptors for the same trigger may be linked to different signal transduction pathways.66,67 Basically, the mitogenic signal of neuropeptides is transmitted into the cell via specific receptors that couple to heterotrimeric G proteins. Subsequent activation of phospholipase C-β influences the activation of PKC and the elevation of intracellular calcium. The antiproliferative effect of the Ca2+-channel blocker verapamil was investigated in vitro on 3 human lung cancer cell lines.68 Verapamil inhibited cell proliferation in the neuroendocrine cancer cell line H727 in the nM range. STIM2 overexpressed in UMC-11 cells functions as highly sensitive Ca2+ sensor in the endoplasmic reticulum and regulates the ion concentration in conjunction with different pumps. Compared with H835, UMC-11 cells showed high expression of adenylate cyclase, establishing cAMP as an important second messenger of growth signals in these resistant carcinoid cells. In conclusion, UMC-11 cells seem to rely on EGF-related, IGF-related, and neuropeptide growth factors for stimulation of proliferation, and this multitude of autocrine and paracrine loops is expected to impede antiproliferative therapy by kinase inhibitors.

Overexpression of a large number of S100 Ca2+-binding proteins in UMC-11 cells pointed to their important functions in cell biology and possibly drug resistance of carcinoids. Intracellular Ca2+ seems to be permanently elevated in UMC-11 cells in response to continuous stimulation by growth factors. Downregulation of proapoptotic genes such as serine/threonine kinase 17a and BNIP3 in association with S100P was reported to be linked to oxaliplatin resistance in the THC8307/L-OHP colon cancer cell line, and 8.7-fold overexpression of S100 protein family members was demonstrated in proteome analysis of cisplatin-resistant ovarian cancer cells.69,70 Furthermore, pancreatic ductal adenocarcinoma showed a correlation between decreased expression of BNIP3 and chemoresistance through repression by S100A2 and S100A4.71 In support of these findings, S100A4 expression was a predictor of poor prognosis for T1N0M0 breast cancer patients.72,73 In conclusion, the expression of a number of S100 proteins may contribute to the broad chemoresistance of carcinoid cells by downregulation of proapoptotic factors.

Numerous members of the cytoskeleton are preferentially expressed in UMC-11 vs H835 cells, including type I and II cytokeratins, CLDNs, VIL, CDH1, TUBB3, and thymosin β4. Most of these components may reflect differences in the histological subtypes of UMC-11 and H835 cells; however, especially TUBB3 overexpression represents a marker of resistance to microtubule-targeting drugs in vitro, in vivo, and clinically for many tumors, including breast cancer, and thymosin β4 proves to be a gene associated with response to therapy in ovarian cancer.74,75

Sulfur-containing molecules such as cysteine, methionine, GSH, metallothioneins, and albumin bind platinum-based compounds and reduce therapeutic efficacy at the level of uptake, excretion, resistance, and toxicity.76 Studies showed that variability in survival can in part be explained by polymorphisms in genes encoding proteins involved in degradation of drugs. Most importantly, such polymorphisms of drug-metabolizing enzymes and transporters demonstrated to influence survival after cancer treatment compass genes of the phase II detoxification enzymes, GSTs.77 For example, the GSTM1-deficient and GSTT1-deficient genotypes have a clear association with longer overall survival in patients treated with respective substrates, such as alkylating agents and platinum compounds. Likewise, genetic polymorphisms of GSTP1 and GSTA1 are also linked with increased overall survival in patients with different malignancies. Levels of thioredoxin (TRX) and GRX were elevated in pancreatic ductal carcinoma tissues compared with pancreatic cystadenocarcinoma or normal pancreatic tissue. Furthermore, cisplatin-resistant subclones of HeLa cells had higher expression of TRX and GRX compared with the parental cells.78

AHR encodes a ligand-activated transcription factor binding to the xenobiotic response element-containing promoter region that is involved in the regulation of biological responses to planar aromatic hydrocarbons. This receptor has been shown to regulate levels of xenobiotic-metabolizing enzymes like CYP, of which CYP26B1 and 2B6 were comparatively overexpressed in UMC-11 cells.79 Human ALDH7A1 protects against hyperosmotic stress through generation of osmolytes and metabolization of toxic aldehydes.80 Human ALDH7A1 expression in Chinese hamster ovary cells attenuated osmotic stress-induced apoptosis caused by increased extracellular concentrations of sucrose or sodium chloride. AKRs are phase I drug-metabolizing enzymes for a variety of carbonyl-containing drugs and are implicated in cancer chemotherapeutic drug resistance. They are stress-regulated genes and play a pivotal role in the cellular response to osmotic, electrophilic, and oxidative stress. The 10 known human AKR enzymes can turnover a range of substrates, including drugs, carcinogens, and reactive aldehydes. AKRs like AKR7A3 are soluble NAD(P)H oxidoreductases that primarily reduce aldehydes and ketones to primary and secondary alcohols, respectively.81

Eukaryotic topo I, a DNA unwinding enzyme, is essential for several cellular functions and is the target of camptothecin derivatives like irinotecan and topotecan. However, regulation of topo I activity was not known for a long time. In an effort to identify potential regulators of TOP1 activity, at least 2 antisense transcripts coded by TOP1 pseudogenes were described.82 Although the function of these TOP1 antisense transcripts has remained unknown, recent studies of naturally occurring antisense RNA demonstrated a number of potential regulatory roles. Downregulation of topo I in carcinoids would increase resistance to camptothecin derivatives.

Conclusion

Multidrug-resistant UMC-11 cells seem to use classical drug transporters, such as P-gp and MRP, drug conjugation and metabolization systems, and possibly new mechanisms, such as suppression of apoptosis by S100 proteins and downregulation of topo I, by its own pseudogene transcripts to achieve chemoresistance. The genome-wide search for drug resistance-associated genes described here provides new evidence for possible targets to circumvent the chemoresistant phenotype of carcinoids. Functional involvement of the candidate genes found needs to be verified by experiments deleting the corresponding transcripts, and, furthermore, it remains to be investigated how far these results apply to other neuroendocrine tumors.83

Acknowledgment and disclosure

This study was supported by a fund of the Jubilaeumsfonds (OENB, grant # 13345). The authors report no conflicts of interest associated with this work.

References

  • 1.Öberg K. Diagnosis and treatment of carcinoid tumors. Expert Rev Anticancer Ther. 2003;3:863–877. doi: 10.1586/14737140.3.6.863. [DOI] [PubMed] [Google Scholar]
  • 2.Modlin IM, Kidd M, Latich I, et al. Current status of gastrointestinal carcinoids. Gastroenterology. 2005;128:1717–1751. doi: 10.1053/j.gastro.2005.03.038. [DOI] [PubMed] [Google Scholar]
  • 3.Pinchot SN, Holen K, Sippel RS, et al. Carcinoid tumors. Oncologist. 2008;13:1255–1269. doi: 10.1634/theoncologist.2008-0207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Zuetenhorst JM, Taal BG. Metastatic carcinoid tumors: a clinical review. Oncologist. 2005;10:123–131. doi: 10.1634/theoncologist.10-2-123. [DOI] [PubMed] [Google Scholar]
  • 5.Granberg D, Oberg K. Neuroendocrine tumours. Cancer Chemother Biol Response Modif. 2005;22:471–483. [PubMed] [Google Scholar]
  • 6.Capella C, Heitz PU, Hofler H, et al. Revised classification of neuroendocrine tumors of the lung, pancreas and gut. Virchows Arch. 1995;425:547–560. doi: 10.1007/BF00199342. [DOI] [PubMed] [Google Scholar]
  • 7.Warren WH, Gould VE, Faber LP, et al. Neuroendocrine neoplasms of the bronchopulmonary tract: a classification of the spectrum of carcinoid to small cell carcinoma and intervening variants. J Thorac Cardiovasc Surg. 1985;89:819–825. [PubMed] [Google Scholar]
  • 8.Smolle-Juttner FM, Popper H, Klemen H, et al. Clinical features and therapy of “typical” and “atypical” bronchial carcinoid tumors (grade 1 and grade 2 neuroendocrine carcinoma) Eur J Cardiothorac Surg. 1993;7:121–125. doi: 10.1016/1010-7940(93)90033-8. [DOI] [PubMed] [Google Scholar]
  • 9.Detterbeck FC. Management of carcinoid tumors. Ann Thorac Surg. 2010;89:998–1005. doi: 10.1016/j.athoracsur.2009.07.097. [DOI] [PubMed] [Google Scholar]
  • 10.Moertel CG, Sauer WG, Dockerty MB, et al. Life history of the carcinoid tumor of the small intestine. Cancer. 1961;14:901–912. doi: 10.1002/1097-0142(196109/10)14:5<901::aid-cncr2820140502>3.0.co;2-q. [DOI] [PubMed] [Google Scholar]
  • 11.Bertino EM, Confer PD, Colonna JE, et al. Pulmonary neuroendocrine/carcinoid tumors: a review article. Cancer. 2009;115:4434–4441. doi: 10.1002/cncr.24498. [DOI] [PubMed] [Google Scholar]
  • 12.Moertel CG. Treatment of the carcinoid tumor and the malignant carcinoid syndrome. J Clin Oncol. 1983;1:727–740. doi: 10.1200/JCO.1983.1.11.727. [DOI] [PubMed] [Google Scholar]
  • 13.Lyons JM, III, Abergel J, Thomson JL, et al. In vitro chemoresistance testing in well-differentiated carcinoid tumors. Ann Surg Oncol. 2009;16:649–655. doi: 10.1245/s10434-008-0261-z. [DOI] [PubMed] [Google Scholar]
  • 14.Kosmidis PA. Treatment of carcinoid of the lung. Curr Opin Oncol. 2004;16:146–149. doi: 10.1097/00001622-200403000-00011. [DOI] [PubMed] [Google Scholar]
  • 15.Sun W, Lipsitz S, Catalano P, et al. Phase II/III study of doxorubicin with fluorouracil compared with streptozocin with fluorouracil or dacarbazine in the treatment of advanced carcinoid tumors: Eastern Cooperative Oncology Group Study E1281. J Clin Oncol. 2005;23:4897–4904. doi: 10.1200/JCO.2005.03.616. [DOI] [PubMed] [Google Scholar]
  • 16.Fiebiger W, Olszewski U, Ulsperger E, et al. In vitro cytotoxicity of novel platinum-based drugs and dichloroacetate against lung carcinoid cell lines. Clin Transl Oncol. doi: 10.1007/s12094-011-0615-z. In press. [DOI] [PubMed] [Google Scholar]
  • 17.Guigay J, Ruffie P, Regnard JF, et al. Chemoresistance mechanisms and neuroendocrine lung tumors (NELT); Paper presented at: 1995 ASCO Annual Meeting; 1995. Abstract 398. [Google Scholar]
  • 18.Furuta E, Okuda H, Kobayashi A, et al. Metabolic genes in cancer: their roles in tumor progression and clinical implications. Biochim Biophys Acta. 2010;1805:141–152. doi: 10.1016/j.bbcan.2010.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Roche TE, Hiromasa Y. Pyruvate dehydrogenase kinase regulatory mechanisms and inhibition in treating diabetes, heart ischemia, and cancer. Cell Mol Life Sci. 2007;64:830–849. doi: 10.1007/s00018-007-6380-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Denko N, Schindler C, Koong A, et al. Epigenetic regulation of gene expression in cervical cancer cells by the tumor microenvironment. Clin Cancer Res. 2000;6:480–487. [PubMed] [Google Scholar]
  • 21.Kasper LH, Brindle PK. Mammalian gene expression program resiliency: the roles of multiple coactivator mechanisms in hypoxia-responsive transcription. Cell Cycle. 2006;5:142–146. doi: 10.4161/cc.5.2.2353. [DOI] [PubMed] [Google Scholar]
  • 22.Salnikow K, Davidson T, Zhang Q, et al. The involvement of hypoxia-inducible transcription factor-1-dependent pathway in nickel carcinogenesis. Cancer Res. 2003;63:3524–3530. [PubMed] [Google Scholar]
  • 23.Riddle SR, Ahmad A, Ahmad S, et al. Hypoxia induces hexokinase II gene expression in human lung cell line A549. Am J Physiol Lung Cell Mol Physiol. 2000;278:L407–L416. doi: 10.1152/ajplung.2000.278.2.L407. [DOI] [PubMed] [Google Scholar]
  • 24.Graven KK, McDonald RJ, Farber HW. Hypoxic regulation of endothelial glyceraldehyde-3-phosphate dehydrogenase. Am J Physiol. 1998;274:C347–C355. doi: 10.1152/ajpcell.1998.274.2.C347. [DOI] [PubMed] [Google Scholar]
  • 25.Koh MY, Spivak-Kroizman TR, Powis G. HIF-1alpha and cancer therapy. Recent Results Cancer Res. 2010;180:15–34. doi: 10.1007/978-3-540-78281-0_3. [DOI] [PubMed] [Google Scholar]
  • 26.Yang Z, Cox JL. Cathepsin L increases invasion and migration of B16 melanoma. Cancer Cell Int. 2007;7:8. doi: 10.1186/1475-2867-7-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Soreide K, Janssen EA, Körner H, et al. Trypsin in colorectal cancer: molecular biological mechanisms of proliferation, invasion, and metastasis. J Pathol. 2006;209:147–156. doi: 10.1002/path.1999. [DOI] [PubMed] [Google Scholar]
  • 28.Nannuru KC, Futakuchi M, Varney ML, et al. Matrix metalloproteinase (MMP)-13 regulates mammary tumor-induced osteolysis by activating MMP9 and transforming growth factor-β signaling at the tumor-bone interface. Cancer Res. 2010;70:3494–3504. doi: 10.1158/0008-5472.CAN-09-3251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Voland P, Besig S, Rad R, et al. Correlation of matrix metalloproteinases and tissue inhibitors of matrix metalloproteinase expression in ileal carcinoids, lymph nodes and liver metastasis with prognosis and survival. Neuroendocrinology. 2009;89:66–78. doi: 10.1159/000151482. [DOI] [PubMed] [Google Scholar]
  • 30.Oikonomopoulou K, Diamandis EP, Hollenberg MD. Kallikrein-related peptidases: proteolysis and signaling in cancer, the new frontier. Biol Chem. 2010;391:299–310. doi: 10.1515/BC.2010.038. [DOI] [PubMed] [Google Scholar]
  • 31.Leja J, Essaghir A, Essand M, et al. Novel markers for enterochromaffin cells and gastrointestinal neuroendocrine carcinomas. Mod Pathol. 2009;22:261–272. doi: 10.1038/modpathol.2008.174. [DOI] [PubMed] [Google Scholar]
  • 32.Franke RM, Scherkenbach LA, Sparreboom A. Pharmacogenetics of the organic anion transporting polypeptide 1A2. Pharmacogenomics. 2009;10:339–344. doi: 10.2217/14622416.10.3.339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Shimizu S, Eguchi Y, Kosaka H, et al. Prevention of hypoxia-induced cell death by Bcl-2 and Bcl-xL. Nature. 1995;374:811–813. doi: 10.1038/374811a0. [DOI] [PubMed] [Google Scholar]
  • 34.Jones PM, O’Mara ML, George AM. ABC transporters: a riddle wrapped in a mystery inside an enigma. Trends Biochem Sci. 2009;34:520–531. doi: 10.1016/j.tibs.2009.06.004. [DOI] [PubMed] [Google Scholar]
  • 35.Goldstein LJ, Galski H, Fojo A, et al. Expression of a multidrug resistance gene in human cancers. J Natl Cancer Inst. 1989;81:116–124. doi: 10.1093/jnci/81.2.116. [DOI] [PubMed] [Google Scholar]
  • 36.Giaccone G, van Ark-Otte J, Rubio GJ, et al. MRP is frequently expressed in human lung-cancer cell lines, in non-small-cell lung cancer and in normal lungs. Int J Cancer. 1996;66:760–767. doi: 10.1002/(SICI)1097-0215(19960611)66:6<760::AID-IJC9>3.0.CO;2-Y. [DOI] [PubMed] [Google Scholar]
  • 37.Ballatori N, Krance SM, Marchan R, et al. Plasma membrane glutathione transporters and their roles in cell physiology and pathophysiology. Mol Aspects Med. 2009;30:13–28. doi: 10.1016/j.mam.2008.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Mijatovic T, Ingrassia L, Facchini V, et al. Na+/K+-ATPase α subunits as new targets in anticancer therapy. Expert Opin Ther Targets. 2008;12:1403–1417. doi: 10.1517/14728222.12.11.1403. [DOI] [PubMed] [Google Scholar]
  • 39.Simpson CD, Mawji IA, Anyiwe K, et al. Inhibition of the sodium potassium adenosine triphosphatase pump sensitizes cancer cells to anoikis and prevents distant tumor formation. Cancer Res. 2009;69:2739–2747. doi: 10.1158/0008-5472.CAN-08-2530. [DOI] [PubMed] [Google Scholar]
  • 40.Brouillard F, Tondelier D, Edelman A, et al. Drug resistance induced by ouabain via the stimulation of MDR1 gene expression in human carcinomatous pulmonary cells. Cancer Res. 2001;61:1693–1698. [PubMed] [Google Scholar]
  • 41.Ahmed Z, Deyama Y, Yoshimura Y, et al. Cisplatin sensitivity of oral squamous carcinoma cells is regulated by Na(+),K(+)-ATPase activity rather than copper-transporting P-type ATPases, ATP7A and ATP7B. Cancer Chemother Pharmacol. 2009;63:643–650. doi: 10.1007/s00280-008-0781-z. [DOI] [PubMed] [Google Scholar]
  • 42.Mijatovic T, Jungwirth U, Heffeter P, et al. The Na+/K+-ATPase is the Achilles heel of multi-drug-resistant cancer cells. Cancer Lett. 2009;282:30–34. doi: 10.1016/j.canlet.2009.02.048. [DOI] [PubMed] [Google Scholar]
  • 43.Geering K. FXYD proteins: new regulators of Na-K-ATPase. Am J Physiol Renal Physiol. 2006;290:F241–F250. doi: 10.1152/ajprenal.00126.2005. [DOI] [PubMed] [Google Scholar]
  • 44.Yamamoto H, Okumura K, Toshima S, et al. FXYD3 protein involved in tumor cell proliferation is overproduced in human breast cancer tissues. Biol Pharm Bull. 2009;32:1148–1154. doi: 10.1248/bpb.32.1148. [DOI] [PubMed] [Google Scholar]
  • 45.Blobe GC, Schiemann WP, Lodish HF. Role of transforming growth factor beta in human disease. N Engl J Med. 2000;342:1350–1358. doi: 10.1056/NEJM200005043421807. [DOI] [PubMed] [Google Scholar]
  • 46.Elliott RL, Blobe GC. Role of transforming growth factor β in human cancer. J Clin Oncol. 2005;23:2078–2093. doi: 10.1200/JCO.2005.02.047. [DOI] [PubMed] [Google Scholar]
  • 47.Zhang J, Iwanaga K, Choi KC, et al. Intratumoral epiregulin is a marker of advanced disease in non-small cell lung cancer patients and confers invasive properties on EGFR-mutant cells. Cancer Prev Res (Phila Pa) 2008;1:201–207. doi: 10.1158/1940-6207.CAPR-08-0014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Busser B, Sancey L, Josserand V, et al. Amphiregulin promotes BAX inhibition and resistance to gefitinib in non-small-cell lung cancers. Mol Ther. 2010;18:528–535. doi: 10.1038/mt.2009.226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Papouchado B, Erickson LA, Rohlinger AL, et al. Epidermal growth factor receptor and activated epidermal growth factor receptor expression in gastrointestinal carcinoids and pancreatic endocrine carcinomas. Mod Pathol. 2005;8:1329–1335. doi: 10.1038/modpathol.3800427. [DOI] [PubMed] [Google Scholar]
  • 50.Rickman OB, Vohra PK, Sanyal B, et al. Analysis of ErbB receptors in pulmonary carcinoid tumors. Clin Cancer Res. 2009;15:3315–3324. doi: 10.1158/1078-0432.CCR-08-2549. [DOI] [PubMed] [Google Scholar]
  • 51.Shimamura T, Shapiro GI. Heat shock protein 90 inhibition in lung cancer. J Thorac Oncol. 2008;3:S152–S159. doi: 10.1097/JTO.0b013e318174ea3a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Egan JE, Hall AB, Yatsula BA, Bar-Sagi D. The bimodal regulation of epidermal growth factor signaling by human Sprouty proteins. Proc Natl Acad Sci U S A. 2002;99:6041–6046. doi: 10.1073/pnas.052090899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Wulbrand U, Remmert G, Zöfel P, et al. mRNA expression patterns of insulin-like growth factor system components in human neuroendocrine tumours. Eur J Clin Invest. 2000;30:729–739. doi: 10.1046/j.1365-2362.2000.00700.x. [DOI] [PubMed] [Google Scholar]
  • 54.Camidge DR, Dziadziuszko R, Hirsch FR. The rationale and development of therapeutic insulin-like growth factor axis inhibition for lung and other cancers. Clin Lung Cancer. 2009;10:262–272. doi: 10.3816/CLC.2009.n.037. [DOI] [PubMed] [Google Scholar]
  • 55.Kawanami Y, Morimoto Y, Kim H, et al. Calcitonin gene-related peptide stimulates proliferation of alveolar epithelial cells. Respir Res. 2009;10:8. doi: 10.1186/1465-9921-10-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Aiyar N, Disa J, Stadel JM, et al. Calcitonin gene-related peptide receptor independently stimulates 3′,5′-cyclic adenosine monophosphate and Ca2+ signaling pathways. Mol Cell Biochem. 1999;197:179–185. doi: 10.1023/a:1006962221332. [DOI] [PubMed] [Google Scholar]
  • 57.Zhu YM, Webster SJ, Flower D, et al. Interleukin-8/CXCL8 is a growth factor for human lung cancer cells. Br J Cancer. 2004;91:1970–1976. doi: 10.1038/sj.bjc.6602227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Olsen SK, Garbi M, Zampieri N, et al. Fibroblast growth factor (FGF) homologous factors share structural but not functional homology with FGFs. J Biol Chem. 2003;278:34226–34236. doi: 10.1074/jbc.M303183200. [DOI] [PubMed] [Google Scholar]
  • 59.Ager EI, Neo J, Christophi C. The renin-angiotensin system and malignancy. Carcinogenesis. 2008;29:1675–1684. doi: 10.1093/carcin/bgn171. [DOI] [PubMed] [Google Scholar]
  • 60.Moody TW, Hill JM, Jensen RT. VIP as a trophic factor in the CNS and cancer cells. Peptides. 2003;24:163–177. doi: 10.1016/s0196-9781(02)00290-5. [DOI] [PubMed] [Google Scholar]
  • 61.Myers RM, Shearman JW, Kitching MO, et al. Cancer, chemistry, and the cell: molecules that interact with the neurotensin receptors. ACS Chem Biol. 2009;4:503–525. doi: 10.1021/cb900038e. [DOI] [PubMed] [Google Scholar]
  • 62.Calhoun K, Toth-Fejel S, Cheek J, et al. Serum peptide profiles in patients with carcinoid tumors. Am J Surg. 2003;186:28–31. doi: 10.1016/s0002-9610(03)00115-6. [DOI] [PubMed] [Google Scholar]
  • 63.Moody TW. Peptide hormones and lung cancer. Panminerva Med. 2006;48:19–26. [PubMed] [Google Scholar]
  • 64.Matsumoto T, Kawashima Y, Nagashio R, et al. A new possible lung cancer marker: VGF detection from the conditioned medium of pulmonary large cell neuroendocrine carcinoma-derived cells using secretome analysis. Int J Biol Markers. 2009;24:282–285. doi: 10.1177/172460080902400411. [DOI] [PubMed] [Google Scholar]
  • 65.Andreis PG, Rucinski M, Neri G, et al. Neuropeptides B and W enhance the growth of human adrenocortical carcinoma-derived NCI-H295 cells by exerting MAPK p42/p44-mediated proliferogenic and antiapoptotic effects. Int J Mol Med. 2005;16:1021–1028. [PubMed] [Google Scholar]
  • 66.Gudermann T, Roelle S. Calcium-dependent growth regulation of small cell lung cancer cells by neuropeptides. Endocr Relat Cancer. 2006;13:1069–1084. doi: 10.1677/erc.1.01302. [DOI] [PubMed] [Google Scholar]
  • 67.Townsend CM, Jr, Bold RJ, Ishizuka J. Gastrointestinal hormones and cell proliferation. Surg Today. 1994;24:772–777. doi: 10.1007/BF01636304. [DOI] [PubMed] [Google Scholar]
  • 68.Schüller HM, Orloff M, Reznik GK. Antiproliferative effects of the Ca2+/calmodulin antagonist B859-35 and the Ca(2+)-channel blocker verapamil on human lung cancer cell lines. Carcinogenesis. 1991;12:2301–2303. doi: 10.1093/carcin/12.12.2301. [DOI] [PubMed] [Google Scholar]
  • 69.Tang H, Liu YJ, Liu M, et al. Establishment and gene analysis of an oxaliplatin-resistant colon cancer cell line THC8307/L-OHP. Anticancer Drugs. 2007;18:633–639. doi: 10.1097/CAD.0b013e3280200428. [DOI] [PubMed] [Google Scholar]
  • 70.Stewart JJ, White JT, Yan X, et al. Proteins associated with Cisplatin resistance in ovarian cancer cells identified by quantitative proteomic technology and integrated with mRNA expression levels. Mol Cell Proteomics. 2006;5:433–443. doi: 10.1074/mcp.M500140-MCP200. [DOI] [PubMed] [Google Scholar]
  • 71.Mahon PC, Baril P, Bhakta V, et al. S100A4 contributes to the suppression of BNIP3 expression, chemoresistance, and inhibition of apoptosis in pancreatic cancer. Cancer Res. 2007;67:6786–6795. doi: 10.1158/0008-5472.CAN-07-0440. [DOI] [PubMed] [Google Scholar]
  • 72.Guo B, Villeneuve DJ, Hembruff SL, et al. 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. doi: 10.1023/B:BREA.0000021046.29834.12. [DOI] [PubMed] [Google Scholar]
  • 73.Lee WY, Su WC, Lin PW, et al. Expression of S100A4 and Met: potential predictors for metastasis and survival in early-stage breast cancer. Oncology. 2004;66:429–438. doi: 10.1159/000079496. [DOI] [PubMed] [Google Scholar]
  • 74.Stengel C, Newman SP, Leese MP, et al. Class III beta-tubulin expression and in vitro resistance to microtubule targeting agents. Br J Cancer. 2010;102:316–324. doi: 10.1038/sj.bjc.6605489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Györffy B, Dietel M, Fekete T, et al. A snapshot of microarray-generated gene expression signatures associated with ovarian carcinoma. Int J Gynecol Cancer. 2008;18:1215–1233. doi: 10.1111/j.1525-1438.2007.01169.x. [DOI] [PubMed] [Google Scholar]
  • 76.Wang X, Guo Z. The role of sulfur in platinum anticancer chemotherapy. Anticancer Agents Med Chem. 2007;7:19–34. doi: 10.2174/187152007779314062. [DOI] [PubMed] [Google Scholar]
  • 77.Ekhart C, Rodenhuis S, Smits PH, et al. An overview of the relations between polymorphisms in drug metabolising enzymes and drug transporters and survival after cancer drug treatment. Cancer Treat Rev. 2009;35:18–31. doi: 10.1016/j.ctrv.2008.07.003. [DOI] [PubMed] [Google Scholar]
  • 78.Nakamura H, Bai J, Nishinaka Y, Ueda S, et al. Expression of thioredoxin and glutaredoxin, redox-regulating proteins, in pancreatic cancer. Cancer Detect Prev. 2000;24:53–60. [PubMed] [Google Scholar]
  • 79.Njar VC. Cytochrome p450 retinoic acid 4-hydroxylase inhibitors: potential agents for cancer therapy. Mini Rev Med Chem. 2002;2:261–269. doi: 10.2174/1389557023406223. [DOI] [PubMed] [Google Scholar]
  • 80.Brocker C, Lassen N, Estey T, et al. Aldehyde dehydrogenase 7A1 (ALDH7A1) is a novel enzyme involved in cellular defense against hyperosmotic stress. J Biol Chem. 2010;285:18452–18463. doi: 10.1074/jbc.M109.077925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Jin Y, Penning TM. Aldo-keto reductases and bioactivation/detoxication. Annu Rev Pharmacol Toxicol. 2007;47:263–292. doi: 10.1146/annurev.pharmtox.47.120505.105337. [DOI] [PubMed] [Google Scholar]
  • 82.Zhou BS, Beidler DR, Cheng YC. Identification of antisense RNA transcripts from a human DNA topoisomerase I pseudogene. Cancer Res. 1992;52:4280–4285. [PubMed] [Google Scholar]
  • 83.Shanker M, Willcutts D, Roth JA, et al. Drug resistance in lung cancer. Lung Cancer: Targets and Ther. 2010;1:23–36. [PMC free article] [PubMed] [Google Scholar]

Articles from Lung Cancer: Targets and Therapy are provided here courtesy of Dove Press

RESOURCES