Skip to main content
  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

  • Register
  • Log in
  • Log out
  • My Cart
Advertisement

Main menu

  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
    • CME
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • CCR Focus Archive
    • Meeting Abstracts
    • Collections
      • Clinical Trials
      • Immunotherapy: Facts and Hopes
      • Editors' Picks
  • For Authors
    • Information for Authors
    • Author Services
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • OnlineFirst
    • Editors' Picks
    • Citation
    • Author/Keyword
  • News
    • Cancer Discovery News
  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

User menu

  • Register
  • Log in
  • Log out
  • My Cart

Search

  • Advanced search
Clinical Cancer Research
Clinical Cancer Research

Advanced Search

  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
    • CME
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • CCR Focus Archive
    • Meeting Abstracts
    • Collections
      • Clinical Trials
      • Immunotherapy: Facts and Hopes
      • Editors' Picks
  • For Authors
    • Information for Authors
    • Author Services
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • OnlineFirst
    • Editors' Picks
    • Citation
    • Author/Keyword
  • News
    • Cancer Discovery News
Molecular Pathways

Molecular Pathways: Targeting Hsp90—Who Benefits and Who Does Not

Maurizio Scaltriti, Shaheenah Dawood and Javier Cortes
Maurizio Scaltriti
Authors' Affiliations: 1Massachusetts General Hospital Cancer Center, Massachusetts General Hospital; 2Harvard Medical School, Boston, Massachusetts;3Medical Oncology Department, Dubai Hospital, UAE Medical, Dubai Health Authority, Dubai; and 4Oncology Department, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, SpainAuthors' Affiliations: 1Massachusetts General Hospital Cancer Center, Massachusetts General Hospital; 2Harvard Medical School, Boston, Massachusetts;3Medical Oncology Department, Dubai Hospital, UAE Medical, Dubai Health Authority, Dubai; and 4Oncology Department, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Shaheenah Dawood
Authors' Affiliations: 1Massachusetts General Hospital Cancer Center, Massachusetts General Hospital; 2Harvard Medical School, Boston, Massachusetts;3Medical Oncology Department, Dubai Hospital, UAE Medical, Dubai Health Authority, Dubai; and 4Oncology Department, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Javier Cortes
Authors' Affiliations: 1Massachusetts General Hospital Cancer Center, Massachusetts General Hospital; 2Harvard Medical School, Boston, Massachusetts;3Medical Oncology Department, Dubai Hospital, UAE Medical, Dubai Health Authority, Dubai; and 4Oncology Department, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI: 10.1158/1078-0432.CCR-11-2138 Published September 2012
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Many kinases and hormone receptors, important for cancer cell proliferation and survival, bind to and are dependent on the Hsp90 cycle for their folding and maturation. This provides the rationale for the development of small-molecule ATP competitors that, inhibiting Hsp90 function, lead to degradation of the "client" proteins. After continual efforts to improve the pharmacologic properties and the tolerability of these molecules, several Hsp90 inhibitors have exhibited activity in both preclinical models and in the clinical setting. As is the case with many other targeted agents, patient selection seems to be the major limitation to the success of these compounds. ERBB2-positive patients with breast cancer are exquisitely sensitive to Hsp90 inhibition. This is because ERBB2 is indispensable for growth and survival of this subtype of cancer, and at the same time ERBB2 is a client protein strictly dependent on Hsp90 for its maturation and stability. Extensive preclinical work identifying other ERBB-like client proteins will likely lead to the ability to enhance selection of appropriate patients for enrollment in more rational clinical trials. Hsp90 inhibition has also been reported to synergize with other therapeutic agents. Several ongoing studies testing different combinations of Hsp90 inhibitors with other targeted agents will confirm whether Hsp90 inhibition can potentiate the efficacy of targeted therapy and/or prevent the emergence of drug resistance. Clin Cancer Res; 18(17); 4508–13. ©2012 AACR.

Background

Hsp90 is a ubiquitous molecular chaperone required for correct folding and maturation of a variety of cellular proteins (1, 2). Hsp90, a protein that is highly conserved from bacteria to mammals, is documented to interact with more than 200 different "client" proteins (3, 4). The rationale that led to the development of Hsp90 inhibitors is as follows: (i) Hsp90 itself is reported to be specifically overexpressed in tumor cells (5–8) in a setting where it would also have more affinity for small-molecule Hsp90 inhibitors (9); (ii) Hsp90 client proteins are frequently oncogenes, mutated, or overexpressed, to which cancer cells are addicted for survival and proliferation (2, 10); and (iii) inhibition of Hsp90 leads to degradation of the client protein (11).

One of the Hsp90 client proteins is the progesterone receptor, which has been used extensively as a model to understand the Hsp90 cycle (refs., 12, 13; Fig 1). Like the other client proteins, the progesterone receptor has no precise sequence consensus for the identification and recruitment (through the interaction with several cochaperones) into the Hsp90 cycle. In fact, the binding of each client protein seems to be conformational, related to a sequence contained in amino-acidic regions that encode a particular conformation (14–16).

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

The Hsp90 cycle. Folding and maturation of the progesterone receptor is a model of how Hsp90 client proteins interact with Hsp90 and its cochaperones. The interaction between the cochaperone Hsp40 and the client protein leads to the recruitment of ATP-bound cochaperone Hsp70 that, following ATP hydrolysis, binds to another complex formed by the adaptor protein Hop and Hsp90 dimers. Binding of ATP to the Hsp90 dimers forces this intermediate complex to assume a closed conformation that facilitates the interaction with the cochaperone p23 and the dissociation from Hop. Hsp90 ATP competitors bind to the ATP-binding pocket of Hsp90 at this stage and impede hydrolysis of ATP to ADP, necessary for intermediate complex disassembly and release of the mature client protein. Nonmature proteins are rapidly degraded.

The first step of the Hsp90 cycle is the binding between the cochaperone Hsp40 and the client protein. This interaction leads to the recruitment of ATP-bound cochaperone Hsp70, only weakly bound at this stage. The formation of this complex triggers ATP hydrolysis, which provokes a conformational change in Hsp70 increasing its affinity for the substrate (12, 17). ADP-bound Hsp70 can interact with Hop, an adaptor protein that simultaneously binds Hsp90 dimers, promoting the formation of an "intermediate complex" (18–21). ATP binding of the intermediate complex is also responsible for the progression to the next step of the Hsp90 cycle, again inducing a conformational change. ATP binds to Hsp90 dimers, forcing them to assume a "closed" conformation, which in turn facilitates the interaction with the cochaperone p23, and the dissociation from Hop (22–25). Subsequently, hydrolysis of ATP to ADP leads to disassembly of the complex and release of the mature client protein. These ATP-dependent conformational arrangements, crucial for the correct folding and consequent stability of the client proteins (26, 27), are the pharmacologic targets of small-molecule Hsp90 ATP competitors. Here, we describe several of the Hsp90 inhibitors currently in clinical development.

Hsp90 inhibitors

Geldanamycin derivatives.

Geldanamycin is considered the prototype of the ansamycin class of Hsp90 inhibitors. Originally characterized as an antibiotic, this natural product showed intrinsic antitumor potential, inhibiting the activity of Src kinase by directly binding Hsp90 and interfering with the formation of the src–Hsp90 complex (28). Geldanamycin, however, was not clinically developed because of its poor solubility and substantial hepatotoxicity (29). These problems were overcome by its semisynthetic derivative 17-allyl-17-demethoxygeldanamycin (17-AAG), obtained by substituting a methoxy group for an allylamino group (30). This molecule was tested in more than 30 clinical studies, as a single agent or in combination with either chemotherapy or U.S. Food and Drug Administration (FDA)–approved drugs (31). Despite encouraging clinical activity, 17-AAG development was limited by its poor water solubility and, perhaps, by nonoptimal patient selection in the first trials. Progress in development was achieved by converting the quinone ring of 17-AAG to the corresponding hydroquinone (IPI-504) by Infinity Pharmaceuticals. This modification significantly improved water solubility, facilitating pharmaceutical preparations (32, 33). In vivo, this molecule has the capability of shifting between the quinone and the hydroquinone form via redox equilibrium, with the hydroquinone form exhibiting more potent inhibition of Hsp90 (33).

Purines.

In their search for synthetic molecules that compete with ATP for the Hsp90 ATP-binding pocket, Chiosis and colleagues identified PU3 as the first scaffold structure of this type having "geldanamycin-like" effects in breast cancer cells and from which more mature inhibitors were developed (34). PU-H71, an example from this research group, was found to be a potent Hsp90 inhibitor and was extremely efficacious in the preclinical models of triple-negative breast cancer. It is now being clinically evaluated in lymphomas (35). Conforma Therapeutics and subsequently Biogen Idec later discovered BIIB021, a derivate of PU3, which is orally available and has significantly improved potency and pharmacologic properties. This molecule showed activity in a number of preclinical models and is now in clinical development (36, 37). Examples of purine-like inhibitors are CUDC-305 (38) and NVP-BEP800 (39). These agents are reported to have high oral bioavailability, increased tumor retention, and, in the case of CUDC-305, exquisite distribution in mouse brain tissue.

Resorcinol derivatives.

Resorcinol is a natural antibiotic that competes with ATP for the binding to Hsp90. Resorcinol is a potent inhibitor of Hsp90 in vitro but, due to its intrinsic metabolic instability, lacks activity in vivo (40). With the use of alternate isolation techniques, other pharmacologically improved agents sharing the resorcinol core were identified by a number of research groups and/or pharmaceutical companies. Molecules such as NVP-AUY922, AT-13387, STA-9090, and KW-2478 have been described to be efficacious in several preclinical models and are all in different phases of clinical development (41).

Clinical–Translational Advances

As highlighted earlier, over the past decade our understanding of the function of Hsp90, together with the wealth of preclinical data validating Hsp90 as a pharmacologic target, has sparked interest in the development of Hsp90 inhibitors as a potential strategy for the treatment of cancer. A number of Hsp90 inhibitors are currently being tested in the clinic. In this section, we review the main cancer sites/subtypes where Hsp90 inhibitors are being investigated, the clinical data currently available, and the various strategies that are being pursued in optimizing their use in the treatment of cancer.

Clinically targetable client proteins of Hsp90

A number of malignancies in which Hsp90 inhibitors may have a potential role in treatment are currently being investigated. Estrogen and progesterone receptors are key drivers of breast cancer and are known to be client proteins of Hsp90 (12, 13). Furthermore, ERBB2, a receptor tyrosine kinase that is overexpressed in 20% of breast cancers, is also among the most sensitive client proteins of Hsp90, with shown preclinical activity of Hsp90 inhibitors in ERBB2-driven xenograft models (42, 43). In hormone-sensitive metastatic prostate cancer, androgen deprivation still is the mainstay of treatment and, given that the androgen receptor is a known client of Hsp90, inhibition of this chaperone may have a potential role in the management of this malignancy (44, 45). BRAF is mutated in 50% to 70% of malignant melanomas, and recent studies have shown that the use of BRAF tyrosine kinase inhibitors is associated with a survival advantage in this mutated subgroup (46). BRAF also depends on Hsp90 for folding and maturation, but, interestingly, Hsp90 inhibitors have been shown to selectively degrade mutant rather than wild-type BRAF (47). In principle, this property could also be exploited in colon cancer, where deregulation of the RAS/RAF/mitogen-activated pathway (48) and mutations of BRAF (49) are very well documented. However, the role of mutant BRAF in colon cancer seems to be less relevant than in melanoma. Other proteins that can potentially be targeted by Hsp90 inhibitors are mutant epidermal growth factor receptor, important in the treatment of non–small cell lung cancer (50), BCR-ABL, known to drive chronic myeloid leukemia (51), ZAP-70, associated with chronic lymphocytic leukemia (52), and CKIT, whose mutations drive gastrointestinal stromal tumors (53). Despite the fact that multiple myeloma does not depend on the classic client proteins described above, Hsp90 inhibitors are being actively investigated in this disease with the biologic rationale that Hsp90 is overexpressed in myeloma cells (54). Moreover, Hsp90 inhibition in this malignancy has shown activity in both in vitro and in vivo preclinical models through a proposed suppression of cytokine-dependent signaling pathways (55).

Single-agent activity of Hsp90 inhibitors

Although active as single agents, the natural Hsp90 inhibitors geldanamycin and resorcinol proved to be too toxic for clinical use. Tanespimycin (17-allylamino-17-demothoxygeldanamycin, 17-AAG), a derivative of geldanaymycin, has been shown to have similar biologic activity as its progenitor, but with an improved toxicity profile. Tenespimycin has shown minimal to moderate activity as a single agent in a number of tumor sites including melanoma, breast cancer, prostate cancer, and renal cell cancer (56, 57). In heavily pretreated patients with multiple myeloma, it has shown a progression-free survival of 3 months among patients with minimal response and a progression-free survival of 2.1 months among patients attaining stable disease (58). Limited success of tanespimycin as a single agent has been attributed, at least in part, to lack of selection of patients most likely to benefit from the drug. In patients with trastuzumab-resistant HER2-positive metastatic breast cancer, the addition of tanespimycin to trastuzumab has been shown to be beneficial. However, the activity of the HSP90 inhibitor as a single agent in a subgroup of women with HER2-positive breast cancer was not studied. Avespimycin (17-dimethylaminoethylaminoethylamino-17-demthoxygeldanamycin, 17-DMAG), an analogue of geldanamycin, has been shown to have single-agent activity in a variety of solid tumors, including castration-resistant prostate cancer, chondrosarcoma, and renal carcinoma (59).

Use of Hsp90 inhibitors in combination with other therapies

Hsp90 inhibitors have shown superior effects when combined with a variety of other therapeutic agents. Perhaps the best example of this success is a recently reported phase II study of 31 patients with ERBB2-overexpressing metastatic breast cancer who progressed on trastuzumab-based therapy. They then received a combination of weekly tanespimycin and trastuzumab (60). The authors of this study reported an overall response rate of 22%, a clinical benefit rate of 59%, a progression-free survival of 6 months, and an overall survival of 17 months. Other interesting results come from a subset of non–small cell lung cancer tumors driven by abnormal activity of the receptor tyrosine kinase, anaplastic lymphoma kinase (ALK), the target for the recently approved inhibitor crizotinib (61). In a recent report, Katayama and colleagues showed that non–small cell lung cancer cell lines that harbor the ALK gene translocation (responsible for ALK hyperactivity) and had become resistant to crizotinib retain sensitivity to the Hsp90 inhibitor tanespimycin (62). Moreover, single-agent retaspimycin hydrochloride (hydroquinone form of 17-AAG) has also shown activity in this subset of lung cancers (63).

BCR ABL, STAT3, and MEK, all client proteins of Hsp90, are (among others) important players in the progression of nonsolid tumors. Inhibition of the Hsp90 cycle has been shown to have synergistic activity in combination with proteasome inhibitors in the management of patients suffering from multiple myeloma (64, 65). In an open label phase I/II trial, Richardson and colleagues (66) reported an overall response rate of 27% with a complete response rate of 3% on a cohort of 63 patients with relapsed or relapsed and refractory multiple myeloma who received a combination of tanespimycin and the proteasome inhibitor bortezomib. Importantly, the authors further noted that this combination effectively inhibited both the proteasome (indicated by decreased 20S proteasome activity) and HSP90 (indicated by increased HSP70 expression).

Hsp90 inhibitors have shown either additive (67) or synergistic (68) activity also in combination with a variety of chemotherapeutic agents, including gemcitabine, carboplatin, docetaxel, and irinotecan (68–71). Significant activity has been also shown when Hsp90 inhibitors were combined with radiotherapy. Among the likely explanations for the observed synergy are enhanced antiangiogenic effects (72), the ability to compromise DNA damage response (73), and concomitant downregulation of ERB receptors (74).

Finally, preclinical evidence indicates that silencing the cochaperones Hsp70, Hsp27, or HSF-1 has been associated with increased sensitivity to Hsp90 inhibition. The activity of inhibitors of these cochaperones in combination with Hsp90 inhibitors is currently under investigation (75–79).

Designing the ideal clinical trial

Most of the clinical trials investigating the activity of Hsp90 inhibitors have been hampered by a variety of factors, including nonoptimal patient selection. It is possible that being driven by an oncogene client of Hsp90 is not a sufficient factor to predict clinical activity of Hsp90 inhibitors. Another possible determinant of activity of these agents will be the degree to which the client protein is dependent on Hsp90 for correct folding, maturation, and stability. Preclinical evidence indicates that ERBB2 is one of the most sensitive client proteins of the Hsp90 machinery. Clinical evidence indicates that breast tumors overexpressing ERBB2 that subsequently progress on trastuzumab still benefit by continued suppression of the ERBB2 pathway (80, 81). The activity shown by the combination of trastuzumab and tenespimycin (60), as described earlier, has been similar if not superior to the combinations of multiple anti-ERBB2 agents. This successful therapeutic strategy was not serendipitous. The rationale for inhibiting Hsp90 in ERBB2-positive cancer cells arose from extensive preclinical work showing how this receptor relies on Hsp90 binding for its maturation, stability, and function. The lesson we may learn from this is that we should first have a strong indication that our "favorite target" really depends on Hsp90 for its activity before we design a clinical trial with Hsp90 inhibitors.

When it comes to designing clinical trials for combinations of Hsp90 inhibitors with other therapeutic agents, we may not have to be so strict about Hsp90 dependence of the client proteins. While therapeutic inhibition of an oncoprotein leads to silencing of the oncogene-dependent signaling, it has been recently observed that it also releases negative feedback, resulting in the activation of upstream receptors and parallel compensatory pathways that limits the efficacy of a given therapy (82). Blockade of these compensatory pathways may result in a "synthetic lethality–like" effect that would limit the emergence of both primary and acquired resistance to a variety of agents used in the management of cancer (82). It is likely that many client proteins of Hsp90 are among the key players/effectors of these compensatory mechanisms and, as a result, the addition of Hsp90 inhibitors would be beneficial. With a systematic identification and characterization of the mechanisms that cancer cells devise to adapt to pharmacologic pressures, we will know whether the use of Hsp90 inhibitors is a valid combinatorial strategy in this setting.

Conclusions

It is becoming evident that being a client protein of Hsp90 does not necessarily mean being a "good" client. Only a fraction of the possible proteins interacting with Hsp90 are expected to serve as clinically exploitable targets. Careful and extensive preclinical work is needed to identify those client proteins that are indispensable for the growth and survival of a given tumor and, at the same time, those that are strictly dependent on Hsp90 for their maturation and function. Future combinatorial studies will confirm whether inhibition of Hsp90 is a good strategy to potentiate target inhibition and/or prevent the insurgence of drug resistance.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interests were disclosed.

Authors' Contributions

Conception and design: M. Scaltriti, S. Dawood, J. Cortes

Development of methodology: J. Cortes

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): J. Cortes

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): J. Cortes

Writing, review, and/or revision of the manuscript: M. Scaltriti, J. Cortes

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): J. Cortes

Study supervision: M. Scaltriti, J. Cortes

Acknowledgments

Given the space limitations of the review, the authors sincerely apologize for their inability to cite everyone who has contributed to this field of inquiry. The authors thank Dr. Andrea Talis for editorial assistance.

  • Received May 8, 2012.
  • Revision received May 30, 2012.
  • Accepted June 1, 2012.
  • ©2012 American Association for Cancer Research.

References

  1. 1.↵
    1. Isaacs JS,
    2. Xu W,
    3. Neckers L
    . Heat shock protein 90 as a molecular target for cancer therapeutics. Cancer Cell 2003;3:213–7.
    OpenUrlCrossRefPubMed
  2. 2.↵
    1. Taipale M,
    2. Jarosz DF,
    3. Lindquist S
    . HSP90 at the hub of protein homeostasis: emerging mechanistic insights. Nat Rev Mol Cell Biol 2010;11:515–28.
    OpenUrlCrossRefPubMed
  3. 3.↵
    1. Banerji U
    . Heat shock protein 90 as a drug target: some like it hot. Clin Cancer Res 2009;15:9–14.
    OpenUrlAbstract/FREE Full Text
  4. 4.↵
    1. Neckers L
    . Heat shock protein 90: the cancer chaperone. J Biosci 2007;32:517–30.
    OpenUrlCrossRefPubMed
  5. 5.↵
    1. Conroy SE,
    2. Latchman DS
    . Do heat shock proteins have a role in breast cancer? Br J Cancer 1996;74:717–21.
    OpenUrlPubMed
  6. 6.↵
    1. Jameel A,
    2. Skilton RA,
    3. Campbell TA,
    4. Chander SK,
    5. Coombes RC,
    6. Luqmani YA
    . Clinical and biological significance of HSP89 alpha in human breast cancer. Int J Cancer 1992;50:409–15.
    OpenUrlPubMed
  7. 7.↵
    1. Li CF,
    2. Huang WW,
    3. Wu JM,
    4. Yu SC,
    5. Hu TH,
    6. Uen YH,
    7. et al.
    Heat shock protein 90 overexpression independently predicts inferior disease-free survival with differential expression of the alpha and beta isoforms in gastrointestinal stromal tumors. Clin Cancer Res 2008;14:7822–31.
    OpenUrlAbstract/FREE Full Text
  8. 8.↵
    1. Pick E,
    2. Kluger Y,
    3. Giltnane JM,
    4. Moeder C,
    5. Camp RL,
    6. Rimm DL,
    7. et al.
    High HSP90 expression is associated with decreased survival in breast cancer. Cancer Res 2007;67:2932–7.
    OpenUrlAbstract/FREE Full Text
  9. 9.↵
    1. Kamal A,
    2. Thao L,
    3. Sensintaffar J,
    4. Zhang L,
    5. Boehm MF,
    6. Fritz LC,
    7. et al.
    A high-affinity conformation of Hsp90 confers tumour selectivity on Hsp90 inhibitors. Nature 2003;425:407–10.
    OpenUrlCrossRefPubMed
  10. 10.↵
    1. Solit DB,
    2. Rosen N
    . Hsp90: a novel target for cancer therapy. Curr Top Med Chem 2006;6:1205–14.
    OpenUrlCrossRefPubMed
  11. 11.↵
    1. Connell P,
    2. Ballinger CA,
    3. Jiang J,
    4. Wu Y,
    5. Thompson LJ,
    6. Hohfeld J,
    7. et al.
    The co-chaperone CHIP regulates protein triage decisions mediated by heat-shock proteins. Nat Cell Biol 2001;3:93–6.
    OpenUrlCrossRefPubMed
  12. 12.↵
    1. Hernandez MP,
    2. Chadli A,
    3. Toft DO
    . HSP40 binding is the first step in the HSP90 chaperoning pathway for the progesterone receptor. J Biol Chem 2002;277:11873–81.
    OpenUrlAbstract/FREE Full Text
  13. 13.↵
    1. Pratt WB,
    2. Galigniana MD,
    3. Morishima Y,
    4. Murphy PJ
    . Role of molecular chaperones in steroid receptor action. Essays Biochem 2004;40:41–58.
    OpenUrlPubMed
  14. 14.↵
    1. Prince T,
    2. Matts RL
    . Exposure of protein kinase motifs that trigger binding of Hsp90 and Cdc37. Biochem Biophys Res Commun 2005;338:1447–54.
    OpenUrlCrossRefPubMed
  15. 15.↵
    1. Terasawa K,
    2. Yoshimatsu K,
    3. Iemura S,
    4. Natsume T,
    5. Tanaka K,
    6. Minami Y
    . Cdc37 interacts with the glycine-rich loop of Hsp90 client kinases. Mol Cell Biol 2006;26:3378–89.
    OpenUrlAbstract/FREE Full Text
  16. 16.↵
    1. Xu Y,
    2. Singer MA,
    3. Lindquist S
    . Maturation of the tyrosine kinase c-src as a kinase and as a substrate depends on the molecular chaperone Hsp90. Proc Natl Acad Sci U S A 1999;96:109–14.
    OpenUrlAbstract/FREE Full Text
  17. 17.↵
    1. Cintron NS,
    2. Toft D
    . Defining the requirements for Hsp40 and Hsp70 in the Hsp90 chaperone pathway. J Biol Chem 2006;281:26235–44.
    OpenUrlAbstract/FREE Full Text
  18. 18.↵
    1. Hernandez MP,
    2. Sullivan WP,
    3. Toft DO
    . The assembly and intermolecular properties of the hsp70-Hop-hsp90 molecular chaperone complex. J Biol Chem 2002;277:38294–304.
    OpenUrlAbstract/FREE Full Text
  19. 19.↵
    1. Smith DF,
    2. Sullivan WP,
    3. Marion TN,
    4. Zaitsu K,
    5. Madden B,
    6. McCormick DJ,
    7. et al.
    Identification of a 60-kilodalton stress-related protein, p60, which interacts with hsp90 and hsp70. Mol Cell Biol 1993;13:869–76.
    OpenUrlAbstract/FREE Full Text
  20. 20.↵
    1. Wayne N,
    2. Bolon DN
    . Dimerization of Hsp90 is required for in vivo function. Design and analysis of monomers and dimers. J Biol Chem 2007;282:35386–95.
    OpenUrlAbstract/FREE Full Text
  21. 21.↵
    1. Wegele H,
    2. Wandinger SK,
    3. Schmid AB,
    4. Reinstein J,
    5. Buchner J
    . Substrate transfer from the chaperone Hsp70 to Hsp90. J Mol Biol 2006;356:802–11.
    OpenUrlCrossRefPubMed
  22. 22.↵
    1. Csermely P,
    2. Kajtar J,
    3. Hollosi M,
    4. Jalsovszky G,
    5. Holly S,
    6. Kahn CR,
    7. et al.
    ATP induces a conformational change of the 90-kDa heat shock protein (hsp90). J Biol Chem 1993;268:1901–7.
    OpenUrlAbstract/FREE Full Text
  23. 23.↵
    1. McLaughlin SH,
    2. Sobott F,
    3. Yao ZP,
    4. Zhang W,
    5. Nielsen PR,
    6. Grossmann JG,
    7. et al.
    The co-chaperone p23 arrests the Hsp90 ATPase cycle to trap client proteins. J Mol Biol 2006;356:746–58.
    OpenUrlCrossRefPubMed
  24. 24.↵
    1. Sullivan W,
    2. Stensgard B,
    3. Caucutt G,
    4. Bartha B,
    5. McMahon N,
    6. Alnemri ES,
    7. et al.
    Nucleotides and two functional states of hsp90. J Biol Chem 1997;272:8007–12.
    OpenUrlAbstract/FREE Full Text
  25. 25.↵
    1. Sullivan WP,
    2. Owen BA,
    3. Toft DO
    . The influence of ATP and p23 on the conformation of hsp90. J Biol Chem 2002;277:45942–8.
    OpenUrlAbstract/FREE Full Text
  26. 26.↵
    1. Obermann WM,
    2. Sondermann H,
    3. Russo AA,
    4. Pavletich NP,
    5. Hartl FU
    . In vivo function of Hsp90 is dependent on ATP binding and ATP hydrolysis. J Cell Biol 1998;143:901–10.
    OpenUrlAbstract/FREE Full Text
  27. 27.↵
    1. Panaretou B,
    2. Prodromou C,
    3. Roe SM,
    4. O'Brien R,
    5. Ladbury JE,
    6. Piper PW,
    7. et al.
    ATP binding and hydrolysis are essential to the function of the Hsp90 molecular chaperone in vivo . EMBO J 1998;17:4829–36.
    OpenUrlAbstract
  28. 28.↵
    1. Whitesell L,
    2. Mimnaugh EG,
    3. De Costa B,
    4. Myers CE,
    5. Neckers LM
    . Inhibition of heat shock protein HSP90-pp60v-src heteroprotein complex formation by benzoquinone ansamycins: essential role for stress proteins in oncogenic transformation. Proc Natl Acad Sci U S A 1994;91:8324–8.
    OpenUrlAbstract/FREE Full Text
  29. 29.↵
    1. Supko JG,
    2. Hickman RL,
    3. Grever MR,
    4. Malspeis L
    . Preclinical pharmacologic evaluation of geldanamycin as an antitumor agent. Cancer Chemother Pharmacol 1995;36:305–15.
    OpenUrlCrossRefPubMed
  30. 30.↵
    1. Schulte TW,
    2. Neckers LM
    . The benzoquinone ansamycin 17-allylamino-17-demethoxygeldanamycin binds to HSP90 and shares important biologic activities with geldanamycin. Cancer Chemother Pharmacol 1998;42:273–9.
    OpenUrlCrossRefPubMed
  31. 31.↵
    1. Kim YS,
    2. Alarcon SV,
    3. Lee S,
    4. Lee MJ,
    5. Giaccone G,
    6. Neckers L,
    7. et al.
    Update on Hsp90 inhibitors in clinical trial. Curr Top Med Chem 2009;9:1479–92.
    OpenUrlCrossRefPubMed
  32. 32.↵
    1. Ge J,
    2. Normant E,
    3. Porter JR,
    4. Ali JA,
    5. Dembski MS,
    6. Gao Y,
    7. et al.
    Design, synthesis, and biological evaluation of hydroquinone derivatives of 17-amino-17-demethoxygeldanamycin as potent, water-soluble inhibitors of Hsp90. J Med Chem 2006;49:4606–15.
    OpenUrlCrossRefPubMed
  33. 33.↵
    1. Sydor JR,
    2. Normant E,
    3. Pien CS,
    4. Porter JR,
    5. Ge J,
    6. Grenier L,
    7. et al.
    Development of 17-allylamino-17-demethoxygeldanamycin hydroquinone hydrochloride (IPI-504), an anti-cancer agent directed against Hsp90. Proc Natl Acad Sci U S A 2006;103:17408–13.
    OpenUrlAbstract/FREE Full Text
  34. 34.↵
    1. Chiosis G,
    2. Timaul MN,
    3. Lucas B,
    4. Munster PN,
    5. Zheng FF,
    6. Sepp-Lorenzino L,
    7. et al.
    A small molecule designed to bind to the adenine nucleotide pocket of Hsp90 causes Her2 degradation and the growth arrest and differentiation of breast cancer cells. Chem Biol 2001;8:289–99.
    OpenUrlCrossRefPubMed
  35. 35.↵
    1. Caldas-Lopes E,
    2. Cerchietti L,
    3. Ahn JH,
    4. Clement CC,
    5. Robles AI,
    6. Rodina A,
    7. et al.
    Hsp90 inhibitor PU-H71, a multimodal inhibitor of malignancy, induces complete responses in triple-negative breast cancer models. Proc Natl Acad Sci U S A 2009;106:8368–73.
    OpenUrlAbstract/FREE Full Text
  36. 36.↵
    1. Kasibhatla SR,
    2. Hong K,
    3. Biamonte MA,
    4. Busch DJ,
    5. Karjian PL,
    6. Sensintaffar JL,
    7. et al.
    Rationally designed high-affinity 2-amino-6-halopurine heat shock protein 90 inhibitors that exhibit potent antitumor activity. J Med Chem 2007;50:2767–78.
    OpenUrlCrossRefPubMed
  37. 37.↵
    1. Lundgren K,
    2. Zhang H,
    3. Brekken J,
    4. Huser N,
    5. Powell RE,
    6. Timple N,
    7. et al.
    BIIB021, an orally available, fully synthetic small-molecule inhibitor of the heat shock protein Hsp90. Mol Cancer Ther 2009;8:921–9.
    OpenUrlAbstract/FREE Full Text
  38. 38.↵
    1. Bao R,
    2. Lai CJ,
    3. Qu H,
    4. Wang D,
    5. Yin L,
    6. Zifcak B,
    7. et al.
    CUDC-305, a novel synthetic HSP90 inhibitor with unique pharmacologic properties for cancer therapy. Clin Cancer Res 2009;15:4046–57.
    OpenUrlAbstract/FREE Full Text
  39. 39.↵
    1. Brough PA,
    2. Barril X,
    3. Borgognoni J,
    4. Chene P,
    5. Davies NG,
    6. Davis B,
    7. et al.
    Combining hit identification strategies: fragment-based and in silico approaches to orally active 2-aminothieno[2,3-d]pyrimidine inhibitors of the Hsp90 molecular chaperone. J Med Chem 2009;52:4794–809.
    OpenUrlCrossRefPubMed
  40. 40.↵
    1. Yang ZQ,
    2. Geng X,
    3. Solit D,
    4. Pratilas CA,
    5. Rosen N,
    6. Danishefsky SJ
    . New efficient synthesis of resorcinylic macrolides via ynolides: establishment of cycloproparadicicol as synthetically feasible preclinical anticancer agent based on Hsp90 as the target. J Am Chem Soc 2004;126:7881–9.
    OpenUrlCrossRefPubMed
  41. 41.↵
    1. Jhaveri K,
    2. Taldone T,
    3. Modi S,
    4. Chiosis G
    . Advances in the clinical development of heat shock protein 90 (Hsp90) inhibitors in cancers. Biochimica Biophys Acta 2012;1823:742–55.
    OpenUrlCrossRefPubMed
  42. 42.↵
    1. Chandarlapaty S,
    2. Scaltriti M,
    3. Angelini P,
    4. Ye Q,
    5. Guzman M,
    6. Hudis CA,
    7. et al.
    Inhibitors of HSP90 block p95-HER2 signaling in Trastuzumab-resistant tumors and suppress their growth. Oncogene 2010;29:325–34.
    OpenUrlCrossRefPubMed
  43. 43.↵
    1. Scaltriti M,
    2. Serra V,
    3. Normant E,
    4. Guzman M,
    5. Rodriguez O,
    6. Lim AR,
    7. et al.
    Antitumor activity of the Hsp90 inhibitor IPI-504 in HER2-positive trastuzumab-resistant breast cancer. Mol Cancer Ther 2011;10:817–24.
    OpenUrlAbstract/FREE Full Text
  44. 44.↵
    1. Chen Y,
    2. Sawyers CL,
    3. Scher HI
    . Targeting the androgen receptor pathway in prostate cancer. Curr Opin Pharmacol 2008;8:440–8.
    OpenUrlCrossRefPubMed
  45. 45.↵
    1. Vanaja DK,
    2. Mitchell SH,
    3. Toft DO,
    4. Young CY
    . Effect of geldanamycin on androgen receptor function and stability. Cell Stress Chaperones 2002;7:55–64.
    OpenUrlCrossRefPubMed
  46. 46.↵
    1. Chapman PB,
    2. Hauschild A,
    3. Robert C,
    4. Haanen JB,
    5. Ascierto P,
    6. Larkin J,
    7. et al.
    Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med 2011;364:2507–16.
    OpenUrlCrossRefPubMed
  47. 47.↵
    1. Dias S,
    2. Friedlos F,
    3. Light Y,
    4. Springer C,
    5. Workman P,
    6. Marais R
    da Rocha Dias S, Friedlos F, Light Y, Springer C, Workman P, Marais R . Activated B-RAF is an Hsp90 client protein that is targeted by the anticancer drug 17-allylamino-17-demethoxygeldanamycin. Cancer Res 2005;65:10686–91.
    OpenUrlAbstract/FREE Full Text
  48. 48.↵
    1. Haigis KM,
    2. Kendall KR,
    3. Wang Y,
    4. Cheung A,
    5. Haigis MC,
    6. Glickman JN,
    7. et al.
    Differential effects of oncogenic K-Ras and N-Ras on proliferation, differentiation and tumor progression in the colon. Nat Genet 2008;40:600–8.
    OpenUrlCrossRefPubMed
  49. 49.↵
    1. Davies H,
    2. Bignell GR,
    3. Cox C,
    4. Stephens P,
    5. Edkins S,
    6. Clegg S,
    7. et al.
    Mutations of the BRAF gene in human cancer. Nature 2002;417:949–54.
    OpenUrlCrossRefPubMed
  50. 50.↵
    1. Shimamura T,
    2. Lowell AM,
    3. Engelman JA,
    4. Shapiro GI
    . Epidermal growth factor receptors harboring kinase domain mutations associate with the heat shock protein 90 chaperone and are destabilized following exposure to geldanamycins. Cancer Res 2005;65:6401–8.
    OpenUrlAbstract/FREE Full Text
  51. 51.↵
    1. Shiotsu Y,
    2. Soga S,
    3. Akinaga S
    . Heat shock protein 90-antagonist destabilizes Bcr-Abl/HSP90 chaperone complex. Leuk Lymphoma 2002;43:961–8.
    OpenUrlPubMed
  52. 52.↵
    1. Castro JE,
    2. Prada CE,
    3. Loria O,
    4. Kamal A,
    5. Chen L,
    6. Burrows FJ,
    7. et al.
    ZAP-70 is a novel conditional heat shock protein 90 (Hsp90) client: inhibition of Hsp90 leads to ZAP-70 degradation, apoptosis, and impaired signaling in chronic lymphocytic leukemia. Blood 2005;106:2506–12.
    OpenUrlAbstract/FREE Full Text
  53. 53.↵
    1. Bauer S,
    2. Yu LK,
    3. Demetri GD,
    4. Fletcher JA
    . Heat shock protein 90 inhibition in imatinib-resistant gastrointestinal stromal tumor. Cancer Res 2006;66:9153–61.
    OpenUrlAbstract/FREE Full Text
  54. 54.↵
    1. Chatterjee M,
    2. Jain S,
    3. Stuhmer T,
    4. Andrulis M,
    5. Ungethum U,
    6. Kuban RJ,
    7. et al.
    STAT3 and MAPK signaling maintain overexpression of heat shock proteins 90alpha and beta in multiple myeloma cells, which critically contribute to tumor-cell survival. Blood 2007;109:720–8.
    OpenUrlAbstract/FREE Full Text
  55. 55.↵
    1. Mitsiades CS,
    2. Mitsiades NS,
    3. McMullan CJ,
    4. Poulaki V,
    5. Kung AL,
    6. Davies FE,
    7. et al.
    Antimyeloma activity of heat shock protein-90 inhibition. Blood 2006;107:1092–100.
    OpenUrlAbstract/FREE Full Text
  56. 56.↵
    1. Banerji U,
    2. O'Donnell A,
    3. Scurr M,
    4. Pacey S,
    5. Stapleton S,
    6. Asad Y,
    7. et al.
    Phase I pharmacokinetic and pharmacodynamic study of 17-allylamino, 17-demethoxygeldanamycin in patients with advanced malignancies. J Clin Oncol 2005;23:4152–61.
    OpenUrlAbstract/FREE Full Text
  57. 57.↵
    1. Solit DB,
    2. Ivy SP,
    3. Kopil C,
    4. Sikorski R,
    5. Morris MJ,
    6. Slovin SF,
    7. et al.
    Phase I trial of 17-allylamino-17-demethoxygeldanamycin in patients with advanced cancer. Clin Cancer Res 2007;13:1775–82.
    OpenUrlAbstract/FREE Full Text
  58. 58.↵
    1. Richardson PG,
    2. Chanan-Khan AA,
    3. Alsina M,
    4. Albitar M,
    5. Berman D,
    6. Messina M,
    7. et al.
    Tanespimycin monotherapy in relapsed multiple myeloma: results of a phase 1 dose-escalation study. Br J Haematol 2010;150:438–45.
    OpenUrlPubMed
  59. 59.↵
    1. Pacey S,
    2. Wilson RH,
    3. Walton M,
    4. Eatock MM,
    5. Hardcastle A,
    6. Zetterlund A,
    7. et al.
    A phase I study of the heat shock protein 90 inhibitor alvespimycin (17-DMAG) given intravenously to patients with advanced solid tumors. Clin Cancer Res 2011;17:1561–70.
    OpenUrlAbstract/FREE Full Text
  60. 60.↵
    1. Modi S,
    2. Stopeck A,
    3. Linden H,
    4. Solit D,
    5. Chandarlapaty S,
    6. Rosen N,
    7. et al.
    HSP90 inhibition is effective in breast cancer: a phase II trial of tanespimycin (17-AAG) plus trastuzumab in patients with HER2-positive metastatic breast cancer progressing on trastuzumab. Clin Cancer Res 2011;17:5132–9.
    OpenUrlAbstract/FREE Full Text
  61. 61.↵
    1. Kwak EL,
    2. Bang YJ,
    3. Camidge DR,
    4. Shaw AT,
    5. Solomon B,
    6. Maki RG,
    7. et al.
    Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer. N Engl J Med 2010;363:1693–703.
    OpenUrlCrossRefPubMed
  62. 62.↵
    1. Katayama R,
    2. Khan TM,
    3. Benes C,
    4. Lifshits E,
    5. Ebi H,
    6. Rivera VM,
    7. et al.
    Therapeutic strategies to overcome crizotinib resistance in non-small cell lung cancers harboring the fusion oncogene EML4-ALK. Proc Natl Acad Sci U S A 2011;108:7535–40.
    OpenUrlAbstract/FREE Full Text
  63. 63.↵
    1. Sequist LV,
    2. Gettinger S,
    3. Senzer NN,
    4. Martins RG,
    5. Janne PA,
    6. Lilenbaum R,
    7. et al.
    Activity of IPI-504, a novel heat-shock protein 90 inhibitor, in patients with molecularly defined non-small-cell lung cancer. J Clin Oncol 2010;28:4953–60.
    OpenUrlAbstract/FREE Full Text
  64. 64.↵
    1. Khong T,
    2. Spencer A
    . Targeting HSP 90 induces apoptosis and inhibits critical survival and proliferation pathways in multiple myeloma. Mol Cancer Ther 2011;10:1909–17.
    OpenUrlAbstract/FREE Full Text
  65. 65.↵
    1. Richardson PG,
    2. Mitsiades CS,
    3. Laubach JP,
    4. Lonial S,
    5. Chanan-Khan AA,
    6. Anderson KC
    . Inhibition of heat shock protein 90 (HSP90) as a therapeutic strategy for the treatment of myeloma and other cancers. Br J Haematol 2011;152:367–79.
    OpenUrlCrossRefPubMed
  66. 66.↵
    1. Richardson PG,
    2. Chanan-Khan AA,
    3. Lonial S,
    4. Krishnan AY,
    5. Carroll MP,
    6. Alsina M,
    7. et al.
    Tanespimycin and bortezomib combination treatment in patients with relapsed or relapsed and refractory multiple myeloma: results of a phase 1/2 study. Br J Haematol 2011;153:729–40.
    OpenUrlCrossRefPubMed
  67. 67.↵
    1. Banerji U,
    2. Sain N,
    3. Sharp SY,
    4. Valenti M,
    5. Asad Y,
    6. Ruddle R,
    7. et al.
    An in vitro and in vivo study of the combination of the heat shock protein inhibitor 17-allylamino-17-demethoxygeldanamycin and carboplatin in human ovarian cancer models. Cancer Chemother Pharmacol 2008;62:769–78.
    OpenUrlCrossRefPubMed
  68. 68.↵
    1. Munster PN,
    2. Basso A,
    3. Solit D,
    4. Norton L,
    5. Rosen N
    . Modulation of Hsp90 function by ansamycins sensitizes breast cancer cells to chemotherapy-induced apoptosis in an RB- and schedule-dependent manner. See: E. A. Sausville, Combining cytotoxics and 17-allylamino, 17-demethoxygeldanamycin: sequence and tumor biology matters, Clin. Cancer Res., 7: 2155–2158, 2001. Clin Cancer Res 2001;7:2228–36.
    OpenUrlAbstract/FREE Full Text
  69. 69.↵
    1. Hubbard J,
    2. Erlichman C,
    3. Toft DO,
    4. Qin R,
    5. Stensgard BA,
    6. Felten S,
    7. et al.
    Phase I study of 17-allylamino-17 demethoxygeldanamycin, gemcitabine and/or cisplatin in patients with refractory solid tumors. Invest New Drugs 2011;29:473–80.
    OpenUrlCrossRefPubMed
  70. 70.↵
    1. Iyer G,
    2. Morris MJ,
    3. Rathkopf D,
    4. Slovin SF,
    5. Steers M,
    6. Larson SM,
    7. et al.
    A phase I trial of docetaxel and pulse-dose 17-allylamino-17-demethoxygeldanamycin in adult patients with solid tumors. Cancer Chemother Pharmacol 2012;69:1089–97.
    OpenUrlCrossRefPubMed
  71. 71.↵
    1. Tse AN,
    2. Klimstra DS,
    3. Gonen M,
    4. Shah M,
    5. Sheikh T,
    6. Sikorski R,
    7. et al.
    A phase 1 dose-escalation study of irinotecan in combination with 17-allylamino-17-demethoxygeldanamycin in patients with solid tumors. Clin Cancer Res 2008;14:6704–11.
    OpenUrlAbstract/FREE Full Text
  72. 72.↵
    1. Kabakov AE,
    2. Makarova YM,
    3. Malyutina YV
    . Radiosensitization of human vascular endothelial cells through Hsp90 inhibition with 17-N-allilamino-17-demethoxygeldanamycin. Int J Radiat Oncol Biol Phys 2008;71:858–65.
    OpenUrlPubMed
  73. 73.↵
    1. Dote H,
    2. Burgan WE,
    3. Camphausen K,
    4. Tofilon PJ
    . Inhibition of hsp90 compromises the DNA damage response to radiation. Cancer Res 2006;66:9211–20.
    OpenUrlAbstract/FREE Full Text
  74. 74.↵
    1. Dote H,
    2. Cerna D,
    3. Burgan WE,
    4. Camphausen K,
    5. Tofilon PJ
    . ErbB3 expression predicts tumor cell radiosensitization induced by Hsp90 inhibition. Cancer Res 2005;65:6967–75.
    OpenUrlAbstract/FREE Full Text
  75. 75.↵
    1. Davenport EL,
    2. Zeisig A,
    3. Aronson LI,
    4. Moore HE,
    5. Hockley S,
    6. Gonzalez D,
    7. et al.
    Targeting heat shock protein 72 enhances Hsp90 inhibitor-induced apoptosis in myeloma. Leukemia 2010;24:1804–7.
    OpenUrlCrossRefPubMed
  76. 76.↵
    1. Evans CG,
    2. Chang L,
    3. Gestwicki JE
    . Heat shock protein 70 (hsp70) as an emerging drug target. J Med Chem 2010;53:4585–602.
    OpenUrlCrossRefPubMed
  77. 77.↵
    1. Hadchity E,
    2. Aloy MT,
    3. Paulin C,
    4. Armandy E,
    5. Watkin E,
    6. Rousson R,
    7. et al.
    Heat shock protein 27 as a new therapeutic target for radiation sensitization of head and neck squamous cell carcinoma. Mol Ther 2009;17:1387–94.
    OpenUrlCrossRefPubMed
  78. 78.↵
    1. Powers MV,
    2. Jones K,
    3. Barillari C,
    4. Westwood I,
    5. van Montfort RL,
    6. Workman P
    . Targeting HSP70: the second potentially druggable heat shock protein and molecular chaperone? Cell Cycle 2010;9:1542–50.
    OpenUrlCrossRefPubMed
  79. 79.↵
    1. Powers MV,
    2. Workman P
    . Inhibitors of the heat shock response: biology and pharmacology. FEBS Lett 2007;581:3758–69.
    OpenUrlCrossRefPubMed
  80. 80.↵
    1. Abramson V,
    2. Arteaga CL
    . New strategies in HER2-overexpressing breast cancer: many combinations of targeted drugs available. Clin Cancer Res 2011;17:952–8.
    OpenUrlAbstract/FREE Full Text
  81. 81.↵
    1. Arteaga CL
    . Why is this effective HSP90 inhibitor not being developed in HER2+ breast cancer? Clin Cancer Res 2011;17:4919–21.
    OpenUrlAbstract/FREE Full Text
  82. 82.↵
    1. Chandarlapaty S
    . Negative feedback and adaptive resistance to the targeted therapy of cancer. Cancer Discov 2012;2:311–9.
    OpenUrlAbstract/FREE Full Text
View Abstract
PreviousNext
Back to top
Clinical Cancer Research: 18 (17)
September 2012
Volume 18, Issue 17
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover

Sign up for alerts

View this article with LENS

Open full page PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Clinical Cancer Research article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Molecular Pathways: Targeting Hsp90—Who Benefits and Who Does Not
(Your Name) has forwarded a page to you from Clinical Cancer Research
(Your Name) thought you would be interested in this article in Clinical Cancer Research.
Citation Tools
Molecular Pathways: Targeting Hsp90—Who Benefits and Who Does Not
Maurizio Scaltriti, Shaheenah Dawood and Javier Cortes
Clin Cancer Res September 1 2012 (18) (17) 4508-4513; DOI: 10.1158/1078-0432.CCR-11-2138

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Molecular Pathways: Targeting Hsp90—Who Benefits and Who Does Not
Maurizio Scaltriti, Shaheenah Dawood and Javier Cortes
Clin Cancer Res September 1 2012 (18) (17) 4508-4513; DOI: 10.1158/1078-0432.CCR-11-2138
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Background
    • Clinical–Translational Advances
    • Conclusions
    • Disclosure of Potential Conflicts of Interest
    • Authors' Contributions
    • Acknowledgments
    • References
  • Figures & Data
  • Info & Metrics
  • PDF
Advertisement

Related Articles

Cited By...

More in this TOC Section

  • Therapeutic Targeting of the Liver Microenvironment
  • Targeting the Protein Kinase Wee1 in Cancer
  • Metabolic Control of Histone Methylation and Gene Expression
Show more Molecular Pathways
  • Home
  • Alerts
  • Feedback
  • Privacy Policy
Facebook  Twitter  LinkedIn  YouTube  RSS

Articles

  • Online First
  • Current Issue
  • Past Issues
  • CCR Focus Archive
  • Meeting Abstracts

Info for

  • Authors
  • Subscribers
  • Advertisers
  • Librarians
  • Reviewers

About Clinical Cancer Research

  • About the Journal
  • Editorial Board
  • Permissions
  • Submit a Manuscript
AACR logo

Copyright © 2019 by the American Association for Cancer Research.

Clinical Cancer Research
eISSN: 1557-3265
ISSN: 1078-0432

Advertisement