Skip to main content
Erschienen in: Molecular Cancer 1/2016

Open Access 01.12.2016 | Research

Insulin-like growth factors are essential to prevent anoikis in oestrogen-responsive breast cancer cells: importance of the type I IGF receptor and PI3-kinase/Akt pathway

verfasst von: Brendan C. Luey, Felicity E. B. May

Erschienen in: Molecular Cancer | Ausgabe 1/2016

Abstract

Background

Detachment of epithelial cells from the extracellular matrix initiates programmed cell death by a process termed anoikis. Malignant cells must acquire anoikis resistance to leave the primary tumour and metastasise. Multiple signal transduction pathways can activate anoikis and confer anoikis resistance, but these are not understood in breast cancer.

Methods

Models for anoikis of oestrogen-responsive breast cancer cells were established and the protective effects of IGF-1 tested. Cleaved PARP was measured by western transfer and cleaved caspase 3 by flow cytometry. Pathways involved in anoikis and in anoikis resistance were investigated with PI3-kinase, Akt, and MEK1 and MEK2 inhibitors. The importance of the type I IGF receptor was investigated by IGF-concentration dependence, siRNA knockdown and pharmacological inhibition. Association between IGF-1R expression and relapse with distant metastasis was analysed in 1609 patients by log rank test.

Results

Unattached breast cancer cells required culture in serum-free medium to induce anoikis. Rapid loss of FAK, Akt and Bad phosphorylation was concurrent with anoiks induction, but ERK1 and ERK2 phosphorylation increased which suggested that anoikis resistance is mediated by the PI3-kinase/Akt rather than the Grb2/Ras/MAP-kinase pathway. IGF-1 conferred anoikis resistance in serum-free medium. IGF-1 activated the PI3-kinase/Akt and Grb2/Ras/MAP-kinase pathways but experiments with PI3-kinase, Akt and MEK1 and MEK2 inhibitors showed that IGF protection is via the PI3-kinase/Akt pathway. The concentration dependence of IGF protection, knockdown experiments with siRNA and pharmacological inhibition with figitumumab, showed that IGF-1 signals through the type I IGF receptor. The crucial role of the type I IGF receptor was demonstrated by induction of anoikis in full serum by figitumumab. High IGF-1R expression was associated with reduced time to relapse with distant metastases in oestrogen receptor-positive patients, especially those with aggressive disease which confirms its relevance in vivo.

Conclusions

Anoikis resistance of oestrogen-responsive breast cancer cells depends upon IGF activation of the type I IGF receptor and PI3-kinase/Akt pathway. Because IGF-dependent evasion of anoikis will facilitate metastasis by malignant breast cancer cells, effective inhibition of IGF signal transduction should be included in combinations of targeted drugs designed to treat metastatic oestrogen receptor-positive breast cancers.
Hinweise

Competing interests

The authors declare no competing interest.

Authors’ contributions

BCL conceived and undertook experiments and analysed data. FEBM conceived experiments. Both authors were involved in preparation of the paper. Both authors read and approved the final manuscript.
Abkürzungen
MAP-kinase
mitogen-activated protein kinase
IGF
insulin-like growth factor
siRNA
small interfering RNA
FAK
focal adhesion kinase
poly-HEMA
poly(hydroxyethyl methacrylic)
RIPA
radioimmunoprecipitate

Background

Worldwide, more than 1.7 million women are diagnosed annually with breast cancer of whom at least 500,000 die as a result of metastatic disease [1]. Metastasis necessitates release of malignant cells from the primary tumour and their movement to and establishment at distant sites. Normal breast epithelial and myoepithelial cells attach to each other and to the extracellular matrix. Loss of these attachments induces programmed cell death in a process called anoikis [24]. Breast cancer cells must become resistant to anoikis as they invade breast and surrounding tissue, intravasate into blood and lymphatic vessels and metastasise [5]. Blockade of the pathways responsible for anoikis resistance offers a powerful strategy for the elimination of metastatic cells.
Integrins are transmembrane proteins that provide connections between extracellular matrix proteins and the actin-based cytoskeleton in normal tissues. Integrins transduce signals via sub-membrane, focal adhesion protein complexes, called focal adhesions, which connect integrins with signal transduction proteins [6, 7]. Integrin engagement activates focal adhesion kinase (FAK) to suppress anoikis [3] by transmission of cell survival signals through multiple signal transduction pathways [3, 8].
The effects of insulin-like growth factors (IGFs) are mediated by the transmembrane type I IGF or insulin receptors and multiple intracellular signal transduction pathways [9]. Work with transgenic animals implicates IGFs in carcinogenesis [10] and they are significant regulators of breast cancer cell proliferation and invasion [1115]. Consequently, the IGF signal transduction pathway has been identified as a therapeutic target and inhibitors of the type I IGF receptor have been developed by pharmaceutical companies [9, 1621].
IGFs confer anoikis resistance in embryonic fibroblasts that have been engineered to overexpress the type I IGF receptor [22] but the importance of IGFs in anoikis resistance and the mechanism by which they might act in oestrogen-responsive breast cancer is unknown. Similarly, the signal transduction pathways involved in the induction of anoikis and in anoikis resistance of breast cancer cells have not been determined.
There have been a few studies that purport to examine the effects of the IGF signal transduction pathway upon anoikis breast cancer cells. However, caspase-dependent programmed cell death was not measured in these studies. In immortalized, normal MCF710A cells that have been modified to overexpress the type I IGF receptor, PTK6 increased signal transduction through the type I IGF receptor and IRS-1 increased the number of viable cells grown in unattached conditions [23]. Another study reported that effects of IGF on the ratio of isoforms of the C/EBPβ protein reduced the proportion of unattached MCF710A cells in sub-G1 phase [24]. Disruption of the type I IGF receptor signal transduction pathway decreased numbers of viable cells of a metastatic variant of MDA-MB-435 breast cancer cell line grown as unattached cells [14]. Another study reported that activation of the p53 pathway after MCF-7 cell detachment leads to a caspase-independent reduction in mitochondrial activity, and that calveolin may reverse the reduction via an increase in type I IGF receptor [25]. Thus, despite the impression conveyed in the titles or abstracts of these articles, there have been no studies of the effects of IGFs on anoikis in human breast cancer cells.
We have shown that IGFs inhibit apoptosis in triple-negative breast cancer cells [12] which suggested that they could protect against breast cancer cell anoikis and that blockade of the IGF signal transduction pathway might offer a strategy for promoting anoikis and reducing metastasis. The overall aim of the current study was to investigate the mechanisms by which oestrogen-responsive breast cancer cells evade anoikis. We established an in vitro model of anchorage-independent, caspase-dependent cell death and investigated the changes in intracellular signal transduction involved, whether IGF-1 protects the cells from anoikis and the receptor and signal transduction pathway through which IGFs act.

Results

Model of anoikis in oestrogen-responsive breast cancer

MCF-7 cells were added to uncoated or poly-HEMA-coated culture wells to prevent cell attachment [26]. After 24 h, cells in the poly-HEMA-coated wells grew as rounded cells in suspension (Fig. 1). To investigate if the unattached MCF-7 cells had undergone programmed cell death via the caspase-dependent pathway, we measured the cleavage of PARP into the 89 kDa catalytic and 24 kDa DNA binding subunits which cannot repair single-strand DNA breaks. No cleaved PARP was detected in attached or unattached cells cultured in maintenance medium. Attached cells grown in serum-free medium for 24 h maintained their characteristic polygonal morphology and PARP cleavage was not detected. PARP cleavage was induced, however, in unattached cells after 24 h in serum-free medium. Culture of attached cells in serum-free medium for up to three days did not induce significant cell death (data not shown).
Oestrogen-responsive breast cancer cells, ZR-75 and EFM-19, also lost their characteristic epithelial appearance and grew as rounded cells after 24 h culture in poly-HEMA-coated wells in serum-free medium (Fig. 1A-C). A small amount of cleaved PARP was detected in unattached ZR-75 after 5 h and substantially more after 24 h. Cleaved PARP was detected readily in unattached EFM-19 at 1 h and was almost maximal after 6 h. Anoikis was induced also in T-47D cells after 24 h (data not shown). PARP cleavage was not detected in attached cells grown in serum-free medium.
Caspase 3 is important in the execution phase of caspase-dependent programmed cell death. It is activated by cleavage into 17 kDa and 12 kDa subunits; the presence of which indicate that cells are in the execution stages of programmed cell death. Activated caspase 3 was detected by flow cytometry in less than 1 % of the cells prior to and in 15 % of the cells after induction of anoikis (Fig. 1D; student t-test; p < 0.01) which confirmed that caspase-dependent anoikis had been induced.

Effect of loss of attachment on signal transduction

To investigate alterations in signal transduction that could account for the anoikic response, EFM-19 cells were grown attached or unattached in serum-free medium for different lengths of time. PARP cleavage was detected after 1 h in the unattached cells and increased thereafter for 24 h (Fig. 2). Little PARP cleavage was detected in attached cells even after 24 h.
The phosphorylation of proteins implicated in anoikis was then measured. There was a dramatic decrease in FAK phosphorylation within 15 min of prevention of cell attachment whereas there was no reduction in FAK phosphorylation in attached cells cultured in serum-free medium for the same length of time. The levels of phosphorylated FAK were minimal by 1 h and decreased further up to 24 h concurrent with the increase in PARP cleavage. There was no concomitant change in the levels of FAK protein. FAK phosphorylation was reduced also in unattached MCF-7 and ZR-75 cultured in poly-HEMA-coated dishes in serum free medium for 24 h. Consistent with a role of the PI3-kinase/Akt pathway in anoikis, Akt phosphorylation was reduced dramatically in MCF-7 and to a lesser extent in ZR-75 and EFM-19. The reduction of Akt phosphorylation led to a reduction in phosphorylation of its downstream target, Bad, in MCF-7 and EFM-19. Reduced phosphorylation of Bad promotes cell death because unphosphorylated Bad sequesters anti-apoptotic Bcl family proteins which prevents their inhibition of pro-apoptotic proteins Bax and Bak.
In contrast to Akt phosphorylation, phosphorylation of ERK1 and ERK2 was increased slightly in MCF-7 and markedly in EFM-19 and ZR-75 cells induced to undergo anoikis which suggests that the Grb2/Ras/MAP-kinase pathway has not been dampened and that it is unlikely to transduce the integrin-dependent cell survival signal. The results are consistent with oestrogen-responsive breast cancer cell attachment activating FAK to signal through the PI3-kinase/Akt/Bad pathway to provide an important cell survival signal in attached oestrogen-responsive breast cancer cells.

IGFs protect oestrogen-responsive breast cancer cells from anoikis

Many factors in serum might account for the anoikis resistance of oestrogen-responsive breast cancer cells. To test if IGFs in serum might contribute to the anoikis resistance of unattached oestrogen-responsive breast cancer cells cultured in maintenance medium, cells were cultured in poly(HEMA)-coated wells in serum-free medium in the absence or presence of IGF-1 (Fig. 3). The amount of cleaved PARP detected was lower in cells cultured in the presence of IGF-1 than in its absence (p < 0.001). IGF-1 prevented completely the induction of anoikis in MCF-7, inhibited it thirty-fold in ZR-75 and ten-fold in EFM-19 cells.
The protective effect of IGF-1 on the induction of anoikis was analysed also by measuring activated caspase 3. The proportion of EFM-19 cells with activated caspase 3 in the execution phase of cell death was reduced by 78 % in cells cultured in poly-HEMA-coated dishes in the presence of IGF-1 compared to in cells cultured in its absence (Fig. 3C; p < 0.01), which confirms that IGF-1 protects cells from caspase-dependent anoikis and suggests that they might be important contributors to the resistance conveyed by culture in serum.
The downstream signal transduction pathways that might be responsible for the protective effect of IGF-1 in oestrogen-responsive breast cancer cells were investigated. IGF-1 stimulated auto-phosphorylation of IGF receptors in all three cell lines (Fig. 4). Activated IGF receptors interact with and induce phosphorylation of adaptor proteins of which IRS-1 is considered to be the most important for signalling via both PI3-kinase/Akt and Grb2/Ras/MAP-kinase pathways [27]. IRS-1 phosphorylation was induced by nanomolar concentrations of IGF-1. Phosphorylation of Akt was stimulated by 2 ng/ml IGF-1 in MCF-7 and ZR-75 and by 20 ng/ml IGF-1 in EFM-19, and of Bad by 0.5 ng/ml IGF-1 in MCF-7 and ZR-75 and by 20 ng/ml IGF-1 in EFM-19. Activation by phosphorylation of ERK1 and ERK2 was stimulated most markedly in ZR-75 cells but required higher IGF-1 concentrations, 20 ng/ml, than for the stimulation of Akt phosphorylation. The total levels of the signal transduction proteins did not change during the IGF-1 treatment.
These experiments show that oestrogen-responsive cells are protected from cell death by anchorage dependent signals from integrins through FAK and Akt (Fig. 5A). Resistance to anoikis can be mediated through the IGF signal transduction pathway. IGFs activate the PI3-kinase/Akt, and the Grb2/Ras/MAP-kinase signal transduction pathways both of which have been implicated in anoikis resistance.

Signal transduction pathway that mediates the anti-anoikic effect of IGF-1

The importance of the PI3-kinase/Akt pathway in the IGF-dependent anoikis resistance was tested with LY294002, a selective, reversible inhibitor of ATP binding in the catalytic subunit of PI3-kinase [28, 29]. Unattached MCF-7 cells were cultured in serum-free medium in poly-HEMA-coated wells in the absence or presence of IGF-1 and LY294002. Neither the IGF receptors nor Akt were phosphorylated in untreated cells which indicates that there is no signalling through the PI3-kinase/Akt pathway (Fig. 6). Phosphorylation of the IGF receptors stimulated by IGF-1 was not affected by LY294002 whereas IGF-stimulated phosphorylation of Akt was prevented completely. Substantial amounts of cleaved PARP were detected in the untreated cells but very little in the presence of IGF-1. The PI3-kinase inhibitor did not increase anoikis in the absence of IGF-1 which is consistent with this pathway being inactivated completely in unattached cells in serum-free medium. The ability of IGF-1 to prevent cell death was reduced 20-fold by the PI3-kinase inhibitor (ANOVA; p < 0.01).
The role of the PI3-kinase/Akt pathway in IGF-protection of breast cancer cells from anoikis was tested further with GSK690693 [30, 31] a specific, ATP-competitive inhibitor of Akt. Activity of the Akt inhibitor was demonstrated by enhanced Akt phosphorylation in the presence of IGF-1 as has been reported previously (data not shown). Phosphorylation of GSK3β, a downstream target of Akt, was stimulated by IGF-1 (Fig. 6) and GSK690693 inhibited this IGF-stimulated phosphorylation. GSK690693 did not increase anoikis in unattached cells but did inhibit the protective effect of IGF-1 which reinforced the supposition that the IGF-protective-effect is transduced via the PI3-kinase/Akt pathway (Fig. 5B).
The contribution of the Grb2/Ras/MAP-kinase pathway to the protection by IGF-1 of oestrogen-responsive breast cancer cells from anoikis was tested with U0126 which is a non-competitive inhibitor of MEK1 and MEK2 [32, 33] that prevents activation of ERK1 and ERK2. Phosphorylated ERK1 and ERK2 were detected in cells in which anoikis had been induced (Fig. 7) which suggests that the Grb2/Ras/MAP-kinase pathway is not inactivated. IGF-1 stimulated receptor phosphorylation, increased ERK1 and ERK2 phosphorylation, and protected cells from anoikis. ERK1 and ERK2 phosphorylation was abrogated completely in the presence of the MEK inhibitor and more cleaved PARP was detected. To investigate if the increased cell death induced by U0126 resulted from increased anoikis or was independent of cell attachment, attached cells were incubated in serum-free medium in the absence and presence of U0126. MCF-7 cultured as attached cells in serum-free medium for up to 24 h did not undergo apoptosis. Apoptosis was induced strongly in the presence of U0126 (Fig. 7C) which indicates that cell death induced by MEK inhibition is independent of cell attachment.
There was no phosphorylated ERK1 or ERK2 in cells incubated in the absence or presence of IGF-1 in the presence of the MEK inhibitor but there was a significant protective effect of IGF-1 against anoikis in the presence of the MEK inhibitor as evidenced by its ability to prevent PARP cleavage. These data demonstrate that the protective effect of IGF-1 against anoikis is not transduced via the Grb2/Ras/MAP-kinase pathway (Fig. 5C).

Importance of the type I IGF receptor

Oestrogen-responsive breast cancer cells express relatively more type I IGF receptor than the insulin receptor [12]. IGF-1 has a higher affinity for the former receptor [9, 34]. To investigate which receptor transmits the IGF-protective signal against anoikis, unattached cells were cultured in the presence of different concentrations of IGF-1 (Fig. 8A). There was a small reduction in the amount of cleaved PARP in MCF-7 cells after culture in 0.5 ng/ml IGF-1 compared to in serum-free medium alone, which was significant in 1 ng/ml IGF-1 and was almost complete in the presence of 2 ng/ml IGF-1 and above. IGF-1 was less potent in EFM-19 than in MCF-7 but in both cells lines the protective effect of IGF-1 was maximal at a concentration of 15 ng/ml which indicates that the protective effect of IGF-1 in oestrogen-responsive breast cancer cells is mediated by the type I IGF receptor. The concentrations of IGF-1 at which significant prevention of anoikis was detected coincided with the concentrations that stimulated Akt and Bad phosphorylation (Fig. 4).
Confirmation of the importance of the type I IGF receptor in the IGF-1 protection was sought with siRNA knockdown. Expression of the type I IGF receptor by MCF-7 cells was reduced to undetectable levels after transfection with an siRNA oligonucleotide directed against the type I IGF receptor (Fig. 8B). PARP cleavage in cells cultured in poly-HEMA-coated wells to induce anoikis was inhibited by IGF-1 in untransfected cells and in cells transfected with scrambled oligonucleotide but not in cells in which expression of the type I IGF receptor had been knocked down. The concentration dependence of the IGF-1 anoikis protection, and the inability of IGF-1 to prevent anoikis in cells without type I IGF receptor expression, confirm that the protective effect of IGF-1 against anoikis is transmitted by the type I IGF receptor in these oestrogen-responsive breast cancer cells.
The importance of the type I IGF receptor was corroborated by pharmacological inhibition with the inhibitory antibody, figitumumab. Expression of the type I IGF receptor was barely detectable after incubation with the anti-receptor antibody (Fig. 8C). Incubation of cells with figitumumab reduced significantly the ability of IGF-1 to protect cells from anoikis (Fig. 8C).
To investigate the generality of the importance of the type I IGF receptor and signalling through the IGF transduction pathway in the anoikis resistance of oestrogen-responsive breast cancer cells, unattached cells were incubated in the presence of figitumumab in serum-containing medium. Incubation with figitumumab reduced the amount of the type I IGF receptor and the activation of Akt by phosphorylation of Ser473, which indicates that the type I IGF receptor and the IGF signal transduction pathway are major activators of the PI3-kinase/Akt pathway in oestrogen-responsive breast cancer cells (Fig. 8D).
Importantly, incubation of the cells with figitumumab induced significantly anoikis in the presence of serum. These data demonstrate that abrogation of the IGF-protective effect by pharmacological inhibition of the type I IGF receptor is able to circumvent the protective effects of anti-anoikis factors that are present in serum (Fig. 5B). The effectiveness of figitumumab indicates that the factors that signal through the IGF signal transduction pathway have a major role in the anoikis resistance of oestrogen-responsive breast cancer cells.
The pivotal role of the type I IGF receptor in protection of oestrogen-responsive breast cancer cells from anoikis, which could be critical for the ectopic survival of the malignant cells, suggests that receptor expression might facilitate metastasis. Amongst all patients, there was a trend towards a longer time to relapse with distant metastasis for patients with high type I IGF receptor expression (Fig. 9A) which is consistent with previous studies that have demonstrated survival benefit of high type I IGF receptor expression [35, 36]. However, amongst the sub-group of patients with oestrogen receptor-positive breast cancer, those with higher type I IGF receptor expression had shorter time to relapse with distant metastasis than those with lower expression (log rank; p = 0.032) (Fig. 9B). The difference in time to relapse between patients with high and low receptor expression was even more significant in the subgroup of patients with oestrogen receptor-positive tumours and lymph node involvement (p = 0.0057), and was most significant in patients with oestrogen receptor-positive, grade 3 tumours (p = 0.0035) (Fig. 9C and D).

Discussion

The majority of breast cancer deaths are caused by the effects of distant metastases in vital organs. Metastatic malignant cells accrue a plethora of characteristics in distinct phases: metastasis initiation, progression and virulence [5]. They must be able to survive without normal cellular and cell matrix attachments to initiate metastasis. The development of resistance to anoikis, the apoptotic process triggered by inappropriate or absence of cell adhesion is an important facet of this transition.
Improvements in the management and survival of breast cancer patients and identification of therapeutic targets will be facilitated by a more profound understanding of metastasis. To this end, we have established a model of oestrogen-responsive breast cancer cell anoikis. Anoikis can be mediated through extrinsic, intrinsic or caspase-independent apoptotic pathways [37, 38]. There have been limited studies on anoikis in breast cancer. Breast cancer cells as exemplified by Hs578T, MDA-MB-231, BT-474 and T-47D have been reported to be inherently resistant to anoikis and it was concluded that this resistance does not involve the PI3-kinase/Akt or Grb2/Ras/MAP-kinase pathways [39]. In contrast, our experiments have identified conditions that allow analysis of breast cancer cell anoikis resistance and have highlighted the importance of the intrinsic apoptotic pathway.
Several signal transduction pathways implicated in anoikis emanate from the focal adhesion complex in response to integrin-mediated cell adhesion to the extracellular matrix. Amongst these, phosphorylated FAK interacts with Grb2 and PI3-kinase [4042] to activate the Ras/MAP-kinase and Akt pathways, respectively [41, 43]. Our data emphasise the importance of the PI3-kinase/Akt pathway in oestrogen-responsive breast cancer as the rapid and dramatic loss of FAK phosphorylation within 15 minutes of detachment, followed by PARP cleavage after 1 hour, was associated with a decrease in phosphorylated Akt but an increase in phosphorylated MAP-kinases. Consistent with involvement of the intrinsic pathway, Bad phosphorylation decreased concomitantly with phosphorylated Akt and caspase 3 was activated. Dephosphorylated Bad forms heterodimers with Bcl-2 and Bcl-XL, thereby preventing them from interacting with and inhibiting Bax and Bak, which would otherwise form pores in the mitochondrial membrane to release cytochrome c and trigger cell death [38].
Our data show that breast cancer cell culture medium contains factors that confer anoikis resistance and that the major factor is a ligand that signals through the type I IGF receptor. A protective effect of IGF-1 against anoikis was demonstrated first in mouse embryonic fibroblasts engineered to overexpress the type I IGF receptor [22]. As far as we are aware, this is the first demonstration that IGFs protect breast cancer cells from caspase-dependent anoikis.
Many mechanisms are suggested to explain how malignant cells that detach from primary tumours evade cell death. These include changes to integrin expression [44], hypoxia which induces ligand-independent activation of growth factor receptors and redox-mediated decrease of pro-apoptotic factors [45], and EMT activation [46]. Our results demonstrate that oestrogen-responsive breast cancer cells avoid anoikis via a prosurvival pathway in which the type I IGF receptor is activated by IGFs. The type I IGF receptor is expressed widely on breast cancer cells and mediates the effects of IGFs on cell migration [13, 47] and proliferation [11, 12, 48]. IGF-1 activated both the PI3-kinase/Akt and Grb2/Ras/MAP-kinase pathways but our experiments with PI3-kinase, Akt and MEK inhibitors establish that anoikis resistance is conferred preferentially through the PI3-kinase/Akt pathway.
Although the Grb2/Ras/MAP-kinase pathway is not involved in the anoikic resistance of oestrogen-responsive breast cancer cells, its inhibition induces significant cell death which is abrogated almost completely by IGF-1. IGF-1 overcomes the cell death signal induced by inhibition of MEK1 and MEK2 without an increase in MAP-kinase phosphorylation. These results suggest that inhibition of the Grb2/Ras/MAP-kinase pathway is unlikely to be effective in the treatment of oestrogen-responsive breast cancer unless treatment is combined with inhibition of IGF signal transduction.
IGFs may signal through the type I IGF receptor, through isoform A and, to a lesser extent, through isoform B of the insulin receptor [9, 34, 49]. The receptor involved is of clinical importance because drugs such as figitumumab, cixutumumab, ganitumab and dalotuzumab are directed specifically against the type I IGF receptor [1619] whereas BMS-754807 and linisitinib inhibit both the type I IGF and insulin receptors [20, 21]. Our data indicate that signalling through the type I IGF receptor is dominant in oestrogen-responsive breast cancer.

Conclusions

We have established a reliable model of caspase-dependent anoikis for oestrogen-responsive breast cancer cells. We demonstrate the importance of the intrinsic pathway in anoikis and that IGF-1 can reinstate anoikis resistance of unattached oestrogen-responsive breast cancer cells cultured in serum-free medium. IGF-1 activated both the PI3-kinase/Akt and Grb2/Ras/MAP-kinase pathways but our experiments with PI3-kinase, Akt and MEK inhibitors show that anoikis resistance is conferred through the PI3-kinase/Akt pathway. Although ERK1 and ERK2 are not important in IGF-dependent anoikis-resistance, IGF-1 is able to circumvent apoptosis induced by inhibition of MEK1 and MEK2 without any increase in MAP-kinase phosphorylation. The IGF-1 signal is transduced via the type I IGF receptor and incubation with type I IGF receptor specific antibody, figitumumab, induces anoikis of cells grown in serum. This is the first demonstration of the importance of IGFs and the type I IGF receptor in the resistance of oestrogen-responsive breast cancer cells to caspase-dependent anoikis. The importance of the type I IGF receptor is supported by the association of higher receptor expression with earlier relapse with distant metastases in oestrogen receptor-positive tumours, especially those of women with more aggressive disease as assessed either by presence of involved lymph nodes or high histological tumour grade. Successful targeting of the type I IGF receptor would abrogate effectively the IGF signal for cell survival. Collectively, our results support the concept that effective combinations of targeted drugs should include abrogation of the activity of the IGF signal transduction pathway.

Methods

Cell culture

Breast cancer cell lines: MCF-7, T-47D, ZR-75 and EFM-19 were obtained from the American Type Culture Collection (Manassas, VA) or DMCSZ and cultured routinely in Dulbecco’s modified Eagle’s medium (DMEM) (Sigma, Poole, United Kingdom), supplemented with 10 % foetal calf serum (FCS) and 1 μgml−1 insulin in a humidified incubator at 37 °C with 5 % CO2.

Anoikis assay

The non-ionic acid poly(2-hydroxyethyl methacrylate) (poly-HEMA; SIGMA) which inhibits matrix deposition and cell attachment [26] was dissolved in 99 % ethanol at 10 mgml−1. Twelve-well tissue culture plates were coated twice with 0.5 ml poly-HEMA solution, allowed to dry, washed with phosphate-buffered saline (PBS) and stored at 4 °C.
Cells were trypsinised, 40 × 104 cells added to 35-mm-diameter poly-HEMA-coated wells and cultured in maintenance medium or in serum-free, phenol red-free DMEM in the absence or presence of IGF-1 for different lengths of time. Cells were recovered, centrifuged, and lysed for protein analysis. Cells were incubated without and with 20 μM PI3-kinase inhibitor, LY294002, (Sigma), 100 nM Akt inhibitor, GSK 690693, (SYN│thesis med chem Pty Ltd., Cambridge, United Kingdom) or 6 μM MEK 1 and MEK2 inhibitor, UO126, (SIgma) for 30 min prior to addition of IGF-1. The type I IGF receptor inhibitory antibody figitumumab [16] was from Pfizer Inc. (Tadworth, United Kingdom). Cells were centrifuged, and lysed for protein analysis.
Activation of caspase 3 was measured with the BD Pharmingen FITC-conjugated active caspase 3 apoptosis kit I (BD Biosciences, Oxford, United Kingdom). Cells were added to 16-mm-diameter poly-HEMA-coated wells at 40 × 104 cells/well in 2 ml of serum-free medium in the absence or presence of 50 ng/ml IGF-1 and incubated for 5 h. Cells were recovered, centrifuged, washed in PBS and, resuspended in 0.2 ml Cytofix/Cytoperm solution and incubated on ice for 20 min. Cells were then washed twice with 0.2 ml Perm/Wash solution, resuspended in 100 μl Perm/Wash solution with 8 μl and FITC-conjugated anti-activated caspase 3 antibody, protected from light and incubated for 30 min. Cells were rewashed, centrifuged and resuspended in 400–500 μl Perm/Wash solution. Fluorescence was measured with a BD FACScan flow cytometer. Excitation was at 488 nm and emission was measured at 530 ± 15 nm.

Knockdown of the type I IGF receptor

The sequence of the double-stranded short interfering RNAs (siRNA) designed to target the type I IGF receptor mRNA was 5’-CGACUAUCAGCAGCUGAAGTT-3’, and equivalent non-silencing scrambled sequence was 5’-UUCUCCGAACGUGUCACGUdTdT-3’ (Sigma). The siRNA duplex was mixed with RNA iMax (Invitrogen, Paisley, United Kingdom) in serum-free DMEM and incubated 30 min at room temperature. MCF-7 cells were trypsinised, resuspended in maintenance medium at a density of 25 × 104 cellsml−1, mixed with the transfection medium and added to a 25 cm2 flask. Cells were tested in anoikis assays or lysed for protein analysis, after incubation for 48 h or 96 h, respectively.

IGF-1 stimulated protein phosphorylation

Cells were added to 16-mm-diameter wells at a density of 15 × 104 cells/well for MCF-7 and ZR-75 cells, and 20 × 104 cells/well for EFM-19 cells. Cells were allowed to attach for 24-48 h and withdrawn from stimulatory factors in serum by culture for 48 h in growth factor-depleted medium, comprising phenol red-free DMEM and 10 % dextran-coated, charcoal-treated serum. Medium was changed daily. Cells were incubated with different concentrations of IGF-1 for 15 min, lysed and analysed by Western transfer [12].

Western transfer analysis

Cells were lysed in radioimmunoprecipitate (RIPA) buffer which comprised 50 mM Tris–HCl pH 7.5, 150 mM NaCl, 1 mM EDTA, 1 % NP-40 (v/v) and 0.25 % sodium deoxycholate (w/v, 1 μgml−1 pepstatin, 1 μgml−1 aprotinin, 1 μgml- 1 leupeptin, 2 mM sodium orthovanadate, 2 mM sodium fluoride and 2 mM phenyl methyl sulphonyl fluoride. Protein concentrations were measured with a bicinchonic acid assay (Thermo Scientific, Loughborough, United Kingdom). Equal amounts of protein were separated by electrophoresis on denaturing 12 % polyacrylamide gels and transferred to a Westran 0.45 μm nitrocellulose membrane (VWR, Leicestershire, United Kingdom) [50]. Membranes were incubated with specific antibodies against: cleaved poly-(ADP-ribose) polymerase (PARP) (#9541), type I IGF receptor (#3027), phosphorylated type I IGF receptor Tyr1135&1136 and insulin receptor Tyr1150&1151. (#3024), Akt (#9272), phosphorylated Akt Ser473 (#4060), FAK (#3285), phosphorylated FAK Tyr397 (#3283), ERK1 and ERK 2 (#9102), phosphorylated ERK 1 and ERK2 Thr 202 or Tyr 204 (#4370), Bad (#9292), phosphorylated Bad Ser136 (#4366), GSK3β (#9315), phosphorylated GSK3β Ser9 (#9323) (Cell Signaling Technologies, Hitchin, United Kingdom), IRS-1 (sc-7200), phosphorylated IRS-1 Tyr632 (sc-17196-R), and GAPDH (sc-25778) (Santa Cruz Biotechnology, Heidelberg, Germany). Membranes were incubated with horseradish peroxidase conjugated secondary antibodies followed by enhanced chemiluminescence with SuperSignal West Dura Substrate (Thermo Scientific) and exposure to SuperRX X-ray film (Fujifilm, Bedford, United Kingdom). The intensity of the protein bands was quantified by densitometry with Labworks 4.0 software (UVP Inc., Cambridge, United Kingdom).

Statistics

For the western transfer images, a representative example is shown. Data were normalized and expressed as a percentage of the maximum cleaved PARP or activated signal transduction protein detected. Results are expressed as means ± S.E.M. Differences between groups were tested by analysis of variance, paired or unpaired t-test; p < 0.05 was considered statistically significant. Experiments were replicated at least thrice. The association between type I IGF receptor expression and time to relapse with distant metastasis was analysed by the log rank test on data from The Cancer Genome Atlas.

Acknowledgements

We are grateful to Dr F. Shaheen for discussion about the inhibition of Akt and for his advice on the analysis of cleaved caspase 3 by FACS. We thank Professors AH Calvert and E Ruth Plummer for their valued support. We thank Pfizer Inc. for the gift of figitumumab. We thank Cancer Research UK for their generous support of our work, C0190/A8310 and C27826/A11524, and for the award of a Clinical Training Fellowship to BCL.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Competing interests

The authors declare no competing interest.

Authors’ contributions

BCL conceived and undertook experiments and analysed data. FEBM conceived experiments. Both authors were involved in preparation of the paper. Both authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat May FEB. Novel drugs that target the estrogen-related receptor alpha: their therapeutic potential in breast cancer. Cancer Manag Res. 2014;6:225–52.PubMedCentralCrossRefPubMed May FEB. Novel drugs that target the estrogen-related receptor alpha: their therapeutic potential in breast cancer. Cancer Manag Res. 2014;6:225–52.PubMedCentralCrossRefPubMed
3.
Zurück zum Zitat Zhan M, Zhao H, Han ZC. Signalling mechanisms of anoikis. Histol Histopathol. 2004;19:973–83.PubMed Zhan M, Zhao H, Han ZC. Signalling mechanisms of anoikis. Histol Histopathol. 2004;19:973–83.PubMed
5.
Zurück zum Zitat Nguyen DX, Bos PD, Massague J. Metastasis: from dissemination to organ-specific colonization. Nat Rev Cancer. 2009;9:274–U265.CrossRefPubMed Nguyen DX, Bos PD, Massague J. Metastasis: from dissemination to organ-specific colonization. Nat Rev Cancer. 2009;9:274–U265.CrossRefPubMed
6.
Zurück zum Zitat Miranti CK, Brugge JS. Sensing the environment: a historical perspective on integrin signal transduction. Nat Cell Biol. 2002;4:E83–90.CrossRefPubMed Miranti CK, Brugge JS. Sensing the environment: a historical perspective on integrin signal transduction. Nat Cell Biol. 2002;4:E83–90.CrossRefPubMed
7.
Zurück zum Zitat Cabodi S, Di Stefano P, Leal MDC, Tinnirello A, Bisaro B, Morello V, et al. Integrins and Signal Transduction, Integrins and Ion Channels. Molecular Complexes and Signaling. 2010;674:43–54. Cabodi S, Di Stefano P, Leal MDC, Tinnirello A, Bisaro B, Morello V, et al. Integrins and Signal Transduction, Integrins and Ion Channels. Molecular Complexes and Signaling. 2010;674:43–54.
8.
Zurück zum Zitat Hehlgans S, Haase M, Cordes N. Signalling via integrins: Implications for cell survival and anticancer strategies. Biochimica Et Biophysica Acta-Rev Cancer. 1775;2007:163–80. Hehlgans S, Haase M, Cordes N. Signalling via integrins: Implications for cell survival and anticancer strategies. Biochimica Et Biophysica Acta-Rev Cancer. 1775;2007:163–80.
9.
Zurück zum Zitat Westley RL, May FEB. A Twenty-First Century Cancer Epidemic Caused by Obesity: The Involvement of Insulin, Diabetes, and Insulin-Like Growth Factors. Int J Endocrinol. 2013;2013:632461.PubMedCentralCrossRefPubMed Westley RL, May FEB. A Twenty-First Century Cancer Epidemic Caused by Obesity: The Involvement of Insulin, Diabetes, and Insulin-Like Growth Factors. Int J Endocrinol. 2013;2013:632461.PubMedCentralCrossRefPubMed
11.
Zurück zum Zitat Stewart AJ, Johnson MD, May FEB, Westley BR. Role of insulin-like growth factors and the type I insulin-like growth factor receptor in the estrogen-stimulated proliferation of human breast cancer cells. J Biol Chem. 1990;265:21172–8.PubMed Stewart AJ, Johnson MD, May FEB, Westley BR. Role of insulin-like growth factors and the type I insulin-like growth factor receptor in the estrogen-stimulated proliferation of human breast cancer cells. J Biol Chem. 1990;265:21172–8.PubMed
12.
Zurück zum Zitat Davison Z, de Blacquiere GE, Westley BR, May FEB. Insulin-like growth factor-dependent proliferation and survival of triple-negative breast cancer cells: implications for therapy. Neoplasia. 2011;13:504–15.PubMedCentralCrossRefPubMed Davison Z, de Blacquiere GE, Westley BR, May FEB. Insulin-like growth factor-dependent proliferation and survival of triple-negative breast cancer cells: implications for therapy. Neoplasia. 2011;13:504–15.PubMedCentralCrossRefPubMed
13.
Zurück zum Zitat de Blaquiere GE, May FEB, Westley BR. Increased expression of both insulin receptor substrates 1 and 2 confers increased sensitivity to IGF-1 stimulated cell migration. Endocr Relat Cancer. 2009;16:635–47.CrossRefPubMed de Blaquiere GE, May FEB, Westley BR. Increased expression of both insulin receptor substrates 1 and 2 confers increased sensitivity to IGF-1 stimulated cell migration. Endocr Relat Cancer. 2009;16:635–47.CrossRefPubMed
14.
Zurück zum Zitat Sachdev D, Zhang X, Matise I, Gaillard-Kelly M, Yee D. The type I insulin-like growth factor receptor regulates cancer metastasis independently of primary tumor growth by promoting invasion and survival. Oncogene. 2010;29:251–62.PubMedCentralCrossRefPubMed Sachdev D, Zhang X, Matise I, Gaillard-Kelly M, Yee D. The type I insulin-like growth factor receptor regulates cancer metastasis independently of primary tumor growth by promoting invasion and survival. Oncogene. 2010;29:251–62.PubMedCentralCrossRefPubMed
15.
Zurück zum Zitat Gooch JL, Van Den Berg CL, Yee D. Insulin-like growth factor (IGF)-I rescues breast cancer cells from chemotherapy-induced cell death--proliferative and anti-apoptotic effects. Breast Cancer Res Treat. 1999;56:1–10.CrossRefPubMed Gooch JL, Van Den Berg CL, Yee D. Insulin-like growth factor (IGF)-I rescues breast cancer cells from chemotherapy-induced cell death--proliferative and anti-apoptotic effects. Breast Cancer Res Treat. 1999;56:1–10.CrossRefPubMed
16.
Zurück zum Zitat Cohen BD, Baker DA, Soderstrom C, Tkalcevic G, Rossi AM, Miller PE, et al. Combination therapy enhances the inhibition of tumor growth with the fully human anti-type 1 insulin-like growth factor receptor monoclonal antibody CP-751,871. Clin Cancer Res. 2005;11:2063–73.CrossRefPubMed Cohen BD, Baker DA, Soderstrom C, Tkalcevic G, Rossi AM, Miller PE, et al. Combination therapy enhances the inhibition of tumor growth with the fully human anti-type 1 insulin-like growth factor receptor monoclonal antibody CP-751,871. Clin Cancer Res. 2005;11:2063–73.CrossRefPubMed
17.
Zurück zum Zitat Rowinsky EK, Schwartz JD, Zojwalla N, Youssoufian H, Fox F, Pultar P, et al. Blockade of insulin-like growth factor type-1 receptor with cixutumumab (IMC-A12): a novel approach to treatment for multiple cancers. Curr Drug Targets. 2011;12:2016–33.CrossRefPubMed Rowinsky EK, Schwartz JD, Zojwalla N, Youssoufian H, Fox F, Pultar P, et al. Blockade of insulin-like growth factor type-1 receptor with cixutumumab (IMC-A12): a novel approach to treatment for multiple cancers. Curr Drug Targets. 2011;12:2016–33.CrossRefPubMed
18.
Zurück zum Zitat Tap WD, Demetri G, Barnette P, Desai J, Kavan P, Tozer R, et al. Phase II Study of Ganitumab, a Fully Human Anti-Type-1 Insulin-Like Growth Factor Receptor Antibody, in Patients With Metastatic Ewing Family Tumors or Desmoplastic Small Round Cell Tumors. J Clin Oncol. 2012;30:1849–56.CrossRefPubMed Tap WD, Demetri G, Barnette P, Desai J, Kavan P, Tozer R, et al. Phase II Study of Ganitumab, a Fully Human Anti-Type-1 Insulin-Like Growth Factor Receptor Antibody, in Patients With Metastatic Ewing Family Tumors or Desmoplastic Small Round Cell Tumors. J Clin Oncol. 2012;30:1849–56.CrossRefPubMed
19.
Zurück zum Zitat Scartozzi M, Bianconi M, Maccaroni E, Giampieri R, Berardi R, Cascinu S. Dalotuzumab, a recombinant humanized mAb targeted against IGFR1 for the treatment of cancer. Curr Opin Mol Ther. 2010;12:361–71.PubMed Scartozzi M, Bianconi M, Maccaroni E, Giampieri R, Berardi R, Cascinu S. Dalotuzumab, a recombinant humanized mAb targeted against IGFR1 for the treatment of cancer. Curr Opin Mol Ther. 2010;12:361–71.PubMed
20.
Zurück zum Zitat Carboni JM, Wittman M, Yang Z, Lee F, Greer A, Hurlburt W, et al. BMS-754807, a small molecule inhibitor of insulin-like growth factor-1R/IR. Mol Cancer Ther. 2009;8:3341–9.CrossRefPubMed Carboni JM, Wittman M, Yang Z, Lee F, Greer A, Hurlburt W, et al. BMS-754807, a small molecule inhibitor of insulin-like growth factor-1R/IR. Mol Cancer Ther. 2009;8:3341–9.CrossRefPubMed
21.
Zurück zum Zitat Mulvihill MJ, Cooke A, Rosenfeld-Franklin M, Buck E, Foreman K, Landfair D, et al. Discovery of OSI-906: a selective and orally efficacious dual inhibitor of the IGF-I receptor and insulin receptor. Future Med Chem. 2009;1:1153–71.CrossRefPubMed Mulvihill MJ, Cooke A, Rosenfeld-Franklin M, Buck E, Foreman K, Landfair D, et al. Discovery of OSI-906: a selective and orally efficacious dual inhibitor of the IGF-I receptor and insulin receptor. Future Med Chem. 2009;1:1153–71.CrossRefPubMed
22.
Zurück zum Zitat Valentinis B, Reiss K, Baserga R. Insulin-like growth factor-I-mediated survival from anoikis: Role of cell aggregation and focal adhesion kinase. J Cell Physiol. 1998;176:648–57.CrossRefPubMed Valentinis B, Reiss K, Baserga R. Insulin-like growth factor-I-mediated survival from anoikis: Role of cell aggregation and focal adhesion kinase. J Cell Physiol. 1998;176:648–57.CrossRefPubMed
23.
Zurück zum Zitat Irie HY, Shrestha Y, Selfors LM, Frye F, Iida N, Wang ZG, et al. PTK6 Regulates IGF-1-Induced Anchorage-Independent Survival. Plos One. 2010;5(7):e11729.PubMedCentralCrossRefPubMed Irie HY, Shrestha Y, Selfors LM, Frye F, Iida N, Wang ZG, et al. PTK6 Regulates IGF-1-Induced Anchorage-Independent Survival. Plos One. 2010;5(7):e11729.PubMedCentralCrossRefPubMed
24.
Zurück zum Zitat Li HL, Baldwin BR, Zahnow CA. LIP expression is regulated by IGF-1R signaling and participates in suppression of anoikis. Mol Cancer. 2011;10:100.PubMedCentralCrossRefPubMed Li HL, Baldwin BR, Zahnow CA. LIP expression is regulated by IGF-1R signaling and participates in suppression of anoikis. Mol Cancer. 2011;10:100.PubMedCentralCrossRefPubMed
25.
Zurück zum Zitat Ravid D, Maor S, Werner H, Liscovitch M. Caveolin-1 inhibits cell detachment-induced p53 activation and anoikis by upregulation of insulin-like growth factor-I receptors and signaling. Oncogene. 2005;24:1338–47.CrossRefPubMed Ravid D, Maor S, Werner H, Liscovitch M. Caveolin-1 inhibits cell detachment-induced p53 activation and anoikis by upregulation of insulin-like growth factor-I receptors and signaling. Oncogene. 2005;24:1338–47.CrossRefPubMed
26.
Zurück zum Zitat Frisch SM, Francis H. Disruption of Epithelial Cell-Matrix Interactions Induces Apoptosis. J Cell Biol. 1994;124:619–26.CrossRefPubMed Frisch SM, Francis H. Disruption of Epithelial Cell-Matrix Interactions Induces Apoptosis. J Cell Biol. 1994;124:619–26.CrossRefPubMed
27.
Zurück zum Zitat Dearth RK, Cui XJ, Kim HJ, Hadsell DL, Lee AV. Oncogenic transformation by the signaling adaptor proteins insulin receptor substrate (IRS)-1 and IRS-2. Cell Cycle. 2007;6:705–13.CrossRefPubMed Dearth RK, Cui XJ, Kim HJ, Hadsell DL, Lee AV. Oncogenic transformation by the signaling adaptor proteins insulin receptor substrate (IRS)-1 and IRS-2. Cell Cycle. 2007;6:705–13.CrossRefPubMed
28.
Zurück zum Zitat Vlahos CJ, Matter WF, Hui KY, Brown RF. A Specific Inhibitor of Phosphatidylnositol 3-Kinase, 2-(4-Morpholinyl)-8-Phenyl-4 h-1-Benzopyran-4-One (Ly294002). J Biol Chem. 1994;269:5241–8.PubMed Vlahos CJ, Matter WF, Hui KY, Brown RF. A Specific Inhibitor of Phosphatidylnositol 3-Kinase, 2-(4-Morpholinyl)-8-Phenyl-4 h-1-Benzopyran-4-One (Ly294002). J Biol Chem. 1994;269:5241–8.PubMed
29.
Zurück zum Zitat Walker EH, Pacold ME, Perisic O, Stephens L, Hawkins PT, Wymann MP, et al. Structural determinants of phosphoinositide 3-kinase inhibition by wortmannin, LY294002, quercetin, myricetin, and staurosporine. Mol Cell. 2000;6:909–19.CrossRefPubMed Walker EH, Pacold ME, Perisic O, Stephens L, Hawkins PT, Wymann MP, et al. Structural determinants of phosphoinositide 3-kinase inhibition by wortmannin, LY294002, quercetin, myricetin, and staurosporine. Mol Cell. 2000;6:909–19.CrossRefPubMed
30.
Zurück zum Zitat Heerding DA, Rhodes N, Leber JD, Clark TJ, Keenan RM, Lafrance LV, et al. Identification of 4-(2-(4-amino-1,2,5-oxadiazol-3-yl)-1-ethyl-7-{[(3S)-3-piperidinylmethyl]oxy}-1H-imidazo[4,5-c]pyridin-4-yl)-2-methyl-3-butyn-2-ol (GSK690693), a novel inhibitor of AKT kinase. J Med Chem. 2008;51:5663–79.CrossRefPubMed Heerding DA, Rhodes N, Leber JD, Clark TJ, Keenan RM, Lafrance LV, et al. Identification of 4-(2-(4-amino-1,2,5-oxadiazol-3-yl)-1-ethyl-7-{[(3S)-3-piperidinylmethyl]oxy}-1H-imidazo[4,5-c]pyridin-4-yl)-2-methyl-3-butyn-2-ol (GSK690693), a novel inhibitor of AKT kinase. J Med Chem. 2008;51:5663–79.CrossRefPubMed
31.
Zurück zum Zitat Rhodes N, Heerding DA, Duckett DR, Eberwein DJ, Knick VB, Lansing TJ, et al. Characterization of an Akt kinase inhibitor with potent pharmacodynamic and antitumor activity. Cancer Res. 2008;68:2366–74.CrossRefPubMed Rhodes N, Heerding DA, Duckett DR, Eberwein DJ, Knick VB, Lansing TJ, et al. Characterization of an Akt kinase inhibitor with potent pharmacodynamic and antitumor activity. Cancer Res. 2008;68:2366–74.CrossRefPubMed
32.
Zurück zum Zitat Favata MF, Horiuchi KY, Manos EJ, Daulerio AJ, Stradley DA, Feeser WS, et al. Identification of a novel inhibitor of mitogen-activated protein kinase kinase. J Biol Chem. 1998;273:18623–32.CrossRefPubMed Favata MF, Horiuchi KY, Manos EJ, Daulerio AJ, Stradley DA, Feeser WS, et al. Identification of a novel inhibitor of mitogen-activated protein kinase kinase. J Biol Chem. 1998;273:18623–32.CrossRefPubMed
33.
Zurück zum Zitat Duncia JV, Santella JB, Higley CA, Pitts WJ, Wityak J, Frietze WE, et al. MEK inhibitors: The chemistry and biological activity of U0126, its analogs, and cyclization products. Bioorg Med Chem Lett. 1998;8:2839–44.CrossRefPubMed Duncia JV, Santella JB, Higley CA, Pitts WJ, Wityak J, Frietze WE, et al. MEK inhibitors: The chemistry and biological activity of U0126, its analogs, and cyclization products. Bioorg Med Chem Lett. 1998;8:2839–44.CrossRefPubMed
34.
Zurück zum Zitat Soos MA, Field CE, Siddle K. Purified hybrid insulin/insulin-like growth factor-I receptors bind insulin-like growth factor-I, but not insulin, with high affinity. Biochem J. 1993;290(Pt 2):419–26.PubMedCentralCrossRefPubMed Soos MA, Field CE, Siddle K. Purified hybrid insulin/insulin-like growth factor-I receptors bind insulin-like growth factor-I, but not insulin, with high affinity. Biochem J. 1993;290(Pt 2):419–26.PubMedCentralCrossRefPubMed
35.
Zurück zum Zitat Peyrat JP, Bonneterre J, Vennin PH, Jammes H, Beuscart R, Hecquet B, et al. Insulin-Like Growth Factor-I Receptors (Igf1-R) and Igf1 in Human Breast-Tumors. J Steroid Biochem Mol Biol. 1990;37:823–7.CrossRefPubMed Peyrat JP, Bonneterre J, Vennin PH, Jammes H, Beuscart R, Hecquet B, et al. Insulin-Like Growth Factor-I Receptors (Igf1-R) and Igf1 in Human Breast-Tumors. J Steroid Biochem Mol Biol. 1990;37:823–7.CrossRefPubMed
36.
Zurück zum Zitat Papa V, Pezzino V, Costantino A, Belfiore A, Giuffrida D, Frittitta L, et al. Elevated insulin receptor content in human breast cancer. J Clin Invest. 1990;86:1503–10.PubMedCentralCrossRefPubMed Papa V, Pezzino V, Costantino A, Belfiore A, Giuffrida D, Frittitta L, et al. Elevated insulin receptor content in human breast cancer. J Clin Invest. 1990;86:1503–10.PubMedCentralCrossRefPubMed
37.
Zurück zum Zitat Tan K, Goldstein D, Crowe P, Yang JL. Uncovering a key to the process of metastasis in human cancers: a review of critical regulators of anoikis. J Cancer Res Clin Oncol. 2013;139:1795–805.CrossRefPubMed Tan K, Goldstein D, Crowe P, Yang JL. Uncovering a key to the process of metastasis in human cancers: a review of critical regulators of anoikis. J Cancer Res Clin Oncol. 2013;139:1795–805.CrossRefPubMed
38.
Zurück zum Zitat Paoli P, Giannoni E, Chiarugi P. Anoikis molecular pathways and its role in cancer progression. Biochimica Et Biophysica Acta-Molecular Cell Res. 1833;2013:3481–98. Paoli P, Giannoni E, Chiarugi P. Anoikis molecular pathways and its role in cancer progression. Biochimica Et Biophysica Acta-Molecular Cell Res. 1833;2013:3481–98.
39.
Zurück zum Zitat Eckert LB, Repasky GA, Ulku AS, McFall A, Zhou H, Sartor CI, et al. Involvement of ras activation in human breast cancer cell signaling, invasion, and anoikis. Cancer Res. 2004;64:4585–92.CrossRefPubMed Eckert LB, Repasky GA, Ulku AS, McFall A, Zhou H, Sartor CI, et al. Involvement of ras activation in human breast cancer cell signaling, invasion, and anoikis. Cancer Res. 2004;64:4585–92.CrossRefPubMed
40.
Zurück zum Zitat Schlaepfer DD, Broome MA, Hunter T. Fibronectin-stimulated signaling from a focal adhesion kinase-c-Src complex: Involvement of the Grb2, p130(cas), and Nck adaptor proteins. Mol Cell Biol. 1997;17:1702–13.PubMedCentralCrossRefPubMed Schlaepfer DD, Broome MA, Hunter T. Fibronectin-stimulated signaling from a focal adhesion kinase-c-Src complex: Involvement of the Grb2, p130(cas), and Nck adaptor proteins. Mol Cell Biol. 1997;17:1702–13.PubMedCentralCrossRefPubMed
41.
Zurück zum Zitat Xia H, Nho RS, Kahm J, Kleidon J, Henke CA. Focal adhesion kinase is upstream of phosphatidylinositol 3-kinase/Akt in regulating fibroblast survival in response to contraction of type I collagen matrices via a beta(1) integrin viability signaling pathway. J Biol Chem. 2004;279:33024–34.CrossRefPubMed Xia H, Nho RS, Kahm J, Kleidon J, Henke CA. Focal adhesion kinase is upstream of phosphatidylinositol 3-kinase/Akt in regulating fibroblast survival in response to contraction of type I collagen matrices via a beta(1) integrin viability signaling pathway. J Biol Chem. 2004;279:33024–34.CrossRefPubMed
42.
Zurück zum Zitat Chen HC, Appeddu PA, Isoda H, Guan JL. Phosphorylation of tyrosine 397 in focal adhesion kinase is required for binding phosphatidylinositol 3-kinase. J Biol Chem. 1996;271:26329–34.CrossRefPubMed Chen HC, Appeddu PA, Isoda H, Guan JL. Phosphorylation of tyrosine 397 in focal adhesion kinase is required for binding phosphatidylinositol 3-kinase. J Biol Chem. 1996;271:26329–34.CrossRefPubMed
43.
Zurück zum Zitat Schlaepfer DD, Hunter T. Focal adhesion kinase overexpression enhances Ras-dependent integrin signaling to ERK2/mitogen-activated protein kinase through interactions with and activation of c-Src. J Biol Chem. 1997;272:13189–95.CrossRefPubMed Schlaepfer DD, Hunter T. Focal adhesion kinase overexpression enhances Ras-dependent integrin signaling to ERK2/mitogen-activated protein kinase through interactions with and activation of c-Src. J Biol Chem. 1997;272:13189–95.CrossRefPubMed
44.
Zurück zum Zitat Mercurio AM, Bachelder RE, Chung J, O'Connor KL, Rabinovitz I, Shaw LM, et al. Integrin laminin receptors and breast carcinoma progression. J Mammary Gland Biol Neoplasia. 2001;6:299–309.CrossRefPubMed Mercurio AM, Bachelder RE, Chung J, O'Connor KL, Rabinovitz I, Shaw LM, et al. Integrin laminin receptors and breast carcinoma progression. J Mammary Gland Biol Neoplasia. 2001;6:299–309.CrossRefPubMed
46.
Zurück zum Zitat Thompson EW, Newgreen DF. Carcinoma invasion and metastasis: A role for epithelial-mesenchymal transition? Cancer Res. 2005;65:5991–5.CrossRefPubMed Thompson EW, Newgreen DF. Carcinoma invasion and metastasis: A role for epithelial-mesenchymal transition? Cancer Res. 2005;65:5991–5.CrossRefPubMed
47.
Zurück zum Zitat Doerr ME, Jones JI. The roles of integrins and extracellular matrix proteins in the insulin-like growth factor I-stimulated chemotaxis of human breast cancer cells. J Biol Chem. 1996;271:2443–7.CrossRefPubMed Doerr ME, Jones JI. The roles of integrins and extracellular matrix proteins in the insulin-like growth factor I-stimulated chemotaxis of human breast cancer cells. J Biol Chem. 1996;271:2443–7.CrossRefPubMed
48.
Zurück zum Zitat Daws MR, Westley BR, May FEB. Paradoxical effects of overexpression of the type I insulin-like growth factor (IGF) receptor on the responsiveness of human breast cancer cells to IGFs and estradiol. Endocrinology. 1996;137:1177–86.PubMed Daws MR, Westley BR, May FEB. Paradoxical effects of overexpression of the type I insulin-like growth factor (IGF) receptor on the responsiveness of human breast cancer cells to IGFs and estradiol. Endocrinology. 1996;137:1177–86.PubMed
49.
Zurück zum Zitat Pandini G, Frasca F, Mineo R, Sciacca L, Vigneri R, Belfiore A. Insulin/insulin-like growth factor I hybrid receptors have different biological characteristics depending on the insulin receptor isoform involved. J Biol Chem. 2002;277:39684–95.CrossRefPubMed Pandini G, Frasca F, Mineo R, Sciacca L, Vigneri R, Belfiore A. Insulin/insulin-like growth factor I hybrid receptors have different biological characteristics depending on the insulin receptor isoform involved. J Biol Chem. 2002;277:39684–95.CrossRefPubMed
50.
Zurück zum Zitat Molloy CA, May FEB, Westley BR. Insulin receptor substrate-1 expression is regulated by estrogen in the MCF-7 human breast cancer cell line. J Biol Chem. 2000;275:12565–71.CrossRefPubMed Molloy CA, May FEB, Westley BR. Insulin receptor substrate-1 expression is regulated by estrogen in the MCF-7 human breast cancer cell line. J Biol Chem. 2000;275:12565–71.CrossRefPubMed
Metadaten
Titel
Insulin-like growth factors are essential to prevent anoikis in oestrogen-responsive breast cancer cells: importance of the type I IGF receptor and PI3-kinase/Akt pathway
verfasst von
Brendan C. Luey
Felicity E. B. May
Publikationsdatum
01.12.2016
Verlag
BioMed Central
Erschienen in
Molecular Cancer / Ausgabe 1/2016
Elektronische ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-015-0482-2

Weitere Artikel der Ausgabe 1/2016

Molecular Cancer 1/2016 Zur Ausgabe

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

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