extracellular hsp90: an emerging target for cancer therapy

Upload: arun-maurya

Post on 04-Jun-2018

220 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/13/2019 Extracellular HSP90: An Emerging Target for Cancer Therapy

    1/8

    Current Signal Transduction Therapy, 2009, 4, 51-58 51

    1574-3624/09 $55.00+.00 2009 Bentham Science Publishers Ltd.

    Extracellular HSP90: An Emerging Target for Cancer Therapy

    Katerina Sidera1and Evangelia Patsavoudi1,2,*

    1Department of Biochemistry, Hellenic Pasteur Institute, Athens, Greece; 2Department of Biomedical Instrumentation

    Technology, Technological Educational Institute of Athens, Greece

    Abstract: Cancer is a genetic disease which progresses from benign to malignant stages through the steady acquisition of

    genomic mutations in key cell-regulatory genes, namely oncogenes, tumor-suppressors, and stability genes. In many ways

    cancer is considered as a disease of de-regulated signal transduction. Oncogenic mutations frequently lead to over-

    expression and/or constitutive activation of signal transduction components, allowing cancer cells to override the control-

    ling mechanisms of signalling networks and acquire the cancer-associated traits known as the six hallmarks of cancer.

    The molecular chaperone HSP90 is viewed as a key player in the subversion of normal cells toward transformation, since

    many of its client proteins are linked to signalling pathways, commonly de-regulated during tumorigenesis. Consequently,

    over the past years HSP90 has emerged as a promising and exciting target for the development of cancer chemotherapeu-

    tics and already several HSP90 inhibitors are under clinical evaluation.

    Recently, a pool of HSP90 was identified at the cell surface, where it was shown to be involved in signalling pathways

    leading to cell motility and invasion. Independent studies suggest that surface HSP90 could be a promising target for the

    development of effective anti-metastatic strategies. Thus a need for the development of novel cell-impermeable HSP90

    inhibitors is emerging.

    Key Words: Extracellular HSP90, cell invasion, signal transduction, cancer therapeutics.

    INTRODUCTION

    Cancer has become in recent years less of an enigma;after decades of rapid advances in cancer research, severallines of evidence indicate that during the course of tumorprogression, cancer cells gradually acquire a number of dy-namic genetic alterations [1]. In 2000, Hanahan and Wein-berg [2], proposed that the complex array of phenotypes dis-played by cancer cells may be organized in six cancer-associated traits. These include the cellular capacities to pro-

    liferate indefinitely (immortalization), to become independ-ent of extracellular growth or anti-growth signals, to evadeapoptosis, to induce a self-sustained supply of nutrients andoxygen (angiogenesis) and ultimately, to invade and metas-tasize to distant sites. These cancer-associated hallmarkcharacteristics actually reflect genetic alterations in multiplesafeguard genes of the cell, namely oncogenes and tumor-suppressor genes. These genes are responsible for the regula-tion of signal transduction molecules, including among oth-ers receptor tyrosine kinases (RTKs), products of the Rasand raf genes, and protein kinases of the mitogen-activatedprotein kinase (MAPK), phosphoinositide 3-kinase (PI3K)and Akt. These molecules in normal cells control the tightcoordination of diverse processes such as cell survival, pro-

    liferation, growth, differentiation and motility [3, 4].

    The broad term signal transduction refers to the flow ofbiological information from the cells micro-environment(extracellular matrix, neighbouring cells) via detection sys-tems such as surface receptors and through intermediatemolecules, inside the cell. The latter include a wide array of

    *Address correspondence to this author at the Department of Biochemistry,Hellenic Pasteur Institute, 127, Vas. Sofias Av.,11521 Athens, Greece; Tel:+30 2106478871; Fax: +30 2106423498; E-mail: [email protected]

    intracellular elements interacting through cascades of chemical signals and ultimately controlling the activities of specific intracellular effectors, resulting in the tight coordinationof fundamental processes of cellular life [5]. Signallingpathways are not isolated from each other, but instead theyare interconnected, forming a labyrinth of intersecting andoverlapping networks, enhancing thus the robustness anddiversity of signalling and permitting fine-tuning and amplification or attenuation of the output [6].

    Advances in the understanding of aberrant signallingpathways in various types of cancers have directly promotedthe development of targeted therapies-that is the development of drugs that influence the action and/or activity of aspecific signalling molecule. Elucidation of the roles omany kinase signalling pathways in cancer, including growthfactor receptors and their downstream effectors, along withthe identification of kinases as an attractive drug-target classhas led to the expectation of innovating cancer-treatmenstrategies with more specific mechanisms of action thanconventional chemotherapeutic agents. However, the cruciachallenge to select appropriate targets/approaches in cancetherapeutics still remains.

    HSP90: MORE THAN A FOLDING TOOL

    Heat shock proteins (HSPs) were first discovered in 1962[7, 8] as a small set of highly conserved proteins whose expression increased, as a response to elevated temperatureSince then, the biology of HSPs has been studied extensivelyand it is generally accepted that these proteins have evolvedas a cellular protection response to a multitude of stresseincluding heat, alcohol, hypoxia, oxidative stress, acidosisnutrient deprivation, and inflammation [9-11]. FuthermoreHSPs function as central components of the molecular chaperone machinery, responsible for the folding, stability, deg

  • 8/13/2019 Extracellular HSP90: An Emerging Target for Cancer Therapy

    2/8

    52 Current Signal Transduction Therapy,2009, Vol. 4, No. 1 Sidera and Patsavoud

    radation and transportation of a multitude of proteins andconsequently affecting diverse cellular processes [12-14].Among the members of the family, the 90-kDa heat shockprotein (HSP90), has recently emerged as a molecule of par-ticular interest due to the fact that many of its "client" pro-teins, including mutant p53, HER-2, HIF-1, Akt/PKB c-Raf -1, hTERT and CDK4, are members of the most-wanted listof proteins considered responsible for the multiple hallmark

    traits of malignancy (see Table 1) [15-18].

    Table 1. HSP90 Clients and the Multiple Hallmarks of

    Cancer

    Cancer-associated Cellular Traits HSP90 Client Proteins

    Self sufficiency in growth signals MEK

    SRC tyrosine kinases

    Steroid hormone receptors

    Insensitivity to anti-growth signals Akt

    CDK2,CDK4

    HIF1a

    RTKsSRC tyrosine kinases

    Steroid hormone receptors

    Evading apoptosis Akt

    RTKs

    Survivin

    Tissue invasion and metastasis MMP2

    Limitless replicative potential CDK2, CDK4

    Telomerase

    Sustained angiogenesis VEGFR2

    HIF1a

    HSP90 is a highly conserved and essential stress protein,expressed throughout the eukaryotic lineage [19-22]. Highereukaryotes possess multiple HSP90 homologues, includingthe highly conserved HSP90 and HSP90 isoforms (86%amino acid conservation) which are mainly cytoplasmic [23],GRP94 in the endoplasmic reticulum [24], and TRAP1 in themitochondrial matrix [25]. Although the HSP90 bacterialhomolog HtpG is typically nonessential [26], eukaryotesrequire a functional cytoplasmic HSP90 for survival underall conditions tested [27]. Indeed whereas the term heat-shock protein is perfectly appropriate for other HSPs, it isreally somewhat of a misnomer for HSP90. In most, if not allcell types, HSP90 is actually a constitutively expressed mo-

    lecular chaperone, already extremely abundant prior to cellu-lar stress and is typically induced only a few-fold understress conditions [21]. More specifically, HSP90 is one ofthe most abundant proteins (1- 2 %) in the cytoplasm of un-stressed cells where it performs housekeeping functions,controlling the stability, maturation, activation, intracellulardisposition and proteolytic turn-over of a plethora of proteinsgenerally termed as client proteins [28-30]. HSP90 exertsits molecular chaperone activity by conformational cycles ofbinding and release which are dependent upon its ATPaseactivity [20, 31-34]. This ATP-driven conformational cycle

    is regulated by specific co-chaperones, such as HSP70, Hopimmunophilins, cdc37 and p23, that complex with HSP90and assemble into the HSP90 chaperone machinery, in ordeto assist the loading and release of client proteins [20, 22, 3132, 35].

    The broad clientele of HSP90 extends to more than 100proteins (see Dr Picards web site for an up-to-date list) andincludes molecules that are structurally and functionally diverse. Among themfeaturemultiple signal transduction molecules such as transcriptional factors, members of the Srckinase family, serine/threonine kinases, and growth factoreceptors. These key-components of the cells signallingmachinery are often activated, mutated and/or over-expressed in cancer cells and are considered responsible for theacquisition of the malignant phenotype [4, 18]. Subsequently, HSP90 is viewed as a key player in the subversionof normal cells towards transformation and therefore an exciting new target for the development of innovating molecular cancer therapeutics [16, 17, 36-42].

    HSP90: THE CANCER CHAPERONE

    HSP90 plays a pivotal role in the acquisition and mainte-nance of the malignant phenotype. Its expression in malignant cells is 2- to 10- fold higher than in normal cells [3643-45]. These higher expression levels are coupled to multiple fundamental oncogenic pathways and indicate a cruciarole associated with the development and maintenance of themalignant phenotype as well as the acquisition of drug resistant phenotypes.

    The abundance of HSP90 in tumours, in part reflects anappropriate cyto-protective stress response to the hostile hypoxic, acidotic and nutrient-deprived tumor microenvironment. Consequently, the increased activities of this molecular chaperone allow tumour cells to cope and adapt to envi-ronmental changes as well as to the imbalanced signalling

    associated with neoplastic transformation, and thereby escape apoptosis.

    Malignant transformation involves the over-expressionand/or mutation of multiple HSP90-dependent key-regulatorof cellular growth. Almost 50 proteins known to play important roles in the control of cell cycle and growth, includingreceptor protein kinases and transcription factors have beenidentified as oncogenic clients of HSP90 [17, 23, 46, 47]HSP90 is indispensable for maintaining these proteins in anactive conformation, and thereby it is considered to drive thecell to self-sufficiency in growth signals. Moreover, HSP90confers survival advantages to cancer cells through its association with important elements of apoptosis such as survivin

    [48], the Akt-survival pathway [49] and the Raf/MAPKgrowth regulatory pathway [50]. Finally HSP90 plays important anti-apoptotic roles by interfering with the intrinsiccaspase apoptotic pathway [51]. In addition to permittingautonomous growth and facilitating cell survival with respecto stressful environmental challenges, HSP90 also allowtumor cells to tolerate genetic alterations, including mutations of critical signalling molecules that would otherwise belethal [52, 53]. It not only moderates the impact of potentially lethal mutations in cancer cells, but also stabilizes andpermits the accumulation of mutant proteins, therefore func

  • 8/13/2019 Extracellular HSP90: An Emerging Target for Cancer Therapy

    3/8

    Extracellular HSP90 Current Signal Transduction Therapy, 2009, Vol. 4, No. 1 53

    tioning as a capacitor of evolution [54]. Thus, HSP90 mightalso serve as a biochemical buffer for the genetic instabilityfrequently found in cancers. As a result of this buffering ca-pacity, phenotypic diversity within the tumour cell popula-tion increases and the evolution of invasive, metastatic anddrug resistant phenotypes accelerates [18, 55].

    To summarize, one can say that HSP90 participates inalmost all the key processes of oncogenesis, such as self-sufficiency in growth signals and stabilization of mutantproteins. The involvement of this molecular chaperone in theacquisition and maintainance of the transformed phenotypeis particularly interesting and suggests that inhibiting HSP90may have a coordinated effect on all of the key alterations onwhich cancer cells depend for their growth and survival.Consequently it is no wonder that HSP90 has emerged as apromising and exciting target for the development of cancertherapeutics.

    HSP90 INHIBITORS: TARGETING SIGNAL TRANS-

    DUCTION IN CANCER

    The earliest inhibitor of HSP90 was the natural product

    geldanamycin (GA) which is a member of the family ofansamycin antibiotics. This agent has featured in the litera-ture for several years as an anti-tumor agent, and was origi-nally isolated based on its ability to promote a significantdecrease in the activity of oncogenic tyrosine kinases such asv-Src and ErbB-2 [37, 56, 57]. In 1994, Whitesell et al. [57]reported that ansamycins and GA in particular, exerts itsfunction by acting as a nucleotide mimetic and by bindingspecifically to the ATPase domain of HSP90, resulting ininhibition of its chaperone function, and consequently in theubiquitination and degradation of client proteins by the pro-teasome pathway [38, 58-60]. Despite their promising activ-ity as anti-cancer agents, these antibiotics proved to havelimited clinical potential, because of their high liver toxicity

    and/or cellular instability [61]. However, subsequent derivati-zation of GA, yielded analogues with reduced liver toxicitythat retained the potent anti-tumor activity of the parentcompound. One such example is the analogue 17-allylamino-17-demethoxygeldanamycin (17AAG) which has alreadyentered clinical trials. [62-65]. At present, several derivativesof natural products or fully synthetic small-molecule drugs

    that target HSP90 have been discovered as potential anticancer agents (see Table 2) [37, 55]. These drugs are considered as unique in that, although they are directed against aspecific molecular target, they simultaneously inhibit multiple signalling pathways by inactivating, destabilizing, andeventually leading to degradation of numerous chaperonedependent client proteins [66, 67]. As a consequence they arecompetent to mount a multi-pronged assault on cancer cells

    Moreover, although initially there were concerns that HSP90targeted drugs would attack proteins expressed both in normal and malignant cells and thus would lack specificity andcause damage to normal tissues, these fears were provedunfounded. Interestingly, exclusively tumour cells wereshown to exhibit sensitivity to HSP90 inhibition, thus lending credence to the feasibility of selectively targeting cancetissues via the pharmacological modulation of HSP90 function [68, 69]. Even more remarkably, HSP90 inhibitors sensitise tumour cells to the cytotoxic effects of a variety ofstandard therapeutic agents. Consequently they are likely tohave broad utility in combination therapy. At present, structurally unique HSP90 inhibitors are undergoing preclinicaand clinical evaluation. These anti-HSP90 drugs show grea

    promise through their potential to block a wide spectrum othe main pathways of autonomous tumor growth.

    EXTRACELLULAR FUNCTIONS OF HSP90

    While HSP90 was originally discovered and perceived aan intracellular molecular chaperone, more than two decadelater, a number of studies challenged this view. In 1986, Ul-rich et al. [70], identified HSP90 as a tumor-specific antigenlocalized on the surface of mouse cells. Since then, severaresearchers have reported the presence of this moleculachaperone on the cell surface. However these studies did noattribute precise functions to this pool of the molecule andthe best studied cases of extracellular HSP90 concerned innate and adaptive immunity [71]. More specifically, HSP90

    has been documented as a key player in antigen processingand presentation during immune responses. It has been detected on the membrane and/or culture media of antigenpresenting cells (APCs) [72] and in surface-peptide complexes [73], where it was reported to play crucial roles inanti-tumor and anti-viral responses through interaction withvarious HSP receptors on APCs, such as CD40, CD91 and

    Table 2. HSP90 Inhibitors

    Name Chemical Class Binding Site

    GA Benzoquinone ansamycin N-terminal ATP-binding pocket

    17-AAG GA-derivative N-terminal ATP-binding pocket

    17-DMAG GA-derivative N-terminal ATP-binding pocket

    Radicicol Macrolide N-terminal ATP-binding pocket

    PU24FC1 Purine scaffold N-terminal ATP-binding pocket

    CCT018159 Pyrazole N-terminal ATP-binding pocket

    Radamycin Hybrid N-terminal ATP-binding pocket

    Novobiocin Noviosylcoumarin crosslinker C-terminus

  • 8/13/2019 Extracellular HSP90: An Emerging Target for Cancer Therapy

    4/8

  • 8/13/2019 Extracellular HSP90: An Emerging Target for Cancer Therapy

    5/8

    Extracellular HSP90 Current Signal Transduction Therapy, 2009, Vol. 4, No. 1 55

    ment of signalling complexes that modulate the actin cy-toskeleton and mediate the extension of membrane protru-sions at the leading edge [94, 95]. Several molecules havebeen described as being tightly involved in the re-organi-zation of the actin cytoskeleton of a cell. These include amongothers adhesion molecules, like integrins and selectins,transmembrane receptor tyrosine kinases (EGF-R, ErbB-2),phospholipids, focal adhesion kinases (FAKs) and GTPases

    [96-98].

    The understanding of the exact molecular mechanismsunderlying the role of surface HSP90 in so complex proc-esses such as cell motility and invasion is a major challengefor the investigators. Which are the extracellular substrate(s)

    composing the clienteleof this extracellular moleculachaperone?

    Eustace et al. [79], focused on MMP-2, an enzymewhose activity in the extracellular matrix is essential for celmigration and invasion, as shown by both in vitroand in vivomodel systems [99, 100]. Extracellular HSP90was shownto associate with, and activate MMP-2, promoting thus theinvasion of fibrosarcoma cells (Fig. 2A). Furthermore, functional inhibition of the molecule by GA covalently affixed tocell-impermeable beads, inhibited cancer cell invasion. Extracellular interaction of HSP90 with MMP-2, associatedwith tumor cell invasion was recently confirmed by Yang eal. [87].

    Fig. (2). Schematic representation of 4 proposed models concerning the involvement of extracellular HSP90 in cell migration and

    invasion.

    A. HSP90 is identified extracellularly, in association with MMP2 and this interaction is suggested to be necessary for the maturation of the

    enzyme, which in turn is crucial for fibrosarcoma cell invasion [79].

    B. Surface HSP90 is shown to participate in melanoma and prostate cancer cell invasion through regulation of focal adhesion formation,

    including ECM-induced c-src/integrin association and re-organization of the actin cytoskeleton [85].

    C.Surface HSP90 is shown to be involved in breast cancer cell invasion, stimulated by the presence of HRG [81]. The model suggests that

    HSP90 exerts its function through an interaction with the extracellular domain of ErbB2, essential for the activation of the downstream signal

    transduction pathways leading to actin cytoskeletal re-arrangement and cell invasion.

    D.TGFa is shown to trigger keratinocytes to secrete HSP90 in the culture medium through the exosome pathway. Secreted HSP90 in turn

    acts as a pro-motility factor for skin cells, through its cell surface receptor LRP-1/CD91 [86].

  • 8/13/2019 Extracellular HSP90: An Emerging Target for Cancer Therapy

    6/8

    56 Current Signal Transduction Therapy,2009, Vol. 4, No. 1 Sidera and Patsavoud

    On the other hand, Tsutsumi et al. [85], suggested thatsurface HSP90 participates in cancer cell invasion throughthe signalling processes involved in the regulation of focaladhesion formation (Fig. 2B). Focal adhesions are siteswhere clusters of integrin and associated proteins such asFAK, c-Src and GTPases mediate adhesion links to the actincytoskeleton [95, 101]. By exploiting the function blockingproperties of a cell-impermeable HSP90 inhibitor named

    DMAG-N-oxide, the authors speculate that surface HSP90 isinvolved in cell motility and invasion processes, throughparticipation in leading-edge actin polymerization and focaladhesion formation. More specifically, they suggest that cellsurface HSP90 is involved in the ECM-induced c-Src/integrinassociation and the re-organization of the actin cytoskeleton.

    In another recent study, surface HSP90 involvement inbreast cancer cell invasion was attributed to a functional in-teraction of this molecule with the extracellular domain ofHER-2 [81], a protein whose intracellular kinase domain isalready known to interact with HSP90 [102-104]. This ex-tracellular interaction was shown to be crucial for maintain-ing the receptor in an active conformation, able to form het-

    erodimers with other ErbB family members and thus activatethe downstream signal transduction pathways leading to cellmotility and actin re-arrangement (Fig. 2C). Disruption ofthis extracellular interaction by the cell-impermeable mAb4C5 resulted in reduced activation of HER-2 and decreasedformation of ErbB heterodimers, accompanied by impaireddownstream kinase signalling, leading to inhibiton of actincytoskeletal re-arrangement and reduced cell invasion.

    Finally, Cheng et al. [86], demonstrated that extracellularHSP90a promoted migration of both epidermal and dermalcells through the cell surface receptor LPR-1/CD91, whichwas found to mediate HSP90a signalling. Furthermore, theauthors linked EGFR activation by TGFa to the exosomepathway leading to the secretion of HSP90a (Fig. 2D).

    The present datasupportthe idea that extracellular HSP90,interacts with a wide-range of molecules, some of them mostprobably implicated in signal transduction processes leadingto cell motility, invasion and metastasis.

    PERSPECTIVES

    The discovery of HSP90 on the surface of cancer cells, incombination with accumulating evidence reporting its in-volvement in invasion and metastasis commences a new andexciting era in the field of cancer therapeutics. Cell surfaceHSP90 provides a novel and promising molecular target forthe development of effective anti-metastatic drugs. Althoughcompounds targeting HSP90 already exist, the need for fur-

    ther development of cell-impermeable HSP90 inhibitors isemerging. In this context, the delineation of the pathwaysthrough which this protein acts and the basis for its varyingactions extracellularly , remains a great challenge.

    ABBREVIATIONS

    17AAG = 17-dimethylaminoethylamino geldanamycin

    APCs = antigen-presenting cells

    FAK = Focal Adhesion Kinase

    GA = Geldanamycin

    HRG = Heregulin

    HSPs = Heat shock proteins

    mAb = Monoclonal antibody

    MAPK = Mitogen-activated protein kinase

    PI3K = Phosphoinositide 3-kinase

    RTKs = Receptor Tyrosine KinasesREFERENCES

    [1] Vogelstein B, Kinzler KW. Cancer genes and the pathways thecontrol. Nat Med 2004; 10: 789-99.

    [2] Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000100: 57-70.

    [3] Blume-Jensen P, Hunter T. Oncogenic kinase signalling. Natur2001; 411: 355-65.

    [4] Martin GS. Cell signaling and cancer. Cancer Cell 2003; 4: 167-74[5] Downward J. The ins and outs of signalling. Nature 2001; 411

    759-62.[6] Hunter T. Signaling-2000 and beyond. Cell 2000; 100: 113-27.[7] Ritossa F. A new puffing pattern induced by temperature shock an

    DNP in Drosophila. Experientia 1962; 19: 571-573.[8] Ritossa F. Discovery of the heat shock response. Cell Stress Chap

    erones 1996; 1: 97-8.

    [9] Lindquist S. The heat-shock response. Annu Rev Biochem 198655: 1151-91.

    [10] Morimoto RI. Cells in stress: transcriptional activation of heashock genes. Science 1993; 259: 1409-10.

    [11] Parsell DA, Lindquist S. The function of heat-shock proteins istress tolerance: degradation and reactivation of damaged proteins

    Annu Rev Genet 1993; 27: 437-96.[12] Bukau B, Horwich AL. The Hsp70 and Hsp60 chaperone ma

    chines. Cell 1998; 92: 351-66.[13] Hartl FU. Molecular chaperones in cellular protein folding. Natur

    1996; 381: 571-9.[14] Hartl FU, Hayer-Hartl M. Molecular chaperones in the cytosol

    from nascent chain to folded protein. Science 2002; 295: 1852-8.[15] Calderwood SK, Khaleque MA, Sawyer DB, Ciocca DR. Hea

    shock proteins in cancer: chaperones of tumorigenesis. Trends Biochem Sci 2006; 31: 164-72.

    [16] Isaacs JS, Xu W, Neckers L. Heat shock protein 90 as a molecula

    target for cancer therapeutics. Cancer Cell 2003; 3: 213-7.[17] Maloney A, Workman P. HSP90 as a new therapeutic target for cance

    therapy: the story unfolds. Expert Opin Biol Ther 2002; 2: 3-24.[18] Whitesell L, Lindquist SL. HSP90 and the chaperoning of cancer

    Nat Rev Cancer 2005; 5: 761-72.[19] Buchner J. Hsp90 & Co. - a holding for folding. Trends Biochem

    Sci 1999; 24: 136-41.[20] Pearl LH, Prodromou C. Structure and mechanism of the Hsp90 mo

    lecular chaperone machinery. Annu Rev Biochem 2006; 75: 271-94.[21] Picard D. Heat-shock protein 90, a chaperone for folding and regu

    lation. Cell Mol Life Sci 2002; 59: 1640-8.[22] Young JC, Moarefi I, Hartl FU. Hsp90: a specialized but essentia

    protein-folding tool. J Cell Biol 2001; 154: 267-73.[23] Sreedhar AS, Kalmar E, Csermely P, Shen YF. Hsp90 isoforms

    functions, expression and clinical importance. FEBS Lett 2004562: 11-5.

    [24] Argon Y, Simen BB. GRP94, an ER chaperone with protein an

    peptide binding properties. Semin Cell Dev Biol 1999; 10: 495505.

    [25] Felts SJ, Owen BA, Nguyen P, Trepel J, Donner DB, Toft DO. Thhsp90-related protein TRAP1 is a mitochondrial protein with distinct functional properties. J Biol Chem 2000; 275: 3305-12.

    [26] Versteeg S, Mogk A, Schumann W. The Bacillus subtilis htpG

    gene is not involved in thermal stress management. Mol Gen Gene1999; 261: 582-8.

    [27] Borkovich KA, Farrelly FW, Finkelstein DB, Taulien J, LindquisS. hsp82 is an essential protein that is required in higher concentrations for growth of cells at higher temperatures. Mol Cell Bio1989; 9: 3919-30.

  • 8/13/2019 Extracellular HSP90: An Emerging Target for Cancer Therapy

    7/8

    Extracellular HSP90 Current Signal Transduction Therapy, 2009, Vol. 4, No. 1 57

    [28] Pratt WB. The role of the hsp90-based chaperone system in signaltransduction by nuclear receptors and receptors signaling viaMAP

    kinase. Annu Rev Pharmacol Toxicol 1997; 37: 297-326.[29] Pratt WB, Toft DO. Steroid receptor interactions with heat shock

    protein and immunophilin chaperones. Endocr Rev 1997; 18: 306-60.

    [30] Pratt WB, Toft DO. Regulation of signaling protein function andtrafficking by the hsp90/hsp70-based chaperone machinery. Exp

    Biol Med (Maywood) 2003; 228: 111-33.[31] Prodromou C, Panaretou B, Chohan S, Siligardi G, O'Brien R,

    Ladbury JE. The ATPase cycle of Hsp90 drives a molecular 'clamp'via transient dimerization of the N-terminal domains. EMBO J2000; 19: 4383-92.

    [32] Prodromou C, Pearl LH. Structure and functional relationships ofHsp90. Curr Cancer Drug Targets 2003; 3: 301-23.

    [33] Prodromou C, Roe SM, Piper PW, Pearl LH. A molecular clamp inthe crystal structure of the N-terminal domain of the yeast Hsp90chaperone. Nat Struct Biol 1997; 4: 477-82.

    [34] Richter K, Muschler P, Hainzl O, Reinstein J, Buchner J. Sti1 is anon-competitive inhibitor of the Hsp90 ATPase. Binding preventsthe N-terminal dimerization reaction during the atpase cycle. J BiolChem 2003; 278: 10328-33.

    [35] Pearl LH, Prodromou C. Structure and in vivofunction of Hsp90.Curr Opin Struct Biol 2000; 10: 46-51.

    [36] Bagatell R, Whitesell L. Altered Hsp90 function in cancer: aunique therapeutic opportunity. Mol Cancer Ther 2004; 3: 1021-30.

    [37] Chaudhury S, Welch TR, Blagg BS. Hsp90 as a target for drug

    development. ChemMedChem 2006; 1: 1331-40.[38] Neckers L, Mimnaugh E, Schulte TW. Hsp90 as an anti-cancer

    target. Drug Resist Updat 1999; 2: 165-172.

    [39] Scheibel T, Buchner J. The Hsp90 complex--a super-chaperonemachine as a novel drug target. Biochem Pharmacol 1998; 56: 675-82.

    [40] Smith DF, Whitesell L, Katsanis E. Molecular chaperones: biologyand prospects for pharmacological intervention. Pharmacol Rev1998; 50: 493-514.

    [41] Workman P. Pharmacogenomics in cancer drug discovery anddevelopment: inhibitors of the Hsp90 molecular chaperone. CancerDetect Prev 2002; 26: 405-10.

    [42] Workman P. Overview: translating Hsp90 biology into Hsp90drugs. Curr Cancer Drug Targets 2003; 3: 297-300.

    [43] Ferrarini M, Heltai S, Zocchi MR, Rugarli C. Unusual expression

    and localization of heat-shock proteins in human tumor cells. Int JCancer 1992; 51: 613-9.

    [44] McCarthy MM, Pick E, Kluger Y, Gould-Rothberg B, Lazova R,Camp RL. HSP90 as a marker of progression in melanoma. AnnOncol 2008; 19: 590-4.

    [45] Yufu Y, Nishimura J, Nawata H. High constitutive expression ofheat shock protein 90 alpha in human acute leukemia cells. LeukRes 1992; 16: 597-605.

    [46] Neckers L. Hsp90 inhibitors as novel cancer chemotherapeuticagents. Trends Mol Med 2002; 8: S55-61.

    [47] Neckers L, Ivy SP. Heat shock protein 90. Curr Opin Oncol 2003;15: 419-24.

    [48] Fortugno P, Beltrami E, Plescia J, Fontana J, Pradhan D, MarchisioPC. Regulation of survivin function by Hsp90. Proc Natl Acad SciUSA 2003; 100: 13791-6.

    [49] Sato S, Fujita N, Tsuruo T. Modulation of Akt kinase activity by

    binding to Hsp90. Proc Natl Acad Sci USA 2000; 97: 10832-7.[50] Schulte TW, Blagosklonny MV, Ingui C, Neckers L. Disruption of

    the Raf-1-Hsp90 molecular complex results in destabilization of

    Raf-1 and loss of Raf-1-Ras association. J Biol Chem 1995; 270:24585-8.

    [51] Pandey P, Saleh A, Nakazawa A, Kumar S, Srinivasula SM, Kumar

    V. Negative regulation of cytochrome c-mediated oligomerizationof Apaf-1 and activation of procaspase-9 by heat shock protein 90.Embo J 2000; 19: 4310-22.

    [52] Takayama S, Reed JC, Homma S. Heat-shock proteins as regula-tors of apoptosis. Oncogene 2003; 22: 9041-7.

    [53] Whitesell L, Bagatell R, Falsey R. The stress response: implica-

    tions for the clinical development of hsp90 inhibitors. Curr CancerDrug Targets 2003; 3: 349-58.

    [54] Rutherford SL, Lindquist S. Hsp90 as a capacitor for morphologi-cal evolution. Nature 1998; 396: 336-42.

    [55] Chiosis G, Vilenchik M, Kim J, Solit D. Hsp90: the vulnerablchaperone. Drug Discov Today 2004; 9: 881-8.

    [56] Sharma SV, Agatsuma T, Nakano H. Targeting of the proteichaperone, HSP90, by the transformation suppressing agent, radicicol. Oncogene 1998; 16: 2639-45.

    [57] Whitesell L, Mimnaugh EG, De Costa B, Myers CE, Neckers LMInhibition of heat shock protein HSP90-pp60v-src heteroproteincomplex formation by benzoquinone ansamycins: essential role fostress proteins in oncogenic transformation. Proc Natl Acad ScUSA 1994; 91: 8324-8.

    [58] Prodromou C, Roe SM, O'Brien R, Ladbury JE, Piper PW, PearLH. Identification and structural characterization of the ATP/ADP

    binding site in the Hsp90 molecular chaperone. Cell 1997; 90: 6575.

    [59] Schulte TW, An WG, Neckers LM. Geldanamycin-induced destabilization of Raf-1 involves the proteasome. Biochem Biophys ReCommun 1997; 239: 655-9.

    [60] Stebbins CE, Russo AA, Schneider C, Rosen N, Hartl FU, Pavletich NP. Crystal structure of an Hsp90-geldanamycin complextargeting of a protein chaperone by an antitumor agent. Cell 199789: 239-50.

    [61] Supko JG, Hickman RL, Grever MR, Malspeis L. Preclinicapharmacologic evaluation of geldanamycin as an antitumor agenCancer Chemother Pharmacol 1995; 36: 305-15.

    [62] Banerji U, O'Donnell A, Scurr M, Pacey S, Stapleton S, Asad YPhase I pharmacokinetic and pharmacodynamic study of 17allylamino, 17-demethoxygeldanamycin in patients with advanced

    malignancies. J Clin Oncol 2005; 23: 4152-61.[63] Kelland LR, Sharp SY, Rogers PM, Myers TG, Workman P. DT

    Diaphorase expression and tumor cell sensitivity to 17-allylamino

    17-demethoxygeldanamycin, an inhibitor of heat shock protein 90J Natl Cancer Inst 1999; 91: 1940-9.

    [64] Pacey S, Banerji U, Judson I, Workman P. Hsp90 inhibitors in thclinic. Handb Exp Pharmacol 2006; 172: 331-58.

    [65] Schulte TW, Neckers LM. The benzoquinone ansamycin 17allylamino-17-demethoxygeldanamycin binds to HSP90 and share

    important biologic activities with geldanamycin. Cancer Chemother Pharmacol 1998; 42: 273-9.

    [66] Neckers L. Heat shock protein 90: the cancer chaperone. J Biosc2007; 32: 517-30.

    [67] Workman P, Burrows F, Neckers L, Rosen N. Drugging the cancechaperone HSP90: combinatorial therapeutic exploitation of onco

    gene addiction and tumor stress. Ann N Y Acad Sci 2007; 1113202-16.

    [68] Kamal A, Thao L, Sensintaffar J, Zhang L, Boehm MF, Fritz LC.Ahigh-affinity conformation of Hsp90 confers tumour selectivity onHsp90 inhibitors. Nature 2003; 425: 407-10.

    [69] Neckers L, Lee YS. Cancer: the rules of attraction. Nature 2003425: 357-9.

    [70] Ullrich SJ, Robinson EA, Law LW, Willingham M, Appella E. Amouse tumor-specific transplantation antigen is a heat shockrelated protein. Proc Natl Acad Sci USA 1986; 83: 3121-5.

    [71] Basu S, Srivastava PK. Heat shock proteins: the fountainhead oinnate and adaptive immune responses. Cell Stress Chaperone2000; 5: 443-51.

    [72] Schmitt E, Gehrmann M, Brunet M, Multhoff G, Garrido C. Intracellular and extracellular functions of heat shock proteins: repercussions in cancer therapy. J Leukoc Biol 2007; 81: 15-27.

    [73] Binder RJ, Vatner R, Srivastava P. The heat-shock protein receptors: some answers and more questions. Tissue Antigens 2004; 64442-51.

    [74] Basu S, Binder RJ, Ramalingam T, Srivastava PK. CD91 is common receptor for heat shock proteins gp96, hsp90, hsp70, andcalreticulin. Immunity 2001; 14: 303-13.

    [75] Binder RJ, Srivastava PK. Essential role of CD91 in re-presentatioof gp96-chaperoned peptides. Proc Natl Acad Sci USA 2004; 1016128-33.

    [76] Triantafilou M, Triantafilou K. Heat-shock protein 70 and heatshock protein 90 associate with Toll-like receptor 4 in response to

    bacterial lipopolysaccharide. Biochem Soc Trans 2004; 32: 636-9.

    [77] Sapozhnikov AM, Ponomarev ED, Tarasenko TN, Telford WGSpontaneous apoptosis and expression of cell surface heat-shock

    proteins in cultured EL-4 lymphoma cells. Cell Prolif 1999; 32363-78.

  • 8/13/2019 Extracellular HSP90: An Emerging Target for Cancer Therapy

    8/8

    58 Current Signal Transduction Therapy,2009, Vol. 4, No. 1 Sidera and Patsavoud

    [78] Liao DF, Jin ZG, Baas AS, Daum G, Gygi SP, Aebersold R. Purifi-cation and identification of secreted oxidative stress-induced fac-

    tors from vascular smooth muscle cells. J Biol Chem 2000; 275:189-96.

    [79] Eustace BK, Sakurai T, Stewart JK, Yimlamai D, Unger C, Zehet-meier C. Functional proteomic screens reveal an essential extracel-lular role for hsp90 alpha in cancer cell invasiveness. Nat Cell Biol2004; 6: 507-14.

    [80] Sidera K, Samiotaki M, Yfanti E, Panayotou G, Patsavoudi E.Involvement of cell surface HSP90 in cell migration reveals a novel

    role in the developing nervous system. J Biol Chem 2004; 279:45379-88.

    [81] Sidera K, Gaitanou M, Stellas D, Matsas R, Patsavoudi E. A criticalrole for HSP90 in cancer cell invasion involves interaction with the ex-tracellular domain of HER-2. J Biol Chem 2008; 283: 2031-41.

    [82] Becker B, Multhoff G, Farkas B, Wild PJ, Landthaler M, Stolz W.Induction of Hsp90 protein expression in malignant melanomas andmelanoma metastases. Exp Dermatol 2004; 13: 27-32.

    [83] Stellas D, Karameris A, Patsavoudi E. Monoclonal antibody 4C5immunostains human melanomas and inhibits melanoma cell inva-sion and metastasis. Clin Cancer Res 2007; 13: 1831-8.

    [84] Li W, Li Y, Guan S, Fan J, Cheng CF, Bright AM. Extracellularheat shock protein-90alpha: linking hypoxia to skin cell motilityand wound healing. Embo J 2007; 26: 1221-33.

    [85] Tsutsumi S, Scroggins B, Koga F, Lee MJ, Trepel J, Felts S. Asmall molecule cell-impermeant Hsp90 antagonist inhibits tumorcell motility and invasion. Oncogene 2008; 27: 2478-87.

    [86] Cheng CF, Guan S, Fedesco M, Fan J, Li Y, Bandyopadhyay B.TGF{alpha}-Stimulated Secretion of HSP90{alpha}: Using LRP-1/CD91 Receptor To Promote Human Skin Cell Migration Against

    TGF{beta}-Rich Environment In Wound Healing. Mol Cell Biol2008; 28: 3344-58.

    [87] Yang Y, Rao R, Shen J, Tang Y, Fiskus W, Nechtman J. Role ofacetylation and extracellular location of heat shock protein 90alphain tumor cell invasion. Cancer Res 2008; 68: 4833-42.

    [88] Tsutsumi S, Neckers L. Extracellular heat shock protein 90: a role

    for a molecular chaperone in cell motility and cancer metastasis.Cancer Sci 2007; 98: 1536-9.

    [89] Bausero MA, Page DT, Osinaga E, Asea A. Surface expression ofHsp25 and Hsp72 differentially regulates tumor growth and metas-tasis. Tumour Biol 2004; 25: 243-51.

    [90] El Hamidieh A, Siganou A, Grammatikakis N, Patsavoudi E.

    Cdc37 is localized at the cell surface and is possibly involved incancer cell invasion. FEBS J 2008; 275: 201.

    [91] Eustace BK, Jay DG. Extracellular roles for the molecular chaperone, hsp90. Cell Cycle 2004; 3: 1098-100.

    [92] Farkas B, Hantschel M, Magyarlaki M, Becker B, Scherer K, Landthaler M. Heat shock protein 70 membrane expression and melanoma-associated marker phenotype in primary and metastatimelanoma. Melanoma Res 2003; 13: 147-52.

    [93] Lee KJ, Kim YM, Kim DY, Jeoung D, Han K, Lee ST. Release oheat shock protein 70 (Hsp70) and the effects of extracellulaHsp70 on matric metalloproteinase-9 expression in human monocytic U937 cells. Exp Mol Med 2006; 38: 364-74.

    [94] Lauffenburger DA, Horwitz AF. Cell migration: a physically integrated molecular process. Cell 1996; 84: 359-69.

    [95] Webb DJ, Parsons JT, Horwitz AF. Adhesion assembly, disassembly and turnover in migrating cells -- over and over and over againNat Cell Biol 2002; 4: E97-100.

    [96] Feldner JC, Brandt BH. Cancer cell motility--on the road from cerbB-2 receptor steered signaling to actin reorganization. Exp CelRes 2002; 272: 93-108.

    [97] Friedl P, Wolf K. Tumour-cell invasion and migration: diversityand escape mechanisms. Nat Rev Cancer 2003; 3: 362-74.

    [98] Miranti CK, Brugge JS. Sensing the environment: a historical perspective on integrin signal transduction. Nat Cell Biol 2002; 4E83-90.

    [99] Murphy G, Gavrilovic J. Proteolysis and cell migration: creating

    path? Curr Opin Cell Biol 1999; 11: 614-21.[100] Seiki M. The cell surface: the stage for matrix metalloproteinas

    regulation of migration. Curr Opin Cell Biol 2002; 14: 624-32.

    [101] Wozniak MA, Modzelewska K, Kwong L, Keely PJ. Focal adhesion regulation of cell behavior. Biochim Biophys Acta 20041692: 103-19.

    [102] Citri A, Kochupurakkal BS, Yarden Y. The achilles heel of ErbB2/HER2: regulation by the Hsp90 chaperone machine and potentiafor pharmacological intervention. Cell Cycle 2004; 3: 51-60.

    [103] Mimnaugh EG, Chavany C, Neckers L. Polyubiquitination anproteasomal degradation of the p185c-erbB-2 receptor proteintyrosine kinase induced by geldanamycin. J Biol Chem 1996; 271

    22796-801.[104] Xu W, Mimnaugh E, Rosser MF, Nicchitta C, Marcu M, Yarden Y

    Sensitivity of mature Erbb2 to geldanamycin is conferred by itkinase domain and is mediated by the chaperone protein Hsp90. Biol Chem 2001; 276: 3702-8.

    Received: July 10, 2008 Revised: August 27, 2008 Accepted: September 09, 2008