Review
Post Screen
Development and application of Hsp90 inhibitors

https://doi.org/10.1016/j.drudis.2007.10.007Get rights and content

Heat shock protein 90 has emerged as an important target in several diseases. The present review will discuss our understanding of the role played by Hsp90 in regulating and maintaining the transformed phenotype in cancers and neurodegenerative diseases, as well as recent findings on its roles in fungal and viral infections. It will also update the reader on the preclinical development and clinical translation of Hsp90 inhibitors.

Introduction

As we have currently come to understand, heat shock protein 90 (Hsp90) is a key component of a multichaperone complex with important roles in the development and progression of pathogenic cellular transformation 1, 2, 3, 4. Its wide-ranging functions result from the ability of Hsp90 to chaperone several ‘client proteins’ that play a central pathogenic role in human diseases including cancer, neurodegenerative diseases, and viral infections. At the time of its discovery, however, it was difficult to imagine Hsp90 as a potential therapeutic target. The chaperone is highly abundant in most tissues, comprising over 1% of the total cellular content even in the absence of stress. Further, genetic knockout of Hsp90 is lethal in eukaryotes and over 100 kinases and transcription factors, many of which are crucial to normal cellular growth and survival, have been shown to interact functionally with Hsp90. The initial interpretation of these findings was that a therapeutic window would be difficult, if not impossible, to achieve with Hsp90 inhibitors. This unfavorable view regarding the potential of Hsp90 as a therapeutic target has changed only after the discovery of geldanamycin (GM) (Figure 1). GM was found as part of a phenotypic screen focused on the identification of agents capable of inducing the morphologic reversion of v-src-transformed 3T3 cells [5]. It was initially hypothesized that GM was a direct inhibitor of v-SRC activity. Whitesell and colleagues later clarified that GM's biologic activity was actually attributable to its ability to bind to and inhibit Hsp90 [6]. Co-crystallization studies demonstrated that GM binds to an ATP/ADP pocket in the N-terminal domain of Hsp90 and that it inhibits Hsp90 function by preventing full chaperone cycling. In cells exposed to GM, unprocessed chaperone-client protein complexes accumulate within the cell leading to recruitment of E3 ubiquitin ligases that target Hsp90 clients for degradation in the proteasome 7, 8.

Much of our understanding of the role of Hsp90 in promoting malignant transformation has been derived from studies using GM and other structurally diverse Hsp90 inhibitors as biologic probes [9]. A surprising observation derived with these agents was that cancer cells were significantly more sensitive to Hsp90 inhibition than non-transformed cells 1, 2, 3, 4, 5, 6, 9. This finding raised the possibility that Hsp90 inhibitors may possess an exploitable therapeutic index and prompted the testing of this strategy using xenograft and transgenic cancer models. These studies, which have used primarily 17-allylamino-17-demethoxy-geldanamycin (17AAG) (Figure 1), a GM derivative with a more favorable profile, but also synthetic small molecule Hsp90 inhibitors, clearly demonstrate that Hsp90 inhibitors possess potent anti-cancer activity at non-toxic doses 10, 11, 12.

Several models have been proposed to explain the selective sensitivity of cancer cells to Hsp90 inhibition 13, 14, 15, 16. One model, akin to the ‘oncogene addiction’ model proposed by Weinstein [13], focuses on the exclusive dependence of some transformed cells on a sensitive Hsp90 client protein. In this model, degradation of a specific Hsp90 client in the appropriate genetic context, for example, BRAF in a melanoma cell with V600E mutant BRAF or Bcr-Abl in chronic myeloid leukemia (CML) 1, 2, 3, 4, results in apoptosis and/or differentiation, whereas its degradation in normal cells leads to little to no effect. As Hsp90 interacts with a large number of oncogenic kinases and transcription factors, this model has been used to justify the clinical development of Hsp90 inhibitors in a broad range of tumor types (Table 1). The importance of this model to the clinical development of Hsp90 inhibitors is that it highlights the relevance of patient selection in the design of Hsp90 inhibitor trials. Unfortunately, only limited animal studies have been performed with these inhibitors to date and, therefore, the ability of 17AAG or other Hsp90 inhibitors to degrade most putative Hsp90 clients in vivo has yet to be explored in relevant animal model systems.

A complementary ‘Hsp90 addiction model’ is based upon the hypothesis that Hsp90 is limiting in many tumor cells because of their increased load of mutated and unfolded proteins 14, 15. An increased requirement for Hsp90 chaperone function in tumors cells may be due to the overexpression of mutated Hsp90 clients or amplification of clients such as HER2. The hypoxic, low pH, and low nutrient conditions found in many tumors may further increase the number of denatured proteins found in tumors, and thus the need for Hsp90 chaperone function. In support of this hypothesis, Kamal et al. have shown that Hsp90 in tumor cells is found entirely in an active complex with co-chaperones, whereas most Hsp90 in normal tissues resides in a free, uncomplexed, or latent state [16]. These data suggest that the sensitivity of some cancer cells to Hsp90 inhibition may not be attributable to their reliance on any individual Hsp90 client or clients, but rather to their increased dependence upon Hsp90 chaperone function. This may explain why therapies that further stress the chaperone system such as anti-angiogenic agents, proteasome inhibitors, cytotoxic chemotherapies, and radiation may broadly synergize with Hsp90 inhibitors even in tumors in which Hsp90 inhibitors alone have little or no activity 1, 2, 3.

Finally, the selective sensitivity of transformed cells for Hsp90 inhibitors may be partly due to the selective accumulation of these drugs in cancer cells [15]. The selective accumulation of Hsp90 inhibitors in tumors has been reported with compounds that display little structural homology and diverse solubility profiles. One possible explanation for this result is that the binding affinity of these compounds may be higher for tumor-derived Hsp90 than recombinant Hsp90 or Hsp90 derived from non-transformed cells because of the higher abundance of activated, co-chaperone bound Hsp90 found in tumors 15, 16.

Section snippets

Application of Hsp90 inhibitors in the treatment of cancer patients (Table 2)

Though useful as biologic probes for studying the role of Hsp90 in mediating transformation, the natural products GM and radicicol (RD) (Figure 1) have several pharmacologic limitations, including poor solubility, limited in vivo stability, and off-target toxicities that have precluded their use as drugs [9]. GM proved too toxic for human use [17] but 17AAG (Figure 1), a carbon-17 substituted derivative, retains activity against Hsp90 but with a more favorable toxicity profile [18]. 17AAG

Development of second generation Hsp90 inhibitors

On the basis of more recent clinical experience, the limited efficacy observed in the initial phase 1 trials of 17AAG was probably due to a lack of patient enrichment for those most likely to benefit and suboptimal target inhibition due to the requirement for intravenous dosing and the off-target toxicities of 17AAG and its DMSO formulation. These findings have catalyzed future efforts directed at both improving the delivery of 17AAG and identifying novel scaffolds with improved pharmacologic

Neurodegenerative diseases

The important functions played by Hsp90 in maintaining the functional stability and viability of cells under a transforming pressure may be clinically exploitable in other pathogenic transformations, such as neurodegeneration. Tauopathies, such as Alzheimer's disease (AD) and frontotemporal dementia (FTD), are neurodegenerative diseases in which transformation is characterized by abnormalities in the protein tau leading to an accumulation of hyperphosphorylated and aggregated tau toxic species

Conclusions

Many cells under a transforming pressure, whether malignant, neurodegenerative, or infective, co-opt Hsp90 to maintain the stability and folding of aberrant, transformation-driving proteins, and to regain a ‘pseudo-stable’ state. As these effects occur in a cell-specific and transformation-specific manner, one may envision that by inhibition of one protein – Hsp90 – biologic activity may be obtained in a wide-range of diseases.

The ability of Hsp90 inhibitors to affect, simultaneously, multiple

Acknowledgements

This work was supported by SynCure Cancer Research Foundation, Susan G. Komen Breast Cancer Foundation, the Translational and Integrative Medicine Research Fund of MSKCC, the Byrne Cancer Research Fund, the Manhasset Women's Coalition against Breast Cancer, the Geoffrey Beene Cancer Research Center of MSKCC and the National Institute of Aging (1R21AG028811).

References (60)

  • C.S. Mitsiades

    Inhibition of the insulin-like growth factor receptor-1 tyrosine kinase activity as a therapeutic strategy for multiple myeloma, other hematologic malignancies, and solid tumors

    Cancer Cell

    (2004)
  • L. Neckers et al.

    Heat-shock protein 90 inhibitors as novel cancer chemotherapeutics—an update

    Expert Opin. Emerg. Drugs

    (2005)
  • M.V. Powers et al.

    Targeting of multiple signalling pathways by heat shock protein 90 molecular chaperone inhibitors

    Endocr. Relat. Cancer

    (2006)
  • G. Chiosis

    Modulating chaperones in transformed systems—a focus on Hsp90 and cancer

    Expert Opin. Ther. Targets

    (2006)
  • L. Whitesell

    Benzoquinonoid ansamycins possess selective tumoricidal activity unrelated to src kinase inhibition

    Cancer Res.

    (1992)
  • L. Whitesell

    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)
  • L. Neckers

    Using natural product inhibitors to validate Hsp90 as a molecular target in cancer

    Curr. Top. Med. Chem.

    (2006)
  • D.B. Solit

    17-Allylamino-17-demethoxygeldanamycin induces the degradation of androgen receptor and HER-2/neu and inhibits the growth of prostate cancer xenografts

    Clin. Cancer Res.

    (2002)
  • S.Y. Sharp

    Inhibition of the heat shock protein 90 molecular chaperone in vitro and in vivo by novel, synthetic, potent resorcinylic pyrazole/isoxazole amide analogues

    Mol. Cancer Ther.

    (2007)
  • I.B. Weinstein

    Cancer. Addiction to oncogenes—the Achilles heal of cancer

    Science

    (2002)
  • P. Workman

    Stress responses in biology and medicine stress of life in molecules, cells, organisms, and psychosocial communities

    Ann. N. Y. Acad. Sci.

    (2007)
  • G. Chiosis et al.

    Tumor selectivity of Hsp90 inhibitors—the explanation remains elusive

    ACS Chem. Biol.

    (2006)
  • A. Kamal

    A high-affinity conformation of Hsp90 confers tumour selectivity on Hsp90 inhibitors

    Nature

    (2003)
  • J.G. Supko

    Preclinical pharmacologic evaluation of geldanamycin as an antitumor agent

    Cancer Chemother. Pharmacol.

    (1995)
  • T.W. Schulte et al.

    The benzoquinone ansamycin 17-allylamino-17-demethoxygeldanamycin binds to HSP90 and shares important biologic activities with geldanamycin

    Cancer Chemother. Pharmacol.

    (1998)
  • U. Banerji

    Phase I pharmacokinetic and pharmacodynamic study of 17-allylamino, 17-demethoxygeldanamycin in patients with advanced malignancies

    J. Clin. Oncol.

    (2005)
  • M.P. Goetz

    Phase I trial of 17-allylamino-17-demethoxygeldanamycin in patients with advanced cancer

    J. Clin. Oncol.

    (2005)
  • J.L. Grem

    Phase I and pharmacologic study of 17-(allylamino)-17-demethoxygeldanamycin in adult patients with solid tumors

    J. Clin. Oncol.

    (2005)
  • R.K. Ramanathan

    Phase I pharmacokinetic-pharmacodynamic study of 17-(allylamino)-17-demethoxygeldanamycin (17AAG, NSC 330507), a novel inhibitor of heat shock protein 90, in patients with refractory advanced cancers

    Clin. Cancer Res.

    (2005)
  • D.B. Solit

    Phase I trial of 17-allylamino-17-demethoxygeldanamycin in patients with advanced cancer

    Clin. Cancer Res.

    (2007)
  • Cited by (0)

    View full text