Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 May 31.
Published in final edited form as: Crit Rev Oncog. 2016;21(3-4):155–168. doi: 10.1615/CritRevOncog.2016016955

Anoikis and EMT: Lethal “Liaisons” during Cancer Progression

Zheng Cao 1, Theodore Livas 1, Natasha Kyprianou 1,*
PMCID: PMC5451151  NIHMSID: NIHMS849553  PMID: 27915969

Abstract

Anoikis is a unique mode of apoptotic cell death that occurs consequentially to insufficient cell–matrix interactions. Resistance to anoikis is a critical contributor to tumor invasion and metastasis. The phenomenon is regulated by integrins, which upon engagement with components of the extracellular matrix (ECM) form adhesion complexes and the actin cytoskeleton drives the formation of cell protrusions used to adhere to ECM, directing cell migration. The epithelial-mesenchymal transition (EMT) confers stem cell properties and leads to acquisition of a migratory and invasive phenotype by causing adherens junction breakdown and circumventing anoikis in the tumor microenvironment. The investigation of drug discovery platforms for apoptosis-driven therapeutics identified several novel agents with antitumor action via reversing resistance to anoikis, inhibiting survival pathways and impacting the EMT landscape in human cancer. In this review, we discuss current evidence on the contribution of the anoikis phenomenon functionally linked to EMT to cancer metastasis and the therapeutic value of antitumor drugs that selectively reverse anoikis resistance and/or EMT to impair tumor progression toward the development/optimization of apoptosis-driven therapeutic targeting of metastatic disease.

Keywords: anoikis, epithelial mesenchymal transition, prostate tumor, integrin, talin

I. INTRODUCTION

Anoikis was originally defined as a unique mode of apoptotic cell eath occurring upon insufficient cell-matrix interactions.1,2 The term is derived from the Greek “without a home” (Anoikos; becoming homeless) to describe the programmed death of epithelial and endothelial cells induced upon detachment from the extracellular matrix (ECM) consequential to insufficient cell-matrix interactions within the microenvironment. The ECM comprises a diverse network of cytokines impacting cell growth, motility, and angiogenesis, which can be made available for cellular use by enzymatic digestion and cytoskeletal remodeling.

The phenomenon of anoikis gained functional recognition and biological relevance in the cancer metastatic spread, as tumor epithelial cells upon release from the primary site and/or during dissemination in the circulation can undergo apoptosis and die prior to the metastatic journey. The development of resistance to anoikis and the mechanistic and phenotypic controls driving the process have been linked with tumor angiogenesis and metastasis1,35 because the mechanisms conferring resistance to this cell death enable the acquisition of migratory and invasive properties of tumor cells to establish metastatic lesions.6 The process of epithelial-mesenchymal transition (EMT) and consequential plasticity from cellular transdifferentiations due mesenchymal-epithelial transition (MET) critically contribute to tumor progression and to metastasis.6 The role of EMT in cancer takes central stage as tumor cells must physically detach from their primary tumor, invade the surrounding microenvironment, intravasate, endure the turbulence of circulation in the blood stream, and extravasate to secondary sites. The functional link between anoikis and EMT in dictating cancer cell survival outcomes has not been exploited.

Prostate cancer is a heterogeneous cancer with a natural history of progression from prostatic intraepithelial neoplasia to locally invasive androgen-dependent to androgen-independent metastatic disease, which is lethal.7,8 This malignancy is currently the second leading cause of cancer-related deaths among American men.9 Impairing the development of metastatic androgen-independent prostate tumors remains a major therapeutic endpoint for improving patient’s survival.10 Androgen-independent prostate tumors develop resistance to standard treatment modalities (such antiandrogen therapy and radiotherapy) due to apoptosis and anoikis evasion and acquisition of invasive and metastatic properties navigated by EMT.5,10,11 Interrogating the mechanisms underlying treatment failure in patients with metastatic prostate tumors is the most critical therapeutic endpoint for improving patient survival.11,12 The emergence of castration-resistant prostate cancer (CRPC) is due to the activation of survival pathways, including apoptosis suppression, anoikis (apoptosis upon detachment from the ECM) resistance, aberrant androgen signaling, and increased neovascularization.6,13,14 The molecular exploitation of apoptotic signaling pathways has fueled the development of effective pharmacotherapies for the treatment of advanced metastatic CRPC.15 This review discusses the current understanding of mechanisms underlying anoikis resistance in metastatic cancer, as well as efforts to overcome such resistance and enhance therapeutic response in patients with CRPC.

II. SIGNALING DEATH TO “HOMELESS” CELLS

A. The Players

The intrinsic pathway of apoptotic death is triggered by intracellular signals that maintain cellular integrity and homeostasis, such as DNA damage and endoplasmic reticulum stress.16 Upon death-signaling activation, the cytoplasmic proapoptotic proteins Bax and Bak are translocated to the outer mitochondrial membrane (OMM). Subsequent oligomerization of Bax and Bak generates a channel within the OMM, contributing to mitochondrial permeabilization and cytochrome C release. Cytochrome C results in formation of the apoptosome, constituting caspase-9, cytochrome C and the cofactor apoptosis protease–activating factor (Apaf), which activates the effector caspase-3 and apoptosis.1719

The proapoptotic BH3-only proteins play a critical role during the intrinsic pathway in anoikis.20 Bid and Bim, two members of this family, are activated by detachment of cells from ECM and facilitate the oligomerization of Bax-Bak within the OMM. The above members of the BH3-only protein family are indicated as activators.21 Specifically, sequestered Bim in the dynein cytoskeletal complexes is released until cells detach from the ECM and are subsequently translocated to the mitochondria.22 Loss of the cell-ECM interaction also leads to Bim accumulation, by inhibiting its proteasome degradation mediated by an ERK (extracellular signal-regulated kinases) and phosphatidylinositol-3-kinase and protein kinase B– (PI3K/Akt-) mediated phosphorylation of Bim, in the context of integrin engagement.22,23 Another group of BH3-only proteins are indicated as sensitizers, including Bad, Bik, Bmf, Noxa, and Puma. The sensitizer BH3–only proteins are incapable of promoting Bax and Bak oligomerization, but result in cell death by preventing activation of the antiapoptotic functions of Bcl-2.2427 Indeed, Bcl-2 is the pivotal antiapoptotic family member which bypasses mitochondrial dysfunction and circumvents apoptosis by regulating Bak/Bax apoptotic members to prevent their clustering into pores, and by sequestering Bid and Bim activator members of the BH3-only proteins to prevent Bak/Bax oligomerization.27,28 Furthermore, other members of the BH3-only family are mechanistically involved in anoikis induction of different cell lines.2931 For instance, Noxa and Puma, following transcriptional regulation by p53, have been shown to functionally contribute to fibroblast anoikis.2931 In addition, in epithelial cells Bmf is capable of recording damage at the actin cytoskeleton and carrying on death signals towards cellular execution. Specifically, loss of the cell-ECM interaction leads to the release of Bmf from the myosin V motor complex with subsequent accumulation in the mitochondria and neutralization of Bcl-2 antiapoptotic action, inducing anoikis.30,31

B. Extrinsic versus Intrinsic Forces of Death

The death receptor pathway is characteristically responsible for driving anoikis in both normal and malignant epithelial cells. Binding of the death ligands, including FasL or TRAIL, at the extracellular domain of the death receptor activates the pathway by causing oligomerization of the receptors.10 Ligand binding contributes to further binding of caspase-8 to the death effector domain (DED) of the Fas-associated death domain (FADD) and mediates the production of a death-inducing signaling complex (DISC). DISC further stimulates caspase-8 dimerization, which is cleaved to become active. The activated caspase-8 is released to the cytoplasm to activate downstream caspase-3 and -7, triggering apoptosis.10,32

FLICE-inhibitory protein (FLIP) is the primary endogenous inhibitor of the death receptor pathway. This effector constitutes two DEDs and a C-terminal caspase–8–like domain with higher affinity to DISC. As such, FLIP is a more potent competitive inhibitor of procaspase-8, and thus blocks binding of caspase-8, preventing subsequent recruitments and activation.33 In normal conditions, loss of cell contact with ECM contributes to upregulation of Fas and FasL and downregulation of FLIP expression. The above changes upon loss of cell-ECM interaction facilitate downstream caspase-8 activation in a FADD-dependent manner, leading to apoptosis.10 However, in malignant cells loss of the cell-ECM interaction fails to activate death receptor-induced caspase–8 despite the upregulation of Fas and FasL, ultimately leading to anoikis resistance. Failure of cancer cells to downregulate FLIP after detachment may be responsible for the above effect.32 Studies have been performed that target FLIP in anoikis-resistant cells to generate a novel therapeutic approach to treating metastatic prostate cancer.34 The involvement of caspases as an absolute requirement for cells in their “death row” has been challenged by the identification of caspase-independent pathways of apoptosis.35 Bit1, a mitochondrial protein, translocates to the cytoplasm upon disruption of integrin-ECM interaction.36 Through a cell death domain,35 Bit1 induces apoptosis upon translocation to the cytoplasm in cells losing the integrin-ECM interaction.37,38 A decrease in Bit1 expression in the cytoplasm facilitates tumor progression in patients with invasive ductal carcinoma.39 Mechanistically, cytoplasmic Bit1 facilitates cell death by reducing the activity of nucleus transducin–like enhancer of Split 1 (TLE1), a member of the Groucho/TLE/Grg protein family that mediates DNA-binding proteins to regulate transcription.40,41 Upon nuclear translocation, TLE1 heterooligomerizes with the amino-terminal enhancer of Split (AES), another member of the Groucho/TLE/Grg family.42 In cancer cells losing the integrin-ECM interaction, the AES/TLE1 heterooligomers are translocated from the nucleus to the cytoplasm following translocation of Bit1 into the cytoplasm.43 The subsequent downregulation of nuclear TLE1 expression activates the death signaling pathway (Fig. 1).44

FIG. 1.

FIG. 1

Anoikis signaling pathway in cancer cells. Cell detachment-induced anoikis signaling can be activated via three pathways: (1) the extrinsic death receptor-mediated apoptotic pathway, (2) the intrinsic apoptotic pathway, and (3) the caspase-independent pathway. In the first pathway, activation of Fas/FADD leads to cleavage of caspase-8 and subsequently activates downstream caspase-3 to facilitate apoptosis. In the second pathway, Cell-ECM interaction mediates integrin-induced activation of Talin1, FAK/Src and ILK-1. ILK-1 activation leads to phosphorylation of GSK3β and subsequent recruitment of Snail, which ultimately activates the PI3-K/Akt cell survival pathway. The loss of cell-ECM interaction contributes to upregulated proapoptotic proteins, such as Bim and downregulated antiapoptotic regulatory proteins, such as Bcl-2. The above changes activate cytochrome C release from the mitochondria, which subsequently activates caspase-9 and -3 to induce apoptosis. In the third pathway, upon cancer cell loss of the ECM interaction, the AES/TLE1 heterooligomers are translocated from the nucleus to the cytoplasm; subsequent downregulation of TLE1 by the proteasome results in the activation of the death signaling pathway.

C. Resistance to Anoikis Navigates Metastatic Journey

Cancer cells are capable of promoting anoikis resistance mainly through the regulation of integrins and the initiation of EMT. Integrins are essential for attachment of epithelial and endothelial cells to the ECM by recognizing fibronectin and laminin, the major adhesive ECM components.45,46 Integrin-mediated cell-ECM interactions are functionally involved in regulating tumor angiogenic response during cancer progression to metastatic disease.4648 Integrins regulate signal transduction from the extracellular space to the intracellular network using integrin-activated signaling molecules, including the focal adhesion kinase (FAK), PI3-K, and members of the ERK1/2 mitogen-activated protein (MAP) kinase family.4749 The loss of integrin-mediated epithelial cell-ECM interactions decreases phosphorylation of downstream effectors such as FAK, PI3-K, ERK1, and MAP kinase, thus mediating cell susceptibility to anoikis. The integrins β1, β3, and β6 are upregulated during prostate cancer progression to metastatic disease.48 Knockdown of β1 integrin prevents anoikis resistance in PC3-MM2 cells, a highly metastatic cell line derived from PC3 cells passaged in mice.50 Noteworthy, there is an association between increased β1 integrin activation and consequential resistance to anoikis with prostate cancer progression and metastatic spread has been observed.50

Mechanistically, taking over dynamic control of the focal-adhesion complex formation, β1-integrin mediates the activity of Src, the oncogenic Rous sarcoma virus.51 Cell adhesion functionally activates Src by ECM-integrin–mediated signals through FAK, while the loss of cell-ECM interaction leads to inactivation of Src, which contributes to anoikis.52,53 Activation of Src is associated with anoikis resistance in various cancers such as osteosarcoma, pancreatic carcinoma, and prostate cancer.5255 Src is widely expressed in prostate cancer and has been studied as a potential therapeutic target to decrease prostate cancer metastases to the lymph nodes.56

The integrin-linked kinase (ILK), a serine/threonine protein kinase, interacts with the cytoplasmic domain of β1- and β3-integrin, and is functionally involved in integrin and Wnt signaling pathways.5759 In prostate cancer, ILK expression is associated with tumor progression and the increased proliferative index during progression; it is inversely correlated with 5-year patient survival.60 ILK is essential in several integrin-involved cellular processes, including anchorage-dependent cell growth, cell adhesion, and fibronectin-ECM assembly.57,61,62 Cell-ECM adhesion contributes to ILK activation and directs phosphorylation of AKT Ser473 and glycogen synthase kinase-3 (GSK3); inhibition of ILK interrupts AKT phosphorylation and reduces cell survival.61,63 Interestingly, ILK-1 contributes to anoikis resistance by inhibiting apoptosis signals, such as initiator caspase-8 and executioner caspase-3, even in the absence of integrin/ECM signal activation, but possibly via a parvin-mediated targeting of AKT signaling to lipid rafts.6466

III. EMT LANDSCAPE IN CONTROL OF CELL FATE

EMT is a physiological process in which epithelial cells remodel cytoskeleton and cause the release of linkage with adjacent cells to generate a motile phenotype.67 Reawakening this program during pathophysiologic conditions confers invasive and migratory properties to epithelia-derived cancer cells via coordinated molecular and genetic alterations. Dramatic phenotypic alterations transform an adherent epithelium into elongated, mesenchymal-like cells that are capable of evading normal tissue architectural constraints, allowing for their escape from the primary tumor mass. Mesenchymal-epithelial transition (MET) transdifferentiations have been implicated in the final stages of metastatic colonization to revert cells back to an epithelial-like phenotype upon settlement in distant tissues.67 In addition, EMT allows cancer cells to detach from neighboring cells, to overcome anoikis, and to move from their primary location to metastatic sites.10 Cell polarity and epithelial integrity are maintained by tight junctions through claudins and occludins with one side attached to the ECM and adjacent cells.

A landscape of dramatic phenotypic alterations transforms an adherent epithelium into mesenchymal-like cells capable of evading normal-tissue architectural constraints, allowing their escape from the primary tumors.6870 During EMT, cell-cell adhesion molecules, such as E-cadherin and γ-catenin, are downregulated, with a parallel upregulation of mesenchymal markers, such as α-smooth muscle actin (SMA), fibronectin, N-cadherin, and vimentin.68 Acquisition of the mesenchymal phenotype is functionally associated with the ability to overcome anoikis.69 Growing evidence links EMT induction to apoptosis evasion via upregulation of Bcl-2 antiapoptotic proteins and/or activation of the PI3-K/Akt prosurvival pathways, often associated with the downregulation of proapoptotic proteins such as Bim, Bax, Noxa, p21, and p53 effector related to pmp22Y.7072

The induction of phenotypic EMT is regulated by key transcription factors, including Snail, Twist, and ZEB1/2. Specifically, Twist activation contributes to EMT by facilitating migration and invasion, and upregulates the antiapoptotic protein Bcl-2 to promote anoikis resistance.73,74 Transcriptional activation of Snail-1, an E-cadherin repressor, is also involved in EMT induction.75 In addition to E-cadherin, Snail can directly suppress the transcription of genes involved in anoikis, such as PTEN, downregulation of which activates the PI3-K/Akt pathway and inactivates the proapoptotic protein Bad, thus contributing to anoikis resistance.76 Moreover, downregulation of E-cadherin during EMT facilitates the accumulation of free β-catenin in the cytosol and subsequent nuclear translocation, which induces genes involved in the regulation of cell migration, invasion, and cell cycle progression, such as c-Myc, cyclin D1, c-Jun, and MMP-1.77 Overexpression of β-catenin and its consistent presence in the cytosol leads to anoikis resistance via regulation of c-Myc, cyclin D1, and MAPK-mediated survival pathways and repression of epithelial genes toward sustaining a mesenchymal phenotype among cancer cell populations.78 The ZEB1 transcription factor facilitates EMT7981 via upregulation of vimentin and downregulation of E-cadherin and semaphorin 3F, resulting in the activation of Akt survival pathway and anoikis resistance.81

Therapeutic failure and subsequent disease progression to CRPC has been attributed to recurrent androgen receptor (AR) activity and the inability to completely suppress AR-mediated signaling.8285 Rapidly growing evidence has defined diverse mechanisms responsible for AR signaling activation in metastatic CRPC, including AR amplification resulting in hypersensitivity86; AR mutations and splice variants which reduce the ligand specificity required for receptor activation8789; alterations of AR coregulators90; ligand-independent AR activation mediated by cross-talk with different signaling networks83; and sustained androgen exposure or intratumoral androgen synthesis.91

Work in our laboratory suggests that modulating androgen exposure to androgen-independent prostate cancer cells contributes to EMT-MET cycling, actin cytoskeleton remodeling, and microtubule rearrangement.92 Furthermore, increasing androgen presence is associated with an augmented EMT phenotype in androgen-independent prostate cancer cells.92 Interestingly, the controversy surrounding the impact of androgen deprivation therapy (ADT) on the EMT landscape in normal mouse prostate tissue and preclinical models of LuCaP35 prostate tumor xenografts93 is propagated in the clinical setting of human disease, where EMT signature profiles have been found in prostate tumors after ADT.94,95 Evidence from independent studies collectively supports elevation of EMT-related markers, including N-cadherin,96 cadherin-11,97,98 and nestin,99 in human prostate cancer specimens after ADT. Considering the transient nature of the EMT-MET cycling dynamic, the therapeutic window, relevant endpoints of treatment, and the EMT signature assessment might require coordination in capturing the temporal effect of androgen deprivation on prostate tumor epithelial cells prior to apoptosis induction among tumor subpopulations.

The canonical EMT signature driving the phenotypic process is programmed by a complex of gene expression changes. Epithelial markers include adherens and tight junction proteins (E-cadherin, ZO-1), and mesenchymal markers include the intermediate filament vimentin.100,101 Loss of cell polarity and gain of migration properties ensue. Genome-wide analysis of gene expression to identify primary targets of DZ-50 (an anticancer drug developed by our group14) identified the downregulation of two EMT effector genes, Snail and insulin growth factor binding protein 3 (IGFBP3), as well as integrin-α6, talin, and thrombospondin, all of which regulate cell-ECM adhesion and angiogenesis; and claudin-11 and -14, which are transmembrane proteins involved in tight junction (TJ) strands formation.14 Selective targeting of claudin-11 during anoikis is significant as this TJ protein interacts with the actin cytoskeleton.102104 The focal adhesion effector, FAK, links AKT survival signaling to EMT via Snail activation.105 IGFBP3, a secretory glycoprotein and IGF signaling regulator that promotes transforming growth factor beta– (TGFβ-) mediated EMT and confers antitumor resistance, is downregulated during anoikis induction by DZ-50.14,102 A large body of evidence supports the functional coupling of phenotypic EMT-MET cycling to anoikis signaling via diverse mechanisms involving the following: (1) FAK regulation of Snail induction via AKT survival signaling;105 (2) coordinated growth factor signaling network suppression of anoikis via proapoptotic proteins;106 (3) recruitment of FAK and paxillin to β1 integrin–promoting cancer cell migration via MAPK activation;107 (4) combined activation of FAK and AKT survival signaling101; and (5) regulators of fibronectin-ECM assembly through EMT transcription factors that dictate anoikis outcomes.57,64,106,108,109

IV. REMODELING THE ACTIN CYTOSKELETON

The actin cytoskeleton is an essential scaffold for integrating membrane and intracellular functions. Growth stimulation promotes the actin polymerization assembly to generate movement, whereas apoptosis elicits cytoskeletal reorganization changes.110,111 The regulation of anoikis outcomes in epithelial cells by the actin cytoskeleton is supported by compelling data on the involvement of key regulators of cytoskeleton organization in cell motility. Thus, the functional loss of cofilin, the main actin regulator, significantly decreases prostate cancer cell adhesion112 while the mutation at the S3A of cofilin compromises cell migration and filopodia formation due to enhanced F-actin filament polymerization.112 Another effector, Ankyrin G, has been shown to functionally link EMT to the cytoskeleton via its localization to AJ and interaction with E-cadherin.113 Moreover, overexpression of LIM kinase-2 (LIMK2), which regulates cofilin phosphorylation and actin dynamics, is associated with invasive prostate cancer114; and claudin-11, a TJ transmembrane protein that interacts with the actin cytoskeleton, is targeted by the anoikis-inducing agent DZ-50.14,102 Taken together, these data support the targeting of interactions between actin regulators and tight junctions responsible for resistance to anoikis to overcome therapeutic failure.

Talin1 is an actin-binding protein that links integrins to the actin cytoskeleton in focal adhesion complexes.115 The integrity of ECM-fibronectin sustains high talin levels, thus conferring resistance to DZ-50-–induced anoikis, evidence that can be of therapeutic significance in targeting of talin in advanced prostate tumors to lift resistance to anoikis.14 This gains indirect support from clinical evidence on the correlation between elevated talin1 and human prostate cancer progression to metastasis.5 The possibility of simultaneous induction of both apoptosis and anoikis, effectively enhancing the therapeutic response to antitumor agents by massive cell killing, gains support from clinical evidence in human breast cancer. During tumor progression, promotion of metastasis is exerted by Bcl-xL, a suppressor of cytoskeleton-dependent apoptosis and enhancer of metastasis but not primary tumor growth.110

In a complex signaling repertoire, talin1 binding to β integrin recruits the focal adhesion partners ILK, FAK, and Src, and activates the downstream signals PI3-K/AKT and ERK, preventing anoikis and promoting cell invasion.14,116,117 Specifically, talin1 S425 phosphorylation, essential in β1 integrin activation, correlates with metastatic properties of prostate cancer cells. Nonphosphorylatable talin1 (S425A) expression in talin1-silenced PC3-MM2 and C4-2B4 prostate cancer cells leads to integrin-mediated adhesion, motility, and increased susceptibility of cells to anoikis, all of which are consequential effects of decreased activation of β1 integrins.118 Work in our laboratory has established that talin’s contribution to anoikis resistance can be overcome by DZ-50, with potential therapeutic targeting value in metastatic prostate cancer.5,10,14 The mechanistic dissection of DZ-50 gene targets in prostate cancer cells reveals key receptor kinases, including LIMK2, GSK3β, AKT, and MAP4K5, that are involved in anoikis regulation. LIM kinases regulate cofilin phosphorylation and actin cytoskeletal dynamics by phosphorylating its Ser3 residue and inactivating its depolymerization activity.8 Actin polymerization regulated by cofilin activation is a convergence point in intracellular signaling through which extracellular stimuli impact the actin cytoskeleton, cell invasion, and apoptosis.119121 In a signaling cascade via a TGF–β–directed mechanism, constitutively active cofilin (S3A mutation) promotes filopodia formation and cell invasion and directs the EMT landscape in prostate cancer cells.112 In clinical specimens of human prostate cancer, there is significant overexpression of cofilin in metastatic compared to primary prostate tumors.112 Considering that the phosphorylation status/activity of cofilin can dictate interactions of the actin cytoskeleton with ECM effectors, enabling acquisition of metastatic properties,59 targeting of cofilin can lift anoikis resistance in metastatic cells with potential therapeutic significance.

V. THERAPEUTIC SIGNIFICANCE OF OVERCOMING ANOIKIS RESISTANCE

A. Quinazoline Derivatives

Our own drug discovery efforts focusing on the structural optimization of a quinazoline-based α1-adrenoceptor antagonist, Doxazosin, led to the generation of a lead derivative, DZ-50. DZ-50 impaired prostate tumor growth and metastasis via anoikis induction and antiangiogenesis in prostate cancer cell lines and human prostate cancer xenografts in nude mice.14,122 The results of a genome-wide microarray analysis in human prostate cancer DU-145 cells to identify the primary molecular targets of DZ-50 indicates that DZ-50 downregulates genes encoding regulators of ECM (fibronectin and integrin-α6), tight junctions (claudin-11), angiogenesis (thrombospondin), and the critical effector of the IGF axis, IGFBP3. IGFBP3 induction is essential for prostate stroma remodeling representing an attractive therapeutic target by the novel quinazoline DZ-50.14,123

B. Src Inhibitors

The Src family of kinases has recently been evaluated as a potential therapeutic target to develop treatment for a variety of cancers. Dasatinib, a Src family tyrosine kinase inhibitor, at low concentrations, represses prostate cancer cell adhesion, migration, and invasion.124 Dasatinib effectively suppresses both tumor growth and development of lymph node metastases in both androgen-sensitive and androgen-resistant prostate tumors in preclinical models.56 Significantly enough, an anoikis-resistant and spheroidogenic intermediate mesenchymal state phenotype of SKOV3 ovarian cancer cells was reversed by the Src kinase inhibitor AZD0530 via increasing E-cadherin promoter activity and E-cadherin expression levels in both in vitro and in vivo preclinical studies.125 The Src inhibitor, Dasatinib, clinically approved for patients with chronic myelogenous leukemia, demonstrated a limited therapeutic response in patients with metastatic CRPC.126 A single patient, however, exhibited a significant therapeutic response, indirectly supporting further molecular profiling to target selective patient populations.126 The clinical evidence, in terms of the combination of such an anoikis-inducing agent with Docetaxel chemotherapy, failed to provide an additional therapeutic benefit in metastatic CRPC.127 Bosutinib, another Src inhibitor, in combination with ABT-263, a Bcl-2 inhibitor, effectively induces anoikis in lung adenocarcinoma and in advanced leukemia resistant to kinase inhibitors.128,129

C. Trks Inhibitors

Tropomyosin-related kinases (Trks), a family of receptor tyrosine kinases (RTK) activated by neurotrophins, are involved in tumor cell growth and survival signaling.130 The pan-Trk inhibitors have been evaluated in tumor xenograft and transplantation models using neuroblastoma, medulloblastoma, and prostatic and pancreatic cancer cell lines.10 Furthermore, a Trk inhibitor, K252a, attenuates brain-derived neurotrophic factor–induced migration and proliferation of nasopharyngeal carcinoma cells by exerting potent anoikis sensitization.130 Trk inhibitors can be used as anoikis-sensitizing agents in diverse human malignancies with a promise of overcoming therapeutic resistance and stopping lethal disease. MGCD516, an RTK inhibitor with Trk inhibitory activity, is being evaluated in a Phase I clinical trial in patients with clear cell renal cell carcinoma resistant to angiogenesis inhibitors and in CRPC patients with advanced bone metastasis (Clinicaltrials.gov).

D. c-Met/VEGFR-2 Inhibitors

c-Met and VEGFR-2 (vascular endothelial growth factor receptor-2), both functionally critical players in cancer initiation and progression to metastasis,131 can be therapeutically targeted by exploitation of anoikis. Indeed, inhibition of c-Met and VEGFR-2 with foretinib reduces ovarian cancer cell adhesion, migration, and invasion via anoikis induction, impairing ovarian tumor growth.132 Considering the evidence discussed so far, one may easily argue that anoikis potentially drives the clinical antitumor action of foretinib in advanced renal cell and gastrointestinal carcinoma.132

E. Metformin

Of direct clinical significance is recent evidence that the antidiabetic agent metformin exhibits anoikis resistance–lifting properties in preclinical models of human cancer. In medullary thyroid cancer (MTC), it inhibits mammalian target of rapamycin (mTOR)/6SK and pERK survival signaling,133 and sensitizes MTC cells to anoikis.133 In clinical trials with prostate cancer patients, metformin significantly reduces biochemical recurrence134 and stabilizes disease in chemotherapy-naive CRPC patients,135 implicating anoikis-mediated therapeutic benefits in patients post-ADT.136

VI. SUMMARY AND FUTURE DIRECTIONS

Both anoikis resistance and EMT reversal to MET have been implicated in the development, progression, and therapeutic failure of different tumors. Considering the transient nature that characterizes both phenomena—anoikis and EMT-MET cycling—targeting these phenotypic processes during a defined therapeutic window during mesenchymal transition—immediately upon disruption of the tumor epithelial cell-EMC interaction and prior to initiation of metastatic dissemination—calls for personalized treatment of cancer patients. Coordinated activation of apoptosis and anoikis signaling mechanisms allow for the combination targeting of primary tumor growth and impairing metastatic progression, depending on the EMT landscape of individual tumors.

Acknowledgments

This work was supported by an American Urological Association-Urology Care Research Fellowship (ZC), the James F. Hardymon Endowment to the Department of Urology, University of Kentucky, and NIH/NIDDK grant no. 2 R01 GRANT00491815.

ABBREVIATIONS

AR

androgen receptor

DED

death effector domain

DISC

death-inducing signaling complex

ECM

extracellular matrix

EGFR

epidermal growth factor receptor

EMT

epithelial-mesenchymal transition

ERK

extracellular signalregulated kinase

FADD

Fas-associated death domain

FAK

focal adhesion kinase

FLIP

FLICE inhibitory protein

GSK-3

glycogen synthase kinase-3

ILK

integrin-linked kinase

MAPK

mitogen-activated protein kinase

MET

mesenchymal-epithelial transition

MMP

metalloproteinase

OMM

outer mitochondrial membrane

PI3K

phosphoinositide-3-OH kinase

PPAR-γ

peroxisome proliferator-activated receptor-gamma

PTEN

phosphatase and tensin homolog deleted on chromosome 10

RTKs

receptor tyrosine kinases

SMA

α-smooth muscle actin

TGFβ

transforming growth factor beta

Trks

tropomyosin-related kinases

VEGFR

vascular endothelial growth factor receptor

References

  • 1.Frisch SM, Ruoslahti E. Integrins and anoikis. Curr Opin Cell Biol. 1997;9(5):701–6. doi: 10.1016/s0955-0674(97)80124-x. [DOI] [PubMed] [Google Scholar]
  • 2.Frisch SM, Screaton RA. Anoikis mechanisms. Curr Opin Cell Biol. 2001;13(5):555–62. doi: 10.1016/s0955-0674(00)00251-9. [DOI] [PubMed] [Google Scholar]
  • 3.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. 2013;139(11):1795–805. doi: 10.1007/s00432-013-1482-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Pencheva N, Tavazoie SF. Control of metastatic progression by microRNA regulatory networks. Nat Cell Biol. 2013;15(6):546–54. doi: 10.1038/ncb2769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Sakamoto S, McCann RO, Dhir R, Kyprianou N. Talin1 promotes tumor invasion and metastasis via focal adhesion signaling and anoikis resistance. Cancer Res. 2010;70(5):1885–95. doi: 10.1158/0008-5472.CAN-09-2833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Rennebeck G, Martelli M, Kyprianou N. Anoikis and survival connections in the tumor microenvironment: is there a role in prostate cancer metastasis? Cancer Res. 2005;65(24):11230–5. doi: 10.1158/0008-5472.CAN-05-2763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Debes JD, Tindall DJ. Mechanisms of androgen-refractory prostate cancer. N Engl J Med. 2004;351(15):1488–90. doi: 10.1056/NEJMp048178. [DOI] [PubMed] [Google Scholar]
  • 8.Wang X, Yin L, Rao P, Stein R, Harsch KM, Lee Z, Heston WD. Targeted treatment of prostate cancer. J Cell Biochem. 2007;102(3):571–9. doi: 10.1002/jcb.21491. [DOI] [PubMed] [Google Scholar]
  • 9.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin. 2015;65(1):5–29. doi: 10.3322/caac.21254. [DOI] [PubMed] [Google Scholar]
  • 10.Sakamoto S, Kyprianou N. Targeting anoikis resistance in prostate cancer metastasis. Mol Aspects Med. 2010;31(2):205–14. doi: 10.1016/j.mam.2010.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.McKenzie S, Kyprianou N. Apoptosis evasion: the role of survival pathways in prostate cancer progression and therapeutic resistance. J Cell Biochem. 2006;97(1):18–32. doi: 10.1002/jcb.20634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Feldman BJ, Feldman D. The development of androgen-independent prostate cancer. Nat Rev Cancer. 2001;1(1):34–45. doi: 10.1038/35094009. [DOI] [PubMed] [Google Scholar]
  • 13.Frisch SM, Francis H. Disruption of epithelial cell-matrix interactions induces apoptosis. J Cell Biol. 1994;124(4):619–26. doi: 10.1083/jcb.124.4.619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Hensley PJ, Desiniotis A, Wang C, Stromberg A, Chen CS, Kyprianou N. Novel pharmacologic targeting of tight junctions and focal adhesions in prostate cancer cells. PloS One. 2014;9(1):e86238. doi: 10.1371/journal.pone.0086238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Siegelin MD, Fimognari C, Kumar NB, Dou QP, Yang H, Samadi AK, Russo GL, Spagnuolo C, Ray SK, Chakrabarti M, Morre JD, Coley HM, Honoki K, Fujii H, Georgakilas AG, Amedei A, Niccolai E, Amin A, Ashraf SS, Helferich WG, Yang X, Boosani CS, Guha G, Bhakta D, Ciriolo MR, Aquilano K, Chen S, Mohammed SI, Keith WN, Bilsland A, Halicka D, Nowsheen S, Azmi AS. Broad targeting of resistance to apoptosis in cancer. Semin Cancer Biol. 2015;35(Suppl):S78–S103. doi: 10.1016/j.semcancer.2015.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Kroemer G, Galluzzi L, Brenner C. Mitochondrial membrane permeabilization in cell death. Physiol Rev. 2007;87(1):99–163. doi: 10.1152/physrev.00013.2006. [DOI] [PubMed] [Google Scholar]
  • 17.Cohen GM. Caspases: the executioners of apoptosis. Biochem J. 1997;326(Pt 1):1–16. doi: 10.1042/bj3260001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Thornberry NA. Caspases: key mediators of apoptosis. Chem Biol. 1998;5(5):R97–103. doi: 10.1016/s1074-5521(98)90615-9. [DOI] [PubMed] [Google Scholar]
  • 19.Zou H, Henzel WJ, Liu X, Lutschg A, Wang X. Apaf-1, a human protein homologous to C. elegans CED-4, participates in cytochrome c-dependent activation of caspase-3. Cell. 1997;90(3):405–13. doi: 10.1016/s0092-8674(00)80501-2. [DOI] [PubMed] [Google Scholar]
  • 20.Bouillet P, Strasser A. BH3-only proteins—evolutionarily conserved proapoptotic Bcl-2 family members essential for initiating programmed cell death. J Cell Sci. 2002;115(Pt 8):1567–74. doi: 10.1242/jcs.115.8.1567. [DOI] [PubMed] [Google Scholar]
  • 21.Taylor RC, Cullen SP, Martin SJ. Apoptosis: controlled demolition at the cellular level. Nat Rev Mol Cell Biol. 2008;9(3):231–41. doi: 10.1038/nrm2312. [DOI] [PubMed] [Google Scholar]
  • 22.Cheng EH, Wei MC, Weiler S, Flavell RA, Mak TW, Lindsten T, Korsmeyer SJ. BCL-2, BCL-X(L) sequester BH3 domain-only molecules preventing BAX- and BAK-mediated mitochondrial apoptosis. Mol Cell. 2001;8(3):705–11. doi: 10.1016/s1097-2765(01)00320-3. [DOI] [PubMed] [Google Scholar]
  • 23.Qi XJ, Wildey GM, Howe PH. Evidence that Ser87 of BimEL is phosphorylated by Akt and regulates BimEL apoptotic function. J Biol Chem. 2006;281(2):813–23. doi: 10.1074/jbc.M505546200. [DOI] [PubMed] [Google Scholar]
  • 24.Belzacq AS, Vieira HL, Kroemer G, Brenner C. The adenine nucleotide translocator in apoptosis. Biochimie. 2002;84(2–3):167–76. doi: 10.1016/s0300-9084(02)01366-4. [DOI] [PubMed] [Google Scholar]
  • 25.Kuwana T, Bouchier-Hayes L, Chipuk JE, Bonzon C, Sullivan BA, Green DR, Newmeyer DD. BH3 domains of BH3-only proteins differentially regulate Bax-mediated mitochondrial membrane permeabilization both directly and indirectly. Mol Cell. 2005;17(4):525–35. doi: 10.1016/j.molcel.2005.02.003. [DOI] [PubMed] [Google Scholar]
  • 26.Letai A, Bassik MC, Walensky LD, Sorcinelli MD, Weiler S, Korsmeyer SJ. Distinct BH3 domains either sensitize or activate mitochondrial apoptosis, serving as prototype cancer therapeutics. Cancer Cell. 2002;2(3):183–92. doi: 10.1016/s1535-6108(02)00127-7. [DOI] [PubMed] [Google Scholar]
  • 27.Vander Heiden MG, Chandel NS, Williamson EK, Schumacker PT, Thompson CB. Bcl-xL regulates the membrane potential and volume homeostasis of mitochondria. Cell. 1997;91(5):627–37. doi: 10.1016/s0092-8674(00)80450-x. [DOI] [PubMed] [Google Scholar]
  • 28.Gazaryan IG, Brown AM. Intersection between mitochondrial permeability pores and mitochondrial fusion/fission. Neurochem Res. 2007;32(4–5):917–29. doi: 10.1007/s11064-006-9252-2. [DOI] [PubMed] [Google Scholar]
  • 29.Nakano K, Vousden KH. PUMA, a novel proapoptotic gene, is induced by p53. Mol Cell. 2001;7(3):683–94. doi: 10.1016/s1097-2765(01)00214-3. [DOI] [PubMed] [Google Scholar]
  • 30.Puthalakath H, Villunger A, O’Reilly LA, Beaumont JG, Coultas L, Cheney RE, Huang DC, Strasser A. Bmf: a proapoptotic BH3-only protein regulated by interaction with the myosin V actin motor complex, activated by anoikis. Science. 2001;293(5536):1829–32. doi: 10.1126/science.1062257. [DOI] [PubMed] [Google Scholar]
  • 31.Shibue T, Takeda K, Oda E, Tanaka H, Murasawa H, Takaoka A, Morishita Y, Akira S, Taniguchi T, Tanaka N. Integral role of Noxa in p53-mediated apoptotic response. Genes Dev. 2003;17(18):2233–8. doi: 10.1101/gad.1103603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Simpson CD, Anyiwe K, Schimmer AD. Anoikis resistance and tumor metastasis. Cancer Lett. 2008;272(2):177–85. doi: 10.1016/j.canlet.2008.05.029. [DOI] [PubMed] [Google Scholar]
  • 33.Safa AR, Pollok KE. Targeting the anti-apoptotic protein c-FLIP for cancer therapy. Cancers (Basel) 2011;3(2):1639–71. doi: 10.3390/cancers3021639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Mawji IA, Simpson CD, Hurren R, Gronda M, Williams MA, Filmus J, Jonkman J, Da Costa RS, Wilson BC, Thomas MP, Reed JC, Glinsky GV, Schimmer AD. Critical role for Fas-associated death domain-like interleukin-1-converting enzyme-like inhibitory protein in anoikis resistance and distant tumor formation. J Natl Cancer Inst. 2007;99(10):811–22. doi: 10.1093/jnci/djk182. [DOI] [PubMed] [Google Scholar]
  • 35.Kairouz-Wahbe R, Biliran H, Luo X, Khor I, Wankell M, Besch-Williford C, Pascual J, Oshima R, Ruoslahti E. Anoikis effector Bit1 negatively regulates Erk activity. Proc Natl Acad Sci U S A. 2008;105(5):1528–32. doi: 10.1073/pnas.0711357105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Ekert PG, Vaux DL. The mitochondrial death squad: hardened killers or innocent bystanders? Curr Opin Cell Biol. 2005;17(6):626–30. doi: 10.1016/j.ceb.2005.09.001. [DOI] [PubMed] [Google Scholar]
  • 37.Biliran H, Jan Y, Chen R, Pasquale EB, Ruoslahti E. Protein kinase D is a positive regulator of Bit1 apoptotic function. J Biol Chem. 2008;283(42):28029–37. doi: 10.1074/jbc.M803139200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Stupack DG, Cheresh DA. A Bit-role for integrins in apoptosis. Nat Cell Biol. 2004;6(5):388–9. doi: 10.1038/ncb0504-388. [DOI] [PubMed] [Google Scholar]
  • 39.Karmali PP, Brunquell C, Tram H, Ireland SK, Ruoslahti E, Biliran H. Metastasis of tumor cells is enhanced by downregulation of Bit1. PloS One. 2011;6(8):e23840. doi: 10.1371/journal.pone.0023840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Gasperowicz M, Otto F. Mammalian Groucho homologs: redundancy or specificity? J Cell Biochem. 2005;95(4):670–87. doi: 10.1002/jcb.20476. [DOI] [PubMed] [Google Scholar]
  • 41.Jan Y, Matter M, Pai JT, Chen YL, Pilch J, Komatsu M, Ong E, Fukuda M, Ruoslahti E. A mitochondrial protein, Bit1, mediates apoptosis regulated by integrins and Groucho/TLE corepressors. Cell. 2004;116(5):751–62. doi: 10.1016/s0092-8674(04)00204-1. [DOI] [PubMed] [Google Scholar]
  • 42.Beagle B, Johnson GV. AES/GRG5: more than just a dominant-negative TLE/GRG family member. Dev Dyn. 2010;239(11):2795–805. doi: 10.1002/dvdy.22439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Jenning S, Pham T, Ireland SK, Ruoslahti E, Biliran H. Bit1 in anoikis resistance and tumor metastasis. Cancer Lett. 2013;333(2):147–51. doi: 10.1016/j.canlet.2013.01.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Seo SW, Lee H, Lee HI, Kim HS. The role of TLE1 in synovial sarcoma. J Orthop Res. 2011;29(7):1131–6. doi: 10.1002/jor.21318. [DOI] [PubMed] [Google Scholar]
  • 45.Giancotti FG, Ruoslahti E. Integrin signaling. Science. 1999;285(5430):1028–32. doi: 10.1126/science.285.5430.1028. [DOI] [PubMed] [Google Scholar]
  • 46.Goel HL, Languino LR. Integrin signaling in cancer. Cancer Treat Res. 2004;119:15–31. doi: 10.1007/1-4020-7847-1_2. [DOI] [PubMed] [Google Scholar]
  • 47.Fornaro M, Manes T, Languino LR. Integrins and prostate cancer metastases. Cancer Metastasis Rev. 2001;20(3–4):321–31. doi: 10.1023/a:1015547830323. [DOI] [PubMed] [Google Scholar]
  • 48.Goel HL, Li J, Kogan S, Languino LR. Integrins in prostate cancer progression. Endocr Relat Cancer. 2008;15(3):657–64. doi: 10.1677/ERC-08-0019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Nikolopoulos SN, Blaikie P, Yoshioka T, Guo W, Giancotti FG. Integrin beta4 signaling promotes tumor angiogenesis. Cancer Cell. 2004;6(5):471–83. doi: 10.1016/j.ccr.2004.09.029. [DOI] [PubMed] [Google Scholar]
  • 50.Lee YC, Jin JK, Cheng CJ, Huang CF, Song JH, Huang M, Brown WS, Zhang S, Yu-Lee LY, Yeh ET, McIntyre BW, Logothetis CJ, Gallick GE, Lin SH. Targeting constitutively activated beta1 integrins inhibits prostate cancer metastasis. Mol Cancer Res. 2013;11(4):405–17. doi: 10.1158/1541-7786.MCR-12-0551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Yeatman TJ. A renaissance for SRC. Nat Rev Cancer. 2004;4(6):470–80. doi: 10.1038/nrc1366. [DOI] [PubMed] [Google Scholar]
  • 52.Giannoni E, Buricchi F, Grimaldi G, Parri M, Cialdai F, Taddei ML, Raugei G, Ramponi G, Chiarugi P. Redox regulation of anoikis: reactive oxygen species as essential mediators of cell survival. Cell Death Differ. 2008;15(5):867–78. doi: 10.1038/cdd.2008.3. [DOI] [PubMed] [Google Scholar]
  • 53.Giannoni E, Fiaschi T, Ramponi G, Chiarugi P. Redox regulation of anoikis resistance of metastatic prostate cancer cells: key role for Src and EGFR-mediated prosurvival signals. Oncogene. 2009;28(20):2074–86. doi: 10.1038/onc.2009.77. [DOI] [PubMed] [Google Scholar]
  • 54.Diaz-Montero CM, Wygant JN, McIntyre BW. PI3-K/Akt-mediated anoikis resistance of human osteosarcoma cells requires Src activation. Eur J Cancer. 2006;42(10):1491–500. doi: 10.1016/j.ejca.2006.03.007. [DOI] [PubMed] [Google Scholar]
  • 55.Duxbury MS, Ito H, Benoit E, Waseem T, Ashley SW, Whang EE. A novel role for carcinoembryonic antigen-related cell adhesion molecule 6 as a determinant of gemcitabine chemoresistance in pancreatic adenocarcinoma cells. Cancer Res. 2004;64(11):3987–93. doi: 10.1158/0008-5472.CAN-04-0424. [DOI] [PubMed] [Google Scholar]
  • 56.Park SI, Zhang J, Phillips KA, Araujo JC, Najjar AM, Volgin AY, Gelovani JG, Kim SJ, Wang Z, Gallick GE. Targeting SRC family kinases inhibits growth and lymph node metastases of prostate cancer in an orthotopic nude mouse model. Cancer Res. 2008;68(9):3323–33. doi: 10.1158/0008-5472.CAN-07-2997. [DOI] [PubMed] [Google Scholar]
  • 57.Hannigan GE, Leung-Hagesteijn C, Fitz-Gibbon L, Coppolino MG, Radeva G, Filmus J, Bell JC, Dedhar S. Regulation of cell adhesion and anchorage-dependent growth by a new beta 1-integrin-linked protein kinase. Nature. 1996;379(6560):91–6. doi: 10.1038/379091a0. [DOI] [PubMed] [Google Scholar]
  • 58.Li F, Drummer HE, Ficorilli N, Studdert MJ, Crabb BS. Identification of noncytopathic equine rhinovirus 1 as a cause of acute febrile respiratory disease in horses. J Clin Microbiol. 1997;35(4):937–43. doi: 10.1128/jcm.35.4.937-943.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Wu C, Dedhar S. Integrin-linked kinase (ILK) and its interactors: a new paradigm for the coupling of extracellular matrix to actin cytoskeleton and signaling complexes. J Cell Biol. 2001;155(4):505–10. doi: 10.1083/jcb.200108077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Graff JR, Deddens JA, Konicek BW, Colligan BM, Hurst BM, Carter HW, Carter JH. Integrin-linked kinase expression increases with prostate tumor grade. Clin Cancer Res. 2001;7(7):1987–91. [PubMed] [Google Scholar]
  • 61.Cieslik K, Zembowicz A, Tang JL, Wu KK. Transcriptional regulation of endothelial nitric-oxide synthase by lysophosphatidylcholine. J Biol Chem. 1998;273(24):14885–90. doi: 10.1074/jbc.273.24.14885. [DOI] [PubMed] [Google Scholar]
  • 62.Radeva G, Petrocelli T, Behrend E, Leung-Hagesteijn C, Filmus J, Slingerland J, Dedhar S. Overexpression of the integrin-linked kinase promotes anchorage-independent cell cycle progression. J Biol Chem. 1997;272(21):13937–44. doi: 10.1074/jbc.272.21.13937. [DOI] [PubMed] [Google Scholar]
  • 63.Lynch DK, Ellis CA, Edwards PA, Hiles ID. Integrin-linked kinase regulates phosphorylation of serine 473 of protein kinase B by an indirect mechanism. Oncogene. 1999;18(56):8024–32. doi: 10.1038/sj.onc.1203258. [DOI] [PubMed] [Google Scholar]
  • 64.Attwell S, Roskelley C, Dedhar S. The integrin-linked kinase (ILK) suppresses anoikis. Oncogene. 2000;19(33):3811–5. doi: 10.1038/sj.onc.1203711. [DOI] [PubMed] [Google Scholar]
  • 65.Fukuda T, Chen K, Shi X, Wu C. PINCH-1 is an obligate partner of integrin-linked kinase (ILK) functioning in cell shape modulation, motility, and survival. J Biol Chem. 2003;278(51):51324–33. doi: 10.1074/jbc.M309122200. [DOI] [PubMed] [Google Scholar]
  • 66.Hannigan G, Troussard AA, Dedhar S. Integrin-linked kinase: a cancer therapeutic target unique among its ILK. Nat Rev Cancer. 2005;5(1):51–63. doi: 10.1038/nrc1524. [DOI] [PubMed] [Google Scholar]
  • 67.Ye X, Weinberg RA. Epithelial-mesenchymal plasticity: a central regulator of cancer progression. Trends Cell Biol. 2015;25(11):675–86. doi: 10.1016/j.tcb.2015.07.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Cao Z, Kyprianou N. Mechanisms navigating the TGF-β pathway in prostate cancer. Asian J Urol. 2015;2(1):11–8. doi: 10.1016/j.ajur.2015.04.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Pu H, Horbinski C, Hensley PJ, Matuszak EA, Atkinson T, Kyprianou N. PARP-1 regulates epithelial-mesenchymal transition (EMT) in prostate tumorigenesis. Carcinogenesis. 2014;35(11):2592–601. doi: 10.1093/carcin/bgu183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Wu Y, Zhou BP. Snail: more than EMT. Cell Adh Migr. 2010;4(2):199–203. doi: 10.4161/cam.4.2.10943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Paoli P, Giannoni E, Chiarugi P. Anoikis molecular pathways and its role in cancer progression. Biochim Biophys Acta. 2013;1833(12):3481–98. doi: 10.1016/j.bbamcr.2013.06.026. [DOI] [PubMed] [Google Scholar]
  • 72.Sanchez-Tillo E, Fanlo L, Siles L, Montes-Moreno S, Moros A, Chiva-Blanch G, Estruch R, Martinez A, Colomer D, Győrffy B, Roué G, Postigo A. The EMT activator ZEB1 promotes tumor growth and determines differential response to chemotherapy in mantle cell lymphoma. Cell Death Differ. 2014;21(2):247–57. doi: 10.1038/cdd.2013.123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Kwok WK, Ling MT, Lee TW, Lau TC, Zhou C, Zhang X, Chua CW, Chan KW, Chan FL, Glackin C, Wong YC, Wang X. Up-regulation of TWIST in prostate cancer and its implication as a therapeutic target. Cancer Res. 2005;65(12):5153–62. doi: 10.1158/0008-5472.CAN-04-3785. [DOI] [PubMed] [Google Scholar]
  • 74.Yang J, Mani SA, Donaher JL, Ramaswamy S, Itzykson RA, Come C, Savagner P, Gitelman I, Richardson A, Weinberg RA. Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell. 2004;117(7):927–39. doi: 10.1016/j.cell.2004.06.006. [DOI] [PubMed] [Google Scholar]
  • 75.Blanco MJ, Moreno-Bueno G, Sarrio D, Locascio A, Cano A, Palacios J, Nieto MA. Correlation of Snail expression with histological grade and lymph node status in breast carcinomas. Oncogene. 2002;21(20):3241–6. doi: 10.1038/sj.onc.1205416. [DOI] [PubMed] [Google Scholar]
  • 76.Barrallo-Gimeno A, Nieto MA. The Snail genes as inducers of cell movement and survival: implications in development and cancer. Development. 2005;132(14):3151–61. doi: 10.1242/dev.01907. [DOI] [PubMed] [Google Scholar]
  • 77.Schmalhofer O, Brabletz S, Brabletz T. E-cadherin, beta-catenin, and ZEB1 in malignant progression of cancer. Cancer Metastasis Rev. 2009;28(1–2):151–66. doi: 10.1007/s10555-008-9179-y. [DOI] [PubMed] [Google Scholar]
  • 78.Orford K, Orford CC, Byers SW. Exogenous expression of beta-catenin regulates contact inhibition, anchorage-independent growth, anoikis, and radiation-induced cell cycle arrest. J Cell Biol. 1999;146(4):855–68. doi: 10.1083/jcb.146.4.855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Gemmill RM, Roche J, Potiron VA, Nasarre P, Mitas M, Coldren CD, Helfrich BA, Garrett-Mayer E, Bunn PA, Drabkin HA. ZEB1-responsive genes in non-small cell lung cancer. Cancer Lett. 2011;300(1):66–78. doi: 10.1016/j.canlet.2010.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Ohira T, Gemmill RM, Ferguson K, Kusy S, Roche J, Brambilla E, Zeng C, Baron A, Bemis L, Erickson P, Wilder E, Rustgi A, Kitajewski J, Gabrielson E, Bremnes R, Franklin W, Drabkin HA. WNT7a induces E-cadherin in lung cancer cells. Proc Natl Acad Sci U S A. 2003;100(18):10429–34. doi: 10.1073/pnas.1734137100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Takeyama Y, Sato M, Horio M, Hase T, Yoshida K, Yokoyama T, Nakashima H, Hashimoto N, Sekido Y, Gazdar AF, Minna JD, Kondo M, Hasegawa Y. Knockdown of ZEB1, a master epithelial-to-mesenchymal transition (EMT) gene, suppresses anchorage-independent cell growth of lung cancer cells. Cancer Lett. 2010;296(2):216–24. doi: 10.1016/j.canlet.2010.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Beer TM, Armstrong AJ, Rathkopf DE, Loriot Y, Sternberg CN, Higano CS, Iversen P, Bhattacharya S, Carles J, Chowdhury S, Davis ID, de Bono JS, Evans CP, Fizazi K, Joshua AM, Kim CS, Kimura G, Mainwaring P, Mansbach H, Miller K, Noonberg SB, Perabo F, Phung D, Saad F, Scher HI, Taplin ME, Venner PM, PREVAIL Investigators Enzalutamide in metastatic prostate cancer before chemotherapy. N Engl J Med. 2014;371(5):424–33. doi: 10.1056/NEJMoa1405095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Kahn B, Collazo J, Kyprianou N. Androgen receptor as a driver of therapeutic resistance in advanced prostate cancer. Int J Biol Sci. 2014;10(6):588–95. doi: 10.7150/ijbs.8671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Tran C, Ouk S, Clegg NJ, Chen Y, Watson PA, Arora V, Wongvipat J, Smith-Jones PM, Yoo D, Kwon A, Wasielewska T, Welsbie D, Chen CD, Higano CS, Beer TM, Hung DT, Scher HI, Jung ME, Sawyers CL. Development of a second-generation antiandrogen for treatment of advanced prostate cancer. Science. 2009;324(5928):787–90. doi: 10.1126/science.1168175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Chen CD, Welsbie DS, Tran C, Baek SH, Chen R, Vessella R, Rosenfeld MG, Sawyers CL. Molecular determinants of resistance to antiandrogen therapy. Nat Med. 2004;10(1):33–9. doi: 10.1038/nm972. [DOI] [PubMed] [Google Scholar]
  • 86.Martin SK, Kyprianou N. Exploitation of the androgen receptor to overcome taxane resistance in advanced prostate cancer. Adv Cancer Res. 2015;127:123–58. doi: 10.1016/bs.acr.2015.03.001. [DOI] [PubMed] [Google Scholar]
  • 87.Li Y, Chan SC, Brand LJ, Hwang TH, Silverstein KA, Dehm SM. Androgen receptor splice variants mediate enzalutamide resistance in castration-resistant prostate cancer cell lines. Cancer Res. 2013;73(2):483–9. doi: 10.1158/0008-5472.CAN-12-3630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Shafi AA, Cox MB, Weigel NL. Androgen receptor splice variants are resistant to inhibitors of Hsp90 and FKBP52, which alter androgen receptor activity and expression. Steroids. 2013;78(6):548–54. doi: 10.1016/j.steroids.2012.12.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Martin SK, Banuelos CA, Sadar MD, Kyprianou N. N-terminal targeting of androgen receptor variant enhances response of castration resistant prostate cancer to taxane chemotherapy. Mol Oncol. 2014 Nov 15; doi: 10.1016/j.molonc.2014.10.014. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Shafi AA, Yen AE, Weigel NL. Androgen receptors in hormone-dependent and castration-resistant prostate cancer. Pharmacol Ther. 2013;140(3):223–38. doi: 10.1016/j.pharmthera.2013.07.003. [DOI] [PubMed] [Google Scholar]
  • 91.Buttigliero C, Tucci M, Bertaglia V, Vignani F, Bironzo P, Di Maio M, Scagliotti GV. Understanding and overcoming the mechanisms of primary and acquired resistance to abiraterone and enzalutamide in castration resistant prostate cancer. Cancer Treat Rev. 2015;41(10):884–92. doi: 10.1016/j.ctrv.2015.08.002. [DOI] [PubMed] [Google Scholar]
  • 92.Zhu ML, Kyprianou N. Role of androgens and the androgen receptor in epithelial-mesenchymal transition and invasion of prostate cancer cells. FASEB J. 2010;24(3):769–77. doi: 10.1096/fj.09-136994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Sun Y, Wang BE, Leong KG, Yue P, Li L, Jhunjhunwala S, Chen D, Seo K, Modrusan Z, Gao WQ, Settleman J, Johnson L. Androgen deprivation causes epithelial-mesenchymal transition in the prostate: implications for androgen-deprivation therapy. Cancer Res. 2012;72(2):527–36. doi: 10.1158/0008-5472.CAN-11-3004. [DOI] [PubMed] [Google Scholar]
  • 94.Lu-Yao GL, Albertsen PC, Moore DF, Shih W, Lin Y, DiPaola RS, Yao SL. Survival following primary androgen deprivation therapy among men with localized prostate cancer. JAMA. 2008;300(2):173–81. doi: 10.1001/jama.300.2.173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Ryan CJ, Shah S, Efstathiou E, Smith MR, Taplin ME, Bubley GJ, Logothetis CJ, Kheoh T, Kilian C, Haqq CM, Molina A, Small EJ. Phase II study of abiraterone acetate in chemotherapy-naive metastatic castration-resistant prostate cancer displaying bone flare discordant with serologic response. Clin Cancer Res. 2011;17(14):4854–61. doi: 10.1158/1078-0432.CCR-11-0815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Jennbacken K, Tesan T, Wang W, Gustavsson H, Damber JE, Welen K. N-cadherin increases after androgen deprivation and is associated with metastasis in prostate cancer. Endocr Relat Cancer. 2010;17(2):469–79. doi: 10.1677/ERC-10-0015. [DOI] [PubMed] [Google Scholar]
  • 97.Chu K, Cheng CJ, Ye X, Lee YC, Zurita AJ, Chen DT, Yu-Lee LY, Zhang S, Yeh ET, Hu MC, Logothetis CJ, Lin SH. Cadherin-11 promotes the metastasis of prostate cancer cells to bone. Mol Cancer Res. 2008;6(8):1259–67. doi: 10.1158/1541-7786.MCR-08-0077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Lee YC, Cheng CJ, Huang M, Bilen MA, Ye X, Navone NM, Chu K, Kao HH, Yu-Lee LY, Wang Z, Lin SH. Androgen depletion up-regulates cadherin-11 expression in prostate cancer. J Pathol. 2010;221(1):68–76. doi: 10.1002/path.2687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Kleeberger W, Bova GS, Nielsen ME, Herawi M, Chuang AY, Epstein JI, Berman DM. Roles for the stem cell associated intermediate filament Nestin in prostate cancer migration and metastasis. Cancer Res. 2007;67(19):9199–206. doi: 10.1158/0008-5472.CAN-07-0806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Fanning AS, Little BP, Rahner C, Utepbergenov D, Walther Z, Anderson JM. The unique-5 and -6 motifs of ZO-1 regulate tight junction strand localization and scaffolding properties. Mol Biol Cell. 2007;18(3):721–31. doi: 10.1091/mbc.E06-08-0764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Horowitz JC, Rogers DS, Sharma V, Vittal R, White ES, Cui Z, Thannickal VJ. Combinatorial activation of FAK and AKT by transforming growth factor-beta1 confers an anoikis-resistant phenotype to myofibroblasts. Cell Signal. 2007;19(4):761–71. doi: 10.1016/j.cellsig.2006.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Natsuizaka M, Ohashi S, Wong GS, Ahmadi A, Kalman RA, Budo D, Klein-Szanto AJ, Herlyn M, Diehl JA, Nakagawa H. Insulin-like growth factor-binding protein-3 promotes transforming growth factor-{beta}1-mediated epithelial-to-mesenchymal transition and motility in transformed human esophageal cells. Carcinogenesis. 2010;31(8):1344–53. doi: 10.1093/carcin/bgq108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Turner JR. ‘Putting the squeeze’ on the tight junction: understanding cytoskeletal regulation. Semin Cell Dev Biol. 2000;11(4):301–8. doi: 10.1006/scdb.2000.0180. [DOI] [PubMed] [Google Scholar]
  • 104.Van Itallie CM, Anderson JM. Claudins and epithelial paracellular transport. Annu Rev Physiol. 2006;68:403–29. doi: 10.1146/annurev.physiol.68.040104.131404. [DOI] [PubMed] [Google Scholar]
  • 105.Li XY, Zhou X, Rowe RG, Hu Y, Schlaepfer DD, Ilić D, Dressler G, Park A, Guan JL, Weiss SJ. Snail1 controls epithelial-mesenchymal lineage commitment in focal adhesion kinase-null embryonic cells. J Cell Biol. 2011;195(5):729–38. doi: 10.1083/jcb.201105103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Reginato MJ, Mills KR, Paulus JK, Lynch DK, Sgroi DC, Debnath J, Muthuswamy SK, Brugge JS. Integrins and EGFR coordinately regulate the pro-apoptotic protein Bim to prevent anoikis. Nat Cell Biol. 2003;5(8):733–40. doi: 10.1038/ncb1026. [DOI] [PubMed] [Google Scholar]
  • 107.Crowe DL, Ohannessian A. Recruitment of focal adhesion kinase and paxillin to beta1 integrin promotes cancer cell migration via mitogen activated protein kinase activation. BMC Cancer. 2004;4:18. doi: 10.1186/1471-2407-4-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Li J, Larocca JN, Rodriguez-Gabin AG, Charron MJ. Expression and signal transduction of the glucagon receptor in betaTC3 cells. Biochim Biophys Acta. 1997;1356(2):229–36. doi: 10.1016/s0167-4889(96)00170-x. [DOI] [PubMed] [Google Scholar]
  • 109.Shibue T, Brooks MW, Weinberg RA. An integrin-linked machinery of cytoskeletal regulation that enables experimental tumor initiation and metastatic colonization. Cancer Cell. 2013;24(4):481–98. doi: 10.1016/j.ccr.2013.08.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Martin SS, Ridgeway AG, Pinkas J, Lu Y, Reginato MJ, Koh EY, Michelman M, Daley GQ, Brugge JS, Leder P. A cytoskeleton-based functional genetic screen identifies Bcl-xL as an enhancer of metastasis, but not primary tumor growth. Oncogene. 2004;23(26):4641–5. doi: 10.1038/sj.onc.1207595. [DOI] [PubMed] [Google Scholar]
  • 111.Sun HQ, Yamamoto M, Mejillano M, Yin HL. Gelsolin, a multifunctional actin regulatory protein. J Biol Chem. 1999;274(47):33179–82. doi: 10.1074/jbc.274.47.33179. [DOI] [PubMed] [Google Scholar]
  • 112.Collazo J, Zhu B, Larkin S, Martin SK, Pu H, Horbinski C, Koochekpour S, Kyprianou N. Cofilin drives cell-invasive and metastatic responses to TGF-β in prostate cancer. Cancer Res. 2014;74(8):2362–73. doi: 10.1158/0008-5472.CAN-13-3058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Kizhatil K, Davis JQ, Davis L, Hoffman J, Hogan BL, Bennett V. Ankyrin-G is a molecular partner of E-cad-herin in epithelial cells and early embryos. J Biol Chem. 2007;282(36):26552–61. doi: 10.1074/jbc.M703158200. [DOI] [PubMed] [Google Scholar]
  • 114.Davila M, Frost AR, Grizzle WE, Chakrabarti R. LIM kinase 1 is essential for the invasive growth of prostate epithelial cells: implications in prostate cancer. J Biol Chem. 2003;278(38):36868–75. doi: 10.1074/jbc.M306196200. [DOI] [PubMed] [Google Scholar]
  • 115.Calderwood DA, Yan B, de Pereda JM, Alvarez BG, Fu-jioka Y, Liddington RC, Ginsberg MH. The phosphotyrosine binding-like domain of talin activates integrins. J Biol Chem. 2002;277(24):21749–58. doi: 10.1074/jbc.M111996200. [DOI] [PubMed] [Google Scholar]
  • 116.Mebratu Y, Tesfaigzi Y. How ERK1/2 activation controls cell proliferation and cell death: is subcellular localization the answer? Cell Cycle. 2009;8(8):1168–75. doi: 10.4161/cc.8.8.8147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Martin SK, Kamelgarn M, Kyprianou N. Cytoskeleton targeting value in prostate cancer treatment. Am J Clin Exp Urol. 2014;2(1):15–26. [PMC free article] [PubMed] [Google Scholar]
  • 118.Jin JK, Tien PC, Cheng CJ, Song JH, Huang C, Lin SH, Gallick GE. Talin1 phosphorylation activates beta1 integrins: a novel mechanism to promote prostate cancer bone metastasis. Oncogene. 2015;34(14):1811–21. doi: 10.1038/onc.2014.116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Meyer G, Kim B, van Golen C, Feldman EL. Cofilin activity during insulin-like growth factor I-stimulated neuroblastoma cell motility. Cell Mol Life Sci. 2005;62(4):461–70. doi: 10.1007/s00018-004-4456-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Niwa R, Nagata-Ohashi K, Takeichi M, Mizuno K, Ue-mura T. Control of actin reorganization by Slingshot, a family of phosphatases that dephosphorylate ADF/cofilin. Cell. 2002;108(2):233–46. doi: 10.1016/s0092-8674(01)00638-9. [DOI] [PubMed] [Google Scholar]
  • 121.Zhu B, Fukada K, Zhu H, Kyprianou N. Prohibitin and cofilin are intracellular effectors of transforming growth factor beta signaling in human prostate cancer cells. Cancer Res. 2006;66(17):8640–7. doi: 10.1158/0008-5472.CAN-06-1443. [DOI] [PubMed] [Google Scholar]
  • 122.Garrison JB, Shaw YJ, Chen CS, Kyprianou N. Novel quinazoline-based compounds impair prostate tumorigenesis by targeting tumor vascularity. Cancer Res. 2007;67(23):11344–52. doi: 10.1158/0008-5472.CAN-07-1662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Sampson N, Zenzmaier C, Heitz M, Hermann M, Plas E, Schäfer G, Klocker H, Berger P. Stromal insulin-like growth factor binding protein 3 (IGFBP3) is elevated in the diseased human prostate and promotes ex vivo fibro-blast-to-myofibroblast differentiation. Endocrinology. 2013;154(8):2586–99. doi: 10.1210/en.2012-2259. [DOI] [PubMed] [Google Scholar]
  • 124.Nam S, Kim D, Cheng JQ, Zhang S, Lee JH, Buettner R, Mirosevich J, Lee FY, Jove R. Action of the Src family kinase inhibitor, dasatinib (BMS-354825), on human prostate cancer cells. Cancer Res. 2005;65(20):9185–9. doi: 10.1158/0008-5472.CAN-05-1731. [DOI] [PubMed] [Google Scholar]
  • 125.Huang RY, Wong MK, Tan TZ, Kuay KT, Ng AH, Chung VY, Chu YS, Matsumura N, Lai HC, Lee YF, Sim WJ, Chai C, Pietschmann E, Mori S, Low JJ, Choolani M, Thiery JP. An EMT spectrum defines an anoikis-resistant and spheroidogenic intermediate mesenchymal state that is sensitive to e-cadherin restoration by a src-ki-nase inhibitor, saracatinib (AZD0530) Cell Death Dis. 2013;4:e915. doi: 10.1038/cddis.2013.442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Twardowski PW, Beumer JH, Chen CS, Kraft AS, Chatta GS, Mitsuhashi M, Ye W, Christner SM, Lilly MB. A phase II trial of dasatinib in patients with metastatic castration-resistant prostate cancer treated previously with chemotherapy. Anticancer Drugs. 2013;24(7):743–53. doi: 10.1097/CAD.0b013e328361feb0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Araujo JC, Trudel GC, Saad F, Armstrong AJ, Yu EY, Bellmunt J, Wilding G, McCaffrey J, Serrano SV, Matveev VB, Efstathiou E, Oudard S, Morris MJ, Sizer B, Goebell PJ, Heidenreich A, de Bono JS, Begbie S, Hong JH, Richardet E, Gallardo E, Paliwal P, Durham S, Cheng S, Logothetis CJ. Docetaxel and dasatinib or placebo in men with metastatic castration-resistant prostate cancer (READY): a randomised, double-blind phase 3 trial. Lancet Oncol. 2013;14(13):1307–16. doi: 10.1016/S1470-2045(13)70479-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Sakuma Y, Tsunezumi J, Nakamura Y, Yoshihara M, Matsukuma S, Koizume S, Miyagi Y. ABT-263, a Bcl-2 inhibitor, enhances the susceptibility of lung adenocarcinoma cells treated with Src inhibitors to anoikis. Oncol Rep. 2011;25(3):661–7. doi: 10.3892/or.2010.1123. [DOI] [PubMed] [Google Scholar]
  • 129.Gambacorti-Passerini C, Kantarjian HM, Kim DW, Khoury HJ, Turkina AG, Brümmendorf TH, Matczak E, Bardy-Bouxin N, Shapiro M, Turnbull K, Leip E, Cortes JE. Long-term efficacy and safety of bosutinib in patients with advanced leukemia following resistance/intolerance to imatinib and other tyrosine kinase inhibitors. Am J Hematol. 2015;90(9):755–68. doi: 10.1002/ajh.24034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Ng YK, Wong EY, Lau CP, Chan JP, Wong SC, Chan AS, Kwan MP, Tsao SW, Tsang CM, Lai PB, Chan AT, Lui VW. K252a induces anoikis-sensitization with suppression of cellular migration in Epstein-Barr virus (EBV)-associated nasopharyngeal carcinoma cells. Invest New Drugs. 2012;30(1):48–58. doi: 10.1007/s10637-010-9513-4. [DOI] [PubMed] [Google Scholar]
  • 131.Qian F, Engst S, Yamaguchi K, Yu P, Won KA, Mock L, Lou T, Tan J, Li C, Tam D, Lougheed J, Yakes FM, Bentzien F, Xu W, Zaks T, Wooster R, Greshock J, Joly AH. Inhibition of tumor cell growth, invasion, and metastasis by EXEL-2880 (XL880, GSK1363089), a novel inhibitor of HGF and VEGF receptor tyrosine kinases. Cancer Res. 2009;69(20):8009–16. doi: 10.1158/0008-5472.CAN-08-4889. [DOI] [PubMed] [Google Scholar]
  • 132.Zillhardt M, Park SM, Romero IL, Sawada K, Montag A, Krausz T, Yamada SD, Peter ME, Lengyel E. Foretinib (GSK1363089), an orally available multikinase inhibitor of c-Met and VEGFR-2, blocks proliferation, induces anoikis, and impairs ovarian cancer metastasis. Clin Cancer Res. 2011;17(12):4042–51. doi: 10.1158/1078-0432.CCR-10-3387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Klubo-Gwiezdzinska J, Jensen K, Costello J, Patel A, Hoperia V, Bauer A, Burman KD, Wartofsky L, Vasko V. Metformin inhibits growth and decreases resistance to anoikis in medullary thyroid cancer cells. Endocr Relat Cancer. 2012;19(3):447–56. doi: 10.1530/ERC-12-0046. [DOI] [PubMed] [Google Scholar]
  • 134.Yu H, Yin L, Jiang X, Sun X, Wu J, Tian H, Gao X, He X. Effect of metformin on cancer risk and treatment outcome of prostate cancer: a meta-analysis of epidemiological observational studies. PloS One. 2014;9(12):e116327. doi: 10.1371/journal.pone.0116327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Rothermundt C, Hayoz S, Templeton AJ, Winterhalder R, Strebel RT, Bärtschi D, Pollak M, Lui L, Endt K, Schiess R, Rüschoff JH, Cathomas R, Gillessen S. Metformin in chemotherapy-naive castration-resistant prostate cancer: a multicenter phase 2 trial (SAKK 08/09) Eur Urol. 2014;66(3):468–74. doi: 10.1016/j.eururo.2013.12.057. [DOI] [PubMed] [Google Scholar]
  • 136.Nobes JP, Langley SE, Klopper T, Russell-Jones D, Laing RW. A prospective, randomized pilot study evaluating the effects of metformin and lifestyle intervention on patients with prostate cancer receiving androgen deprivation therapy. BJU Int. 2012;109(10):1495–502. doi: 10.1111/j.1464-410X.2011.10555.x. [DOI] [PubMed] [Google Scholar]

RESOURCES