1,209
Views
63
CrossRef citations to date
0
Altmetric
Original Article

Heat shock proteins in breast cancer progression–A suitable case for treatment?

Pages 681-685 | Received 11 Feb 2010, Accepted 28 Apr 2010, Published online: 23 Jul 2010

Abstract

Heat shock proteins (HSP) and heat shock factor 1 (HSF1), key factors in the heat shock response (HSR) have been implicated in the etiology of breast cancer. At least two members of the HSP family, Hsp27 and Hsp70 undergo significant increases in cellular concentration during the transformation of mammary cells. These changes result in HSP-mediated inhibition of tumour cell inactivation through blockade of the apoptosis and replicative senescence pathways. The increases in HSP thus mediate two of the common hallmarks of cancer and favour cell birth over cell death. In addition, Hsp90 plays a role in facilitating transformation by stabilising the mutated and over-expressed oncoproteins found in breast tumours, and permitting the activation of growth stimulatory and transforming pathways in the absence of growth factors. HSF1 appears to play a similar role as a facilitator of transformation in mammary carcinoma. Induction of some facets of the HSR in breast cancer cells therefore leads to growth stimulation and inhibits cell death. Pharmacological targeting of HSP and HSF1 is therefore indicated and in the case of Hsp90, inhibitory drugs are undergoing clinical trial for treatment of breast carcinoma and other cancers.

Introduction

Heat shock proteins were first discovered as mediators of resistance to hyperthermia in all cellular organisms Citation[1]. They have subsequently been characterised as members of the molecular chaperones, a group of proteins that play essential roles in the correct folding of a large proportion of cellular proteins Citation[2], Citation[3]. The development of thermotolerance involves this property of HSPs and is associated with the resolution of toxic protein aggregates that accumulate in hyperthermia Citation[4–6]. Thermotolerance involves several mechanisms including: (1) the refolding of some proteins in pathway facilitated by HSPs as well as (2) degradation of proteins that are not refolded through the proteasome pathway, another HSP-mediated process Citation[7]. In addition, HSPs participate directly in cell survival during hyperthermia by inhibiting programmed cell death and cell senescence Citation[8–11]. The transcription of HSP genes is regulated by transcription factor HSF1, that senses cellular exposure to stress and turns on rapid induction of HSPs Citation[12], Citation[13]. These properties of the HSR appear to have been hijacked during malignant progression and aid in the development of cancer Citation[14], Citation[15]. HSP levels become elevated in a wide spectrum of malignant cells including mammary carcinoma cells Citation[16]. Hsp27 and Hsp70 appear to foster mammary tumorigenesis by inhibiting apoptosis and senescence. Hsp90 appears to play a key role in facilitating tumour progression by chaperoning the mutated and over-expressed oncogenes that fuel transformation and tumour progression Citation[17], Citation[18]. Indeed, Hsp90 may contribute to the evolution of treatment-resistant cell populations by permitting the emergence of variant proteins that can overcome the stresses of cancer therapy Citation[17–20], Citation[21]. It has also been shown recently that the mitochondrial Hsp90 family member TRAP-1 plays a versatile role in transformation by mediating membrane transitions within this organelle and inhibiting apoptosis at source Citation[22]. Mitochondria possess a subclass of HSPs including the Hsp70 homologue mortalin and TRAP-1 as well as Hsp60, each of which appear important in progression of a wide class of cancers Citation[22], Citation[23]. In addition, recent studies indicate a key role for HSF1 in breast carcinoma. HSF1 may function in breast cancer progression by inducing HSPs Citation[21]. However, HSF1 appears to play additional roles in addition to HSP induction, including activation of metastasis by the silencing of anti-metastatic processes, activating pro-malignant signalling cascades and regulating the mitotic spindle checkpoint Citation[24–26]. These multiple components of the heat shock response appear to be utilised in tumorigenesis in order for cells to escape the pathways of tumour suppression, to promote evolution into advanced and treatment-resistant modes, and to facilitate metastasis. They would therefore seem suitable candidates for molecular targeting (summarised in ).

Figure 1. Role of the HSR in breast cancer. Activated nuclear HSF1 is depicted leading to the expression of Hsp27 and Hsp70 that block programmed cell death and Hsp90 that fosters the accumulation of oncoproteins. Oncoproteins may be mutated (oncogeme) or over-expressed [oncogene, oncogene, oncogene]. Apoptosis is inhibited by Hsp27 and Hsp70 in the cytosol or by TRAP-1 and/or mortalin in the mitochondria.

Figure 1. Role of the HSR in breast cancer. Activated nuclear HSF1 is depicted leading to the expression of Hsp27 and Hsp70 that block programmed cell death and Hsp90 that fosters the accumulation of oncoproteins. Oncoproteins may be mutated (oncogeme) or over-expressed [oncogene, oncogene, oncogene]. Apoptosis is inhibited by Hsp27 and Hsp70 in the cytosol or by TRAP-1 and/or mortalin in the mitochondria.

Targeting Hsp90

Hsp90 has emerged as important agent in cancer therapy and a number of drug classes (including the ansamycins and novobiocin homologues) appear able to target its ATP binding domain and inhibit activity Citation[27], Citation[28]. Binding and hydrolysis of ATP is essential for its molecular chaperone function Citation[29]. Hsp90 is not generally expressed at elevated levels in cancer although its basal levels are already high in the majority of cells. However Hsp90 activity appears essential for growth of breast and other cancers due to properties that include its ability to chaperone a wide spectrum of oncogenic proteins. These include proteins important in breast cancer progression such as Her2/neu and c-src Citation[17], Citation[18]. Many such proteins become over-expressed or mutated in mammary cancer progression and such tumours appear to be dependent on, or ‘addicted to’, Hsp90 to maintain the levels of these proteins and drugs that inactivate the chaperone target this dependency Citation[17], Citation[18]. Hsp90 drugs have the advantage of multi-targeting, in that many of the oncogenic proteins depend on Hsp90 activity and a dynamic mechanism of inhibiting the evolution of treatment resistant cells Citation[17], Citation[18], Citation[30]. Hsp90 is known to maintain pools of mutant/polymorphic molecules in the cell that could potentially be recruited for the evolution of new traits Citation[19]. Inhibition of the protein is predicted to cause loss of the mutant pool and inability to respond to changes in microenvironment and to anticancer treatments. Clinical trials are beginning on a number of the ansamycin-derived Hsp90 drugs including 17-AAG and others Citation[31].

Hsp90 carries out its molecular chaperone role in cells in association with a cohort of accessory proteins called co-chaperones. One of these proteins in particular, p50/Cdc37 is expressed to high level in multiple cancers and is required in the maturation of a wide spectrum of oncogenic protein kinases. Targeting Cdc37 in cancer has been attempted and appears highly effective at least in prostate cancer Citation[32]. Cdc37 offers the potential of a therapeutic gain due to its elevated expression in cancer and experiments are underway to target the protein in breast cancer Citation[33].

A role for Hsp70

Hsp70, like Hsp90 mediates the chaperoning of a wide spectrum of cellular proteins in an ATP-dependent manner Citation[3]. Hsp70 is expressed at high levels in a wide spectrum of cancer cells and is induced through activation of the Her2/neu pathway that is important in advanced breast cancer Citation[21]. Hsp70 is involved in the early stages of protein folding, chaperoning a proportion of polypeptides undergoing synthesis on ribosomes before passing on some clients to Hsp90. Hsp70 does not, however seem required for the long-term stabilisation of a wide spectrum of clients as observed with Hsp90. Thus Hsp70 addiction in cancer cells is not as commonly observed as with the more ubiquitous Hsp90. Hsp70 appears to function in breast cancer due to its ability to inhibit programmed cell death and senescence when expressed at high levels; important features of malignant transformation Citation[10], Citation[11]. Thus, targeting Hsp70 can lead to cell inactivation by permitting programmed cell death. However, unlike Hsp90, really effective drugs are not currently available for targeting the ATP binding domain of Hsp70. However, since Hsp70 blocks apoptosis at the post-mitochondrial level by inactivating the apoptosome as well as the apoptosis inducing factor (AIF), strategies targeting Hsp70 appear to be effective in overcoming tumour cell resistance. It has recently been shown that decoy targets of Hsp70 derived from AIF can sensitise cancer cells to apoptosis induction by neutralising Hsp70 functions. AIF-derived peptides mimic a domain of the AIF protein (amino acids 150 to 228) required for Hsp70 binding Citation[34]. Hence, they bind to Hsp70 but lack an autonomous pro-apoptotic function. Experiments using different cancer cell lines (leukaemia, colon cancer, breast cancer and cervical cancer) demonstrate that several among these peptides strongly increase the sensitivity to chemotherapy in vitro. This effect was related to their ability to neutralise endogenous Hsp70, because this pro-apoptotic activity was lost in Hsp70-negative cells Citation[35].

The small HSP family, especially Hsp27

Hsp27 is expressed to high levels in breast carcinoma and many other types of cancer, compared to normal cells in which expression is moderate in the absence of stress Citation[36]. Hsp27 is a member of the small HSP (sHSP) family and is a potent mediator of protein folding Citation[11]. The sHSP are highly pleiotropic molecules both in terms of molecular function and cell biology properties. In breast cancer, Hsp27 exerts pro-malignant effects largely due to its properties of inhibiting programmed cell death and senescence Citation[37], Citation[38]. Hsp27 is activated in stress both by transcriptional activation and post-translational modification (phosphorylation) downstream of the p38 MAPK stress kinase pathway, although it is not clear which of these processes is involved in Hsp27 up-regulation in cancer Citation[39]. However, HSF1 becomes activated in breast cancer and Hsp27 levels may increase secondarily to this change. Hsp27 mediates its molecular activities after activation through phosphorylation-dependent changes in oligomerisation that are involved in its protein folding and cell regulatory functions Citation[39]. Unlike the other HSPs, the sHSPs do not bind ATP, a property that may make this molecule problematic for targeting with small compounds. Many effective anti-cancer agents are aimed at the ATP binding domains of protein kinases or other molecular chaperones Citation[17], Citation[40].

HSF1

As the central regulator of HSP expression, HSF1 would seem a focal point in designing anti-breast cancer approaches. HSF1 inactivation has been shown to inhibit the progression of a wide spectrum of cancers Citation[26], Citation[41]. In addition, activation of the Her2/neu pathway has been shown to induce HSF1 and HSP expression and render cells resistant to spontaneous and drug-induced programmed cell death Citation[21]. HSF1 may also mediate tumorigenic effects through an alternative pathway involving the recruitment of the pro-metastatic gene co-repressor MTA1 which inhibits the expression of (oestrogen-induced) anti-metastatic genes Citation[24]. MTA1 thus appears to stand at the crossroads between oestrogen and Her2/neu regulated signalling, encouraging metastasis and HSF1 appears complicit in these effects. HSF1 also possesses additional properties in cancer including the enhancement of pro-malignant signalling through the ERK, PKA and TOR pathways Citation[26]. Chemical inhibitors of HSF1 activation have been described, including genistein, a compound which has the interesting property of synergising with hyperthermia in cancer regression in vivo Citation[42], Citation[43]. However, genistein is a highly pleiotropic molecule with many potential targets in cancer cells and it is not clear what proportion of its effects on tumour growth involve HSF1 inhibition. Additional HSF1 inhibitors include KNK437 and Triptolide and information on their specificity, effectiveness in treatment of breast cancer is awaited Citation[44], Citation[45]. In addition, an inhibitor of the heat shock response termed emunin has been described Citation[46]. Emunin inhibits HSP expression in cancer cells by unknown mechanisms and potentiates mammary tumour cell killing by agents such as Hsp90 inhibitors and proteasome inhibitors Citation[46]. It is thus apparent that chemical targeting of HSF1 is at an early stage in development and advances may be expected.

Discussion

The heat shock response thus appears to have become ‘hijacked’ in the processes of mammary cell transformation and progression. Transformation is accompanied by HSF1 activation and involves multiple downstream effects of this molecule (). These effects include the induction of Hsp27 and Hsp70, powerful antagonists of programmed cell death. However, in addition to HSR activation in cancer, tumour cells also appear to become ‘addicted’ to basal expression of Hsp90 and its co-chaperones. Hsp90 appears to be required in order to permit accumulation of over-expressed and mutated oncogenes. However, other mechanisms may also be involved. At least in the case of Hsp90, effective classes of inhibitory drugs are available and are undergoing clinical trial in breast cancer treatment. For other arms of the HSR, agents are currently being developed although it is too early to assess their worth in treatment of breast cancer.

Cancer research is currently undergoing a major re-evaluation as its cell biological parameters come under increased scrutiny. Understanding the relative roles of cancer stem cells, progenitor cells and differentiated cancer cells in treatment response may alter our assessments of the molecular biology studies carried out on bulk populations of cells in recent years Citation[47], Citation[48]. The role of HSF1 and HSPs in breast cancer stem cell growth, survival and response to treatment is currently unknown but may be highly significant in future approaches to breast cancer treatment.

Declaration of interest: This work was supported by NIH research grants R01CA119045, R01CA047407, R01CA094397. The authors report no conflicts of interest. The authors alone are responsible for the content and writing of the paper.

References

  • Calderwood SK, Ciocca DR. Heat shock proteins: Stress proteins with Janus-like properties in cancer. Int J Hyperthermia 2008; 24: 31–39
  • Lindquist S, Craig EA. The heat shock proteins. Ann Rev Genet 1988; 22: 631–637
  • Bukau B, Weissman J, Horwich A. Molecular chaperones and protein quality control. Cell 2006; 125: 443–451
  • Subjeck JR, Sciandra JJ, Johnson RJ. Heat shock proteins and thermotolerance: A comparison of induction kinetics. Br J Radiol 1982; 55: 579–584
  • Landry J, Bernier D, Chretien P, Nicole LM, Tanguay RM, Marceau N. Synthesis and degradation of heat shock proteins during the development and decay of thermotolerance. Cancer Research 1982; 42: 2457–2461
  • Li GC, Werb Z. Correlation between the synthesis of heat shock proteins and the development of thermotolerance in CHO fibroblasts. Proc Natl Acad Sci USA 1982; 79: 3218–3222
  • Calderwood SK, Murshid A, Prince T. The shock of aging: Molecular chaperones and the heat shock response in longevity and aging–A mini-review. Gerontology 2009; 55: 550–558
  • Tang D, Khaleque MA, Jones EL, Theriault JR, Li C, Wong WH, Stevenson MA, Calderwood SK. Expression of heat shock proteins and heat shock protein messenger ribonucleic acid in human prostate carcinoma in vitro and in tumors in vivo. Cell Stress Chaperones 2005; 10: 46–58
  • Jones EL, Zhao MJ, Stevenson MA, Calderwood SK. The 70 kilodalton heat shock protein is an inhibitor of apoptosis in prostate cancer. Int J Hyperthermia 2004; 20: 835–849
  • Gabai VL, Meriin AB, Yaglom JA, Volloch VZ, Sherman MY. Role of Hsp70 in regulation of stress-kinase JNK: Implications in apoptosis and aging. FEBS Lett 1998; 438: 1–4
  • Garrido C, Brunet M, Didelot C, Zermati Y, Schmitt E, Kroemer G. Heat shock proteins 27 and 70: Anti-apoptotic proteins with tumorigenic properties. Cell Cycle 2006; 5: 2592–2601
  • Sorger PK, Pelham HRB. Yeast heat shock factor is an essential DNA-binding protein that exhibits temperature-dependent phosphorylation. Cell 1988; 54: 855–864
  • Wu C. Heat shock transcription factors: Structure and regulation. Ann Rev Cell Dev Biol 1995; 11: 441–469
  • Calderwood SK, Khaleque MA, Sawyer DB, Ciocca DR. Heat shock proteins in cancer: Chaperones of tumorigenesis. Trends Biochem Sci 2006; 31: 164–172
  • Ciocca DR, Calderwood SK. Heat shock proteins in cancer: Diagnostic, prognostic, predictive, and treatment implications. Cell Stress Chaperones 2005; 10: 86–103
  • Ciocca DR, Vargas-Roig LM. Hsp27 as a prognostic and predictive factor in cancer. Prog Mol Subcell Biol 2002; 28: 205–218
  • Kamal A, Thao L, Sensintaffar J, Zhang L, Boehm MF, Fritz LC, Burrows FJ. A high-affinity conformation of Hsp90 confers tumour selectivity on Hsp90 inhibitors. Nature 2003; 425: 407–410
  • Neckers L, Lee YS. Cancer: The rules of attraction. Nature 2003; 425: 357–359
  • Rutherford SL, Lindquist S. Hsp90 as a capacitor for morphological evolution. Nature 1998; 396: 336–342
  • Sangster TA, Salathia N, Lee HN, Watanabe E, Schellenberg K, Morneau K, Wang H, Undurraga S, Queitsch C, Lindquist S. HSP90-buffered genetic variation is common in Arabidopsis thaliana. Proc Natl Acad Sci USA 2008; 105: 2969–2974
  • Khaleque MA, Bharti A, Sawyer D, Gong J, Benjamin IJ, Stevenson MA, Calderwood SK. Induction of heat shock proteins by heregulin beta1 leads to protection from apoptosis and anchorage-independent growth. Oncogene 2005; 24: 6564–6573
  • Leav I, Plescia J, Goel HL, Li J, Jiang Z, Cohen RJ, Languino LR, Altieri DC. Cytoprotective mitochondrial chaperone TRAP-1 as a novel molecular target in localized and metastatic prostate cancer. Am J Pathol, 176: 393–401
  • Wadhwa R, Taira K, Kaul SC. An Hsp70 family chaperone, mortalin/mthsp70/PBP74/Grp75: What, when, and where?. Cell Stress Chaperones 2002; 7: 309–316
  • Khaleque MA, Bharti A, Gong J, Gray PJ, Sachdev V, Ciocca DR, Stati A, Fanelli M, Calderwood SK. Heat shock factor 1 represses estrogen-dependent transcription through association with MTA1. Oncogene 2008; 27: 1886–1893
  • Wang Y, Theriault JR, He H, Gong J, Calderwood SK. Expression of a dominant negative heat shock factor-1 construct inhibits aneuploidy in prostate carcinoma cells. J Biol Chem 2004; 279: 32651–32659
  • Dai C, Whitesell L, Rogers AB, Lindquist S. Heat shock factor 1 is a powerful multifaceted modifier of carcinogenesis. Cell 2007; 130: 1005–1018
  • Kawabe M, Mandic M, Taylor JL, Vasquez CA, Wesa AK, Neckers LM, Storkus WJ. Heat shock protein 90 inhibitor 17-dimethylaminoethylamino-17-demethoxygeldanamycin enhances EphA2+ tumor cell recognition by specific CD8+ T cells. Cancer Res 2009; 69: 6995–7003
  • Tsutsumi S, Beebe K, Neckers L. Impact of heat-shock protein 90 on cancer metastasis. Future Oncol 2009; 5: 679–688
  • Neckers L, Ivy SP. Heat shock protein 90. Curr Opin Oncol 2003; 15: 419–424
  • Whitesell L, Lindquist SL. HSP90 and the chaperoning of cancer. Nat Rev Cancer 2005; 5: 761–772
  • Kim YS, Alarcon SV, Lee S, Lee MJ, Giaccone G, Neckers L, Trepel JB. Update on Hsp90 inhibitors in clinical trial. Curr Top Med Chem 2009; 9: 1479–1492
  • Gray PJ, Jr, Stevenson MA, Calderwood SK. Targeting Cdc37 inhibits multiple signaling pathways and induces growth arrest in prostate cancer cells. Cancer Res 2007; 67: 11942–11950
  • Gray PJ, Jr, Prince T, Cheng J, Stevenson MA, Calderwood SK. Targeting the oncogene and kinome chaperone CDC37. Nat Rev Cancer 2008; 8: 491–495
  • Gurbuxani S, Schmitt E, Cande C, Parcellier A, Hammann A, Daugas E, Kouranti I, Spahr C, Pance A, Kroemer G, et al. Heat shock protein 70 binding inhibits the nuclear import of apoptosis-inducing factor. Oncogene 2003; 22: 6669–6678
  • Schmitt E, Parcellier A, Gurbuxani S, Cande C, Hammann A, Morales MC, Hunt CR, Dix DJ, Kroemer RT, Giordanetto F, et al. Chemosensitization by a non-apoptogenic heat shock protein 70-binding apoptosis-inducing factor mutant. Cancer Res 2003; 63: 8233–8240
  • Ciocca DR, Oesterreich S, Chamness GC, McGuire WL, Fuqua SA. Biological and clinical implications of heat shock protein 27,000 (Hsp27): A review. J Natl Cancer Inst 1993; 85: 1558–1570
  • Sherman MY, Gabai V, O'Callaghan C, Yaglom J. Molecular chaperones regulate p53 and suppress senescence programs. FEBS Lett 2007; 581: 3711–3715
  • Sherman MY, Goldberg AL. Cellular defenses against unfolded proteins: A cell biologist thinks about neurodegenerative diseases. Neuron 2001; 29: 15–32
  • Brunet Simioni M, De Thonel A, Hammann A, Joly AL, Bossis G, Fourmaux E, Bouchot A, Landry J, Piechaczyk M, Garrido C. Heat shock protein 27 is involved in SUMO-2/3 modification of heat shock factor 1 and thereby modulates the transcription factor activity. Oncogene 2009; 28: 3332–3344
  • Buchdunger E, Matter A, Druker BJ. Bcr-Abl inhibition as a modality of CML therapeutics. Biochim Biophys Acta 2001; 1551: M11–18
  • Min JN, Huang L, Zimonjic DB, Moskophidis D, Mivechi NF. Selective suppression of lymphomas by functional loss of Hsf1 in a p53-deficient mouse model for spontaneous tumors. Oncogene 2007; 26: 5086–5097
  • Asea A, Ara G, Teicher BA, Stevenson MA, Calderwood SK. Effects of the flavonoid drug quercetin on the response of human prostate tumours to hyperthermia in vitro and in vivo. Int J Hyperthermia 2001; 17: 347–356
  • Price BD, Calderwood SK. Ca2+ is essential for multistep activation of the heat shock factor in permeabilized cells. Mol Cell Biol 1991; 11: 3365–3368
  • Martin CA, Kurkowski DL, Valentino AM, Santiago-Schwarz F. Increased intracellular, cell surface, and secreted inducible heat shock protein 70 responses are triggered during the monocyte to dendritic cell (DC) transition by cytokines independently of heat stress and infection and may positively regulate DC growth. J Immunol 2009; 183: 388–399
  • Koishi M, Yokota S, Mae T, Nishimura Y, Kanamori S, Horii N, Shibuya K, Sasai K, Hiraoka M. The effects of KNK437, a novel inhibitor of heat shock protein synthesis, on the acquisition of thermotolerance in a murine transplantable tumor in vivo. Clin Cancer Res 2001; 7: 215–219
  • Zaarur N, Gabai VL, Porco J, Calderwood SK, Sherman MY. Novel inhibitors of heat shock protein expression sensitize cancer cells to proteasome and hsp90 inhibitors. Cancer Res, 66: 1783–1791
  • Reya T, Clevers H. Wnt signalling in stem cells and cancer. Nature 2005; 434: 843–850
  • Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature 2001; 414: 105–111

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.