Targeted cancer therapy through 17-DMAG as an Hsp90 inhibitor: Overview and current state of the art

Targeted cancer therapy through 17-DMAG as an Hsp90 inhibitor: Overview and current state of the art

Biomedicine & Pharmacotherapy 102 (2018) 608–617 Contents lists available at ScienceDirect Biomedicine & Pharmacotherapy journal homepage: www.elsev...

1MB Sizes 0 Downloads 26 Views

Biomedicine & Pharmacotherapy 102 (2018) 608–617

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy journal homepage: www.elsevier.com/locate/biopha

Review

Targeted cancer therapy through 17-DMAG as an Hsp90 inhibitor: Overview and current state of the art

T

Hassan Mellatyara,b, Sona Talaeia,b, Younes Pilehvar-Soltanahmadia,b, Abolfazl Barzegarc, ⁎ Abolfazl Akbarzadehd, Arman Shahabie, Mazyar Barekati-Mowahedf, Nosratollah Zarghamia,b, a

Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran c Research Institute for Fundamental Sciences (RIFS), University of Tabriz, Tabriz, Iran d Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran e Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran f Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA b

A R T I C LE I N FO

A B S T R A C T

Keywords: Heat shock protein 90 17-DMAG Cancer therapy Inflammatory diseases

Heat shock protein 90 (Hsp90) is an evolutionary preserved molecular chaperone which mediates many cellular processes such as cell transformation, proliferation, and survival in normal and stress conditions. Hsp90 plays an important role in folding, maturation, stabilization and activation of Hsp90 client proteins which all contribute to the development, and proliferation of cancer as well as other inflammatory diseases. Functional inhibition of Hsp90 can have a massive effect on various oncogenic and inflammatory pathways, and will result in the degradation of their client proteins. This turns it into an interesting target in the treatment of different malignancies. 17-dimethylaminoethylamino-17-demethoxygeldanamycin (17-DMAG) as a semi-synthetic derivative of geldanamycin, has several advantages over 17-Allylamino-17-demethoxygeldanamycin (17-AAG) such as higher water solubility, good bioavailability, reduced metabolism, and greater anti-tumour capability. 17-DMAG binds to the Hsp90, and inhibits its function which eventually results in the degradation of Hsp90 client proteins. Here, we reviewed the pre-clinical data and clinical trial data on 17-DMAG as a single agent, in combination with other agents and loaded on nanomaterials in various cancers and inflammatory diseases.

1. Hsp90 and its biological role Heat shock protein 90 is the most plentiful molecular chaperone in eukaryotic organisms which comprises about 1–2% of cytosolic proteins [1–3]. Hsp90 is tightly conserved in the course of evolution from bacteria to homo sapiens. This implies its essential niche in several cellular processes including cell transformation, proliferation, and survival under normal and stressed conditions [4,5]. Eukaryotic cells have three types of Hsp90s: cytosolic Hsp90 with two isoforms of Hsp90α and Hsp90β, Grp94 (glucose-regulated protein 94) of the endoplasmic reticulum (ER) and mitochondrial Trap1 (tumor necrosis receptor-associated protein 1) [6,7]. Hsp90 is composed of three functional domains: N-terminal, middle and C-terminal domains. All of the domains discussed bind to the ATP which is a primary function of Hsp90 [2,6] (Fig. 1). Hsp90 has a primary role in folding, maturation, stabilization and activation of a wide range proteins which are known as Hsp90 client proteins in both normal and cancer cells [8]. In normal cells, Hsp90 also



plays a key role in intracellular transport, cell signaling and maintenance of genome stability [9]. In these cells, freshly synthesized or stress-induced denatured client proteins achieve an innate state which is mediated via Hsp90. Hsp90 also protects these proteins from proteasomal degradation [10,11]. To materialize this, Hsp90 forms the multi-chaperone complex known as the Hsp90 chaperone machine [12]. This complex which is made up from Hsp90 juxtaposed to Hsp70, Hsp40, P23, cdc37, immunophilins (IPs) and HOP (Hsp70 and Hsp90 organizing protein) [3,13,14]. The Hsp90 chaperone machinery is regulated via the consecutive binding and hydrolysis of ATP [15] (Fig. 2). On the other hand, Hsp90 plays an essential role in the assembly and maintenance of the 26 S proteasome that is responsible for degradation of misfolded and damaged proteins marked for destruction by the polyubiquitation pathway in normal conditions of eukaryotic cells [16]. The Hsp90 client proteins can be classified into three main classes: steroid hormone receptors, tyrosine and serine/threonine kinases, and proteins with various other functions [17–20] (Table 1). These proteins

Corresponding author at: Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, 13191-45156, Iran. E-mail address: [email protected] (N. Zarghami).

https://doi.org/10.1016/j.biopha.2018.03.102 Received 16 January 2018; Received in revised form 6 March 2018; Accepted 17 March 2018 0753-3322/ © 2018 Elsevier Masson SAS. All rights reserved.

Biomedicine & Pharmacotherapy 102 (2018) 608–617

H. Mellatyar et al.

tumors [21,24]. Also its ATPase activity is enhanced 50X in a cancerous microenvironment [25]. When cells are stressed by external stressors, Hsp90 assists in the recovery from stress through at least two general ways. First, Hsp90 facilitates protein refolding (correct folding) and increases the rate at which a damaged protein is reactivated. Second, Hsp90 directs nonfunctional proteins toward proteasomal degradation by the polyubiquitation pathway. Thus, Hsp90 restores protein homeostasis and promotes cell survival in stress conditions [26,27]. On the other hand, cancer cells overexpress a number of Hsp90 client proteins, including signal transduction proteins and growth factor receptors that degradation of these proteins induce apoptosis [28]. Hsp90 also stabilizes mutant proteins that appear during cell transformation, thereby enabling malignant transformation [29]. Hsp90 significantly contributes to the microenvironment in which the cancer cells thrive. The inhibition and disruption of Hsp90 affects processes involved in the initiation of cancer which all holistically can be regarded as the “Hallmarks of Cancer”. [6,30–32] (Fig. 3). A noticeable chunk of Hsp90 client proteins are involved in stages of carcinogenesis. Accordingly inhibition of Hsp90 by inhibitors and proteasomal degradation of these proteins can be effective in cancer therapy [17,19,33].

Fig. 1. Schematic structure of Hsp90. There are three functional domains in Hsp90: N-terminal domain with ATP-binding site and drugs such as geldanmycin and 17-DMAG, middle domain with binding site for client proteins, and C-terminal homodimerization domain with binding site for ATP. The charged domain provides a flexible linker in structure of Hsp90. EEVD motif within Cterminal domain is essential for the interaction, and is recognized by co-chaperones carrying a tetratricopeptide repeat (TPR) domain. Client protein binding to the middle domain induces conformational changes in structure of Hsp90, and leads to a closed form formation. In closed form, Hsp90 can exert its activity.

2. Discovery and development of 17-DMAG as an Hsp90 inhibitor Geldanamycin is a natural product and a member of the family of benzoquinone ansamycins that was first derived from Streptomyces hygrocopicus [34,35]. Benzoquinone ansamycins have demonstrated anti-tumor and anti-proliferative characteristics [36]. Initially, the potent antitumor activity of geldanamycin on cancer cells was proposed to be done via inhibition of c-Src kinases catalytic activity, but subsequent studies have indicated that inhibition of Hsp90 was responsible for its antitumor activity [13,37].

all possess critical roles in the signal transduction pathways as well as the cell cycle [21–23]. Hsp90 is upregulated in response to external stressors such as heat, nutrient absence and oxidative stress conditions in various human

Fig. 2. The Hsp90 chaperoning cycle. Initially, HSP70, HSP40, HIP and a client protein form an early complex that interacts with the Hsp90 homodimer via the adaptor protein HOP which will result into an intermediate complex. The ATP binding at aminoterminal region of Hsp90, and its following hydrolysis detaches the HSP70, HSP40, HIP and HOP from the intermediate complex and Hsp90 forms a mature complex, containing p23, cdc37 and IPs. This prepares the structural maturation of the client protein. Binding of 17-DMAG to the ATPbinding site of Hsp90, blocks formation of mature complex and leads to the ubiquitin proteasome-dependent degradation of client proteins by the CHIP (C-terminus of HSP70-interacting protein) ligase.

609

Biomedicine & Pharmacotherapy 102 (2018) 608–617

H. Mellatyar et al.

Table 1 Partial list of Hsp90 client proteins. Protein Steroid hormone receptors AR ER Progesterone receptor Ser/Thr and Tyr kinases HER-2 EGFR Raf-1 B-Raf Chk1 IGF1-R FAK Wee1 PLK AKT CDK-4 Bcr-Abl c-MET FLT-3 Proteins with various other functions Mutant p53 HIF-1α hTERT Survivin ZAP-70

Disease

Function

Prostate Breast Breast

Ligand mediated gene transcription [108,109] Ligand mediated gene transcription [110,111] Ligand mediated gene transcription [112]

Breast, Ovarian Lung, Breast, Head and neck and Colorectal Melanoma Melanoma AML Multiple Myeloma Breast, Colon Lung Colorectal Lung NHL CML Head and neck, Lung, Prostate AML

PI3 kinase signaling [113] Signal transduction [114,115] MAPK signaling [116,117] MAPK signaling [116] Cell cycle regulation [118] Signal transduction [119] Actin-based cell motility [120] Cell cycle regulation [121,122] Cell cycle regulation [123,124] PI3 kinase signaling [125,126] Cell cycle regulation [127] Pathogenesis of myeloid leukaemia [128,129] HGF/SF-MET motility signaling [130] PI3K/AKT signaling [131]

Lung, Colorectal Breast Prostate GBM CLL

Mutant form of cell cycle checkpoint protein [132] Hypoxia-induced angiogenesis [133–135] Cell mortality and senescence [136,137] Inhibitor of apoptosis [138] Signal transduction [124]

AR: Androgen receptor; ER: Estrogen receptor; Chk1: Check point kinase 1; EGFR: Epidermal growth factor receptor; IGF1-R: Insulin-like growth factor 1 receptor; HER-2: Human epidermal growth factor receptor 2; AML: Acute myelogenous leukemia; FAK: Focal adhesion kinase; PLK: Polo-1 kinase; CDK-4: Cyclin-dependent kinase-4; NHL: Non-Hodgkin Lymphoma ;CML: Chronic myeloid leukemia; FLT-3: FMS-like tyrosine kinase-3; HIF-1α: Hypoxia-inducible factor-1α; hTERT: Catalytic subunit of telomerase; GBM: Glioblastoma multiforme; ZAP-70: Zeta-associated protein of 70 kDa; CLL: Chronic lymphocytic leukemia.

Fig. 3. Hsp90 and the hallmark traits of cancer cells.

17-DMAG has multiple superiorities which makes it more potent clinical agent vis-à-vis the 17-AAG. These include higher water solubility which leads to the use of an improved formulation. Better bioavailability facilitates the oral use, as well as a reduced metabolism (reduced potential toxic metabolites) which leads to wider distribution to animal tissues, and greater antitumour potency against cancer cell lines in culture and in xenografts models [3,18,50,51]. Also, 17-DMAG has a lower liver toxicity than geldanamycin [52]. These advantages make 17-DMAG superior in humans [53,54]. 17-DMAG binds to the Hsp90 ATP-binding motif, and inhibits the ATP binding, and accordingly the chaperoning role of Hsp90. This results in the misfolding, ubiquitylation and the eventual degradation of the Hsp90 client proteins by the proteasome. [54–56]. It is noteworthy that the 17-DMAG specifically binds to the tumor cells, and inhibits the tumor growth [57] (Fig. 2). Additionally, 17-DMAG decreases the inflammatory response via up-regulating the heat shook proteins (HSPs), and subsequent

Geldanamycin is a compound which competitively binds to the ATP/adenosine diphosphate (ADP)–binding site on Hsp90 and inhibits its intrinsic ATPase activity [10,38]. Geldanamycin was the first clinically used as a Hsp90 inhibitor, but its clinical usage was limited due to liver toxicity, metabolic instability and poor solubility, and promoted the clinicians to use its safer derivatives [39,40]. 17-AAG is an analogue of geldanamycin with higher binding affinity to the ATP binding site of Hsp90, and lower toxicity for use in clinical trials [41–43]. Allyl amino group replaces the methoxy at the 17-position of geldanamycin in this analogue [13]. Although, 17-AAG is now undergoing phase I–III clinical trials for the treatment of patients with solid tumors and various malignancies, its poor water solubility, short stability, potential liver toxicity, short biological half-life, and the requirement to be dissolved in dimethyl sulfoxide (DMSO) and Cremophor EL has limited the progress of its clinical usage [44–48]. 17-DMAG is a semi-synthetic derivative of geldanamycin which differs from 17AAG in position 17 side chain of the ansa ring [18,49].

610

Biomedicine & Pharmacotherapy 102 (2018) 608–617

H. Mellatyar et al.

inhibition of pro-inflammatory transcription factor function [58].

thus function via the modulation Hsp90 [18].

2.1. 17-DMAG in pre-clinical trials

2.1.1.3. Breast and ovarian cancers. HER-2 is a receptor tyrosine kinase. The overexpression of HER-2 enhances the cell proliferation, migration, and invasive in breast and ovarian cancers. HER-2 is deemed to be one of the most sensitive Hsp90 client proteins. With regards to 17-DMAG, it will lead to the inhibition of Hsp90 that accordingly degraded the HER-2 [66].

17-DMAG has been studied in the preclinical and clinical trials. The following summarizes the reported pre-clinical data with 17-DMAG, alone, and in conjunction with other agents. 2.1.1. 17-DMAG and its antiangiogenic and antitumor properties in cancer The transcription factor HIF-1, a client protein of Hsp90, is induced by hypoxia. HIF-1, during the angiogenic response, drives the transcription of vascular endothelial growth factor (VEGF) which is involved in tumor cell hypoxia adaption. Hsp90 also regulates the antiapoptotic properties of VEGF in leukemic and endothelial cells, and VEGF-induced endothelial cell migration in vitro. Therefore, Hsp90 is deemed as primary regulator of signaling pathways which promotes the activation of inducible nitric oxide synthase (iNOS) and proangiogenic effects mediated by the nitric oxide. 17-DMAG inhibits the VEGF and fibroblast growth factor (FGF)-2-induced endothelial cell proliferation. This facilitates the apoptosis through proteasome degradation via c-Raf1, AKT, and extracellular signal-regulated kinase (ERK) protein kinases. Therefore, antiangiogenic properties of 17-DMAG is related to its direct effects on endothelial cell functions [59].

2.1.1.4. Medulloblastoma. The p53 protein is a tumor suppressor which reduces the onset of tumorigenic events by inducing apoptosis in tumor cells. Therefore, a mutation in p53 facilitates the emergence of tumorigenesis, and makes tumor microenvironment robust. Ayrault et al have located a correlation between Hsp90 and p53 activities. This correlation will necessitate that 17-DMAG would require a thorough p53 activity in order to apply its antitumor function. They have suggested that the state of p53 is a probable predictor of the sensitiveness of tumors to 17-DMAG. This may be an efficient treatment for medulloblastoma in which the tumors contain functional p53 [57]. 2.1.1.5. Cervical cancer. FAK, a nonreceptor protein tyrosine kinase, is a primary mediator of integrin signaling. FAK is tightly correlated with neoplasias where it is one of the underlying factors in cell proliferation, resistance to apoptosis and anoikis, angiogenesis, and metastasis. FAK depends on the Hsp90 for its stability and proper activity. Also in the context of cervical cancer, Hsp90 is overexpressed. Schwock et al have elucidated the importance of FAK with respect to the tumor growth in cervical cancer. They also have suggested the disruption of FAK signaling by 17-DMAG via inhibition of Hsp90, perpetuates suppression of tumor growth and metastasis [67].

2.1.1.1. Leukemia. Panarsky et al studied the effect which 17-DMAG exerts in a murine model with localized tumor-like structures at the site of acute myeloid leukemia (AML) cell implantation. They have shown that a treatment of 17-DMAG brings down the level of HIF-1 and VEGF inside measured within the tumor tissues, and impedes the tumor growth, and thus the leukemia [60]. In another study, Ghoshal et al have demonstrated that 17-DMAG has a synergistic effect combined with the arsenic trioxide (ATO). The synergy down-regulates the activation of transcription 3 (STAT3) in patients with AML. Both ATO and17-DMAG contribute to the up-regulation of Hsp70, an antiapoptotic protein. It can be concluded that the down-regulation of Hsp70 enhances their anti-leukemic activity [61]. The transcription factor nuclear factor-κB (NF-κB), becomes overtly functional in chronic lymphocytic leukemia (CLL). Its functions include regulation of a variety of main antiapoptotic proteins and oncogenes, such as c-FLI, Bcl2, MCL1 and XIAP. It is worth mentioning that the NFκB-activated I-κ-B kinase (IKK) complex is a client protein of Hsp90. 17DMAG treatment effectively degrades the presence of IKK in CLL cells, and subsequently suppresses the NF-κB DNA binding, and the transcription of its target genes. Therefore, the caspase-dependent apoptosis ensues [62]. Gao et al investigated the effect of 17-DMAG in proliferation and apoptosis of leukemia cells K562 and acute lymphocytic leukemia cell lines Jurkat. They showed that 17-DMAG inhibits the cell growth and increase the cell apoptosis in dose and time dependent manners. They also indicated that after treatment of K562 and Jurkat cells with 17DMAG, the Hsp90 mRNA expression lessened significantly [63,64].

2.1.1.6. Multiple myeloma. In the context of human multiple myeloma (MM) cells,17-DMAG promotes apoptosis and autophagy via the inhibition of mTOR (mammalian target of rapamycin) and microtubule-associated proteins. The process of light chain 3-I conversion to LC3-II is an indicator of autophagy. Autophagy is a mechanism of intracellular protein degradation which is activated due to a stress exerted on the endoplasmic reticulum. Interestingly enough, Palacios et al have claimed that the inhibition of autophagy increases 17-DMAG-induced apoptosis via the activation of caspase, and thus the release of cytochrome c from mitochondria and cleavage of Poly-ADP ribose polymerase (PARP) in MM cells [68]. 2.1.1.7. Lung cancer. The mutant EGFR is considered to be a Hsp90 client protein. In the context of non-small-cell lung cancers (NSCLC), also, EGFR mutations are seen to be responsive to 17-DMAG. Treatment with 17-DMAG deregulates phosphorylation of EGFR and degrades phospho-EGFR, phospho-Akt and phospho-MAPK in EGFR-mutant cell lines than in EGFR-wild type cell line, and also have promoted apoptosis in EGFR-mutant cell lines [69]. EML4-ALK (echinoderm microtubule-associated protein-like 4-anaplastic lymphoma kinase) fusion oncoprotein, also a Hsp90 client protein, is known to be a primary oncogenic driver in NSCL. Therefore, the 17-DMAG treatment reduces the levels of this proteins. This will distort the oncogenic signaling pathways, and orchestrates apoptosis or cell cycle arrest [70]. NF-κB signaling pathway has a vital function in the rise of lung cancer. This pathway is induced by the tumor necrosis factor (TNF), a key player in a wide spectrum of cellular processes. TNF contributes to the tumor development via induction of cell proliferation and survival signaling pathways. On the other hand, it potentially could induce apoptosis. IκB kinase (IKK) complex formation includes IKKα, IKKβ, and IKKγ. IKK is essential during the TNF-induced signaling. Since IKKβ is identified as a client protein of Hsp90,17-DMAG treatment reduces the levels of IKKβ in lung cancer cells. Therefore, a combination of 17-

2.1.1.2. Melanoma. Hollingshead et al studied the in vivo antitumor effectiveness of 17-DMAG in metastatic pancreatic carcinoma. They also made use of subcutaneous xenograft melanoma and lung carcinoma models for further development. They have demonstrated that 17-DMAG possesses antitumor activity in orthotropic and subcutaneous models of pancreatic cancer as well as melanomas when administered orally. Further, 17-DMAG has a better activity compared with the 17-AAG in parenteral routes when administered to subcutaneous melanoma and lung carcinoma xenografts [65]. Using the molecular results obtained from treating melanoma cells with both 17-AAG and 17-DMAG, Smith et al comparatively studied their in vitro antitumor activities. They elaborated that 17-DMAG has higher antitumor activity than 17-AAG within the melanoma cells. They also concluded which of the two drugs have similar mechanisms, and 611

Biomedicine & Pharmacotherapy 102 (2018) 608–617

H. Mellatyar et al.

tumor radiosensitivity. This sensitization seemed to be as a result of abrogation of the G2 and S phases checkpoints. Levels of radiosensitivity-associated proteins, Akt, Raf-1, and ErbB2, are all lessened when they are exposed to 17-DMAG in the tumor cells [78]. In the past recent years, the radioprotector effects of 17-DMAG in Tcells may prove effective in the radiation-based cancer therapy. Radiation exposure raises iNOS expression and nitric oxide (NO) production by NF-κB and Kruppel like factor 6 (KLF6) transcription factors. The production of iNOS-mediated NO increases of caspase 3 activity and results in apoptosis and cell death. Via inhibiting the up-regulation of iNOS, and blocking the production of NO and the subsequent caspase-3 activation 17-DMAG protects the T-cells from radiation-induced apoptosis [79]. In addition to the cases discussed above, ionizing radiation increases p53 aggregation, acute p53 phosphorylation and Bax expression in T cells. Also, p53 – Hsp90 interaction occurs post-irradiation. 17-DMAG, via inhibiting the radiation-induced p53 phosphorylation, and the preventing p53 – Hsp90 complex formation, lowers the p53 level and p53-dependant apoptosis in T cells [52].

DMAG and TNF treatments may be potentially effective in the treatment of lung cancer cells [71]. 2.1.1.8. Liver cancer. Survivin, cyclin D1, NF-κB and p53, all client proteins of Hsp90, are the central proteins in the primary liver cancer. Hsp90 is deemed to play a main role in hepatocellular carcinoma, and tumor survival through regulation of these proteins levels. 17-DMAG inhibits Hsp90, and induces apoptosis by degrading the survivin, which is a strong inhibitor of apoptosis. Also, the relocation of NF-κB to the nucleus is downscaled. On the same token the upregulation of p53 protein levels through depletion of its inhibitors, and reduction of cyclin D1 expression all occur [49]. Zhang et al have shown that the Hsp90 might be prominent in the cell cycle control of hepatocellular carcinoma cells. This occurs by the regulating the levels of cyclin B1, a Hsp90 client protein, that is crucial for the G2/M phase transition in cell cycle. They reported that 17DMAG induces aggregation of cyclin B1, therefore, prevents tumor cell growth [72].

2.1.2. 17-DMAG loaded in nanocarriers and its antitumor properties in cancer The rapid progress of nanotechnology provides alternative attitudes to overcome numerous drawbacks of conventional anti-cancer treatment [80]. Drug targeting employing functionalized nanoparticles to improve their transport to the dedicated location, became a new criterion in novel antineoplastic approaches [81–83]. In effect, the utilization of nanoparticles during design of anti-cancer drugs aids to enhance pharmacokinetic properties, with subsequent development of non-toxic, high specific and biocompatible anti-cancer drugs [84–86]. There have been two nano-formulation studies of 17-DMAG that have been reported so far by our group. In these studies, polymeric nanoparticles (NPs) used for the delivery of 17-DMAG. Polylactidecoglycolide–polyethylene glycol ((PLGA-PEG)) is one of the most common co-polymers that are used to encapsulate the drugs. Studies have shown that encapsulation of drug to (PLGA-PEG) lowers the dose of drug, and its adverse side effects [87]. We studied inhibitory effects of the (PLGAPEG)-17-DMAG complex in the context of lung and breast cancers. The results demonstrated that the complex can be more effective when compared to the 17-DMAG when it comes to down-regulation of Hsp90 expression. It is noteworthy to mention that this happens through enhancing uptake by cells [88,89].

2.1.1.9. Neuroblastoma. Anaplastic lymphoma kinase (ALK), a receptor tyrosine kinase, regulates transcription of proto-oncogene, MYCN, in neuroblastoma (NB) cells. The amplification of ALK and MYCN is commonly found in high-risk NB patients. N-Myc protein encoded by MYCN gene and activated ALK act as the downstream effector of AKT/ phosphatidylinositol-3 kinase (PI3K), RAS/ERK and STAT3 signaling pathways. This can promote cell proliferation and migration, induce cell transformation, inhibit neural cell differentiation, and prevent cell apoptosis. Therefore, application of 17-DMAG through the degradation of proteins involved in these pathways inhibits the NB cells growth and induces apoptosis [73]. 2.1.1.10. Colon cancer. Ma et al have identified 16 proteins related to the 17-DMAG treatment in colon cancer cells. They have suggested that Hsp71 (Heat shock 70 kDa protein 1 A/1B) as well as SRC8 (Cortactin) might be potential client proteins of Hsp90. They have elucidated that the 17-DMAG treatment enhances the expression level of Hsp71 and SRC8, and thus inhibits cell proliferation, and induces apoptosis in colon cancer cells [74]. Glucose-regulated protein 78 (GRP78), an ER stress protein, is a member of the Hsp70 family. GRP78 is a chief biomarker of various cancers. It also reciprocally prevents apoptosis by inhibiting Bad and Bax activation and caspase 7. A decrease in the expression levels of GRP78 elevates the 17-DMAG treatment efficiency in colon cancer cells. In the light of this, GRP78 may be a powerful predictive marker of 17DMAG efficiency. Colon cancer cells treated with 17-DMAG post-GRP78 knock down exhibited a reduced level of Bcl-2, subsequently and an increased level of Bad and Bax [75].

2.1.3. The 17-DMAG and its antioxidant and anti-inflammatory properties in inflammatory diseases 17-DMAG carries antiangiogenic characteristics in cancer cells, but in tibial dyschondroplasia(TD), it does the opposite. It switches, and thus enhanced the blood-vessel formation, mediated by the VEGF signaling pathway [90]. Shahzad et al. have scrutinized the antioxidant effects of 17-DMAG in the thiram-induced TD. They showed that the 17DMAG recuperate the liver damages caused by thiram [91]. In the other study, Madrigal-Matute et al. investigated the antioxidant properties of 17-DMAG in atherosclerosis. As previously discussed the 17-DMAG regulates several signaling pathways such as mitogen-activated protein kinases (MAPKs). MAPK kinase (MEK), a client protein of Hsp90, is involved in the phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2). ERK1/2 upon activation regulates main processes of vascular smooth muscle cells (VSMCs) in atherosclerosis. Therefore, reduced levels of ERK1/2 activation observed in 17-DMAG-treated VSMCs. 17-DMAG also lowers the reactive oxygen species (ROS) levels. Their participation is the underlying mechanism in the atherogenesis. On the other hand, treatment with 17DMAG up-regulates atheroprotective HSPs (HSP27 and HSP70) levels in VSMCs [58]. Thus, results suggest that 17-DMAG inhibits oxidative stress via reducing the pro-oxidative factors in the pathogenesis of atherosclerosis [92]. 17-DMAG exerts its anti-inflammatory effects through various

2.1.1.11. Gastric cancer. Kim et al studied the anticancer effects of 17DMAG in gastric cancer. They demonstrated that 17-DMAG, exerts its inhibitory effects against gastric cancer cells through down- and upregulation of antioxidant enzymes and ROS, respectively. Therefore, they identified a new therapeutic approach based on non-canonical (ROS-generating) pathway in gastric cancer treatment [76]. 2.1.1.12. Lymphoma. Signaling pathways proteins e.g., c-RAF, AKT, ZAP-70, IKKα, and the cell cycle regulatory proteins e.g., CDK4, p21, CHK1 are key proteins in mantle cell lymphoma (MCL) and known as Hsp90 client proteins. Co-treatment with 17- DMAG and vorinostat (pan-histone deacetylase inhibitor) synergistically induces the apoptosis of MCL cells. 17-DMAG promotes proteasomal degradation of Hsp90 client proteins, and arrests the MCL cells in the G2-M phase of cell cycle. Vorinostat facilitates the hyperacetylation of Hsp90 and disrupts its association with neighboring client proteins and cochaperones [77]. 17-DMAG also exerts its antitumor effects via enhancement of 612

613

weekly 80 mg/m2 Trastuzumab 4 mg/kg

Diarrhea, headache, myalgia, fatigue, nausea, arthralgia, blurry vision, back pain and dry eyes [106]

Twice weekly weekly 24 mg/m2 80 mg/m2

28

24 25

Table 2 17-DMAG in Phase I Trials.

2.2.1. Pharmacokinetics 17-DMAG went into Phase I clinical trials in several different dosing schedules. The 17-DMAG area under the curve (AUC) raised linearly with dosages from 1.5 mg/m2 to 80 mg/m2 [102–106]. On a daily × 3

31

2.2. 17-DMAG in clinical trials Having shown meaningful antitumor activities in pre-clinical studies in animals and Pediatric Preclinical Testing Program in vitro models [99–101], 17-DMAG has been studied in a number of phase I trials as a single drug, and also in combination with other drugs in hematological malignancies as well as solid tumors (Table 2).

Liver function test elevation, diarrhea, pneumonitis, fatigue nausea and thrombocytopenia [102] Peripheral neuropathy and renal dysfunction [104] daily × 3 or 5 d, every 3 weeks Twice-weekly 16 mg/m2 × 5 days or 25 mg/m2 × 3 days, every 3 weeks 21 mg/m2

Colorectal (17), Esophageal (5), Ovarian (4), Sarcoma (4), Head and neck (4), Breast (3), Salivary gland (3), Neuroendocrine (3), Melanoma (3), Prostate (2),Lung (2), Other (6) Colorectal (6), Lung (4), Pancreas (3), Peritoneal mesothelioma (1), Pheochromocytoma (1), Other (16) Acute myeloid leukemia Melanoma (7), Soft tissue sarcoma (3), Prostate (3), Breast (3), Colon (2), Pancreas (2), Cervix (2), Cholangiocarcinoma (1), Kidney (1), Uterine (1), Breast (25), Ovarian (3) 56

Schedule Cancer Number of Patients

Recommended Phase II Dose

DLTs

mechanisms. Among them is the degradation of NF-κB in autoimmune diseases. The Hsp90 has a vital niche in antigen presentation and activation of lymphocytes, the Hsp90 inhibition can be a useful therapy to improve the inflammatory pathways in autoimmune diseases. T helper 1 (Th1) and T helper 17 (Th17) cells are necessary to progress of different autoimmune diseases and Hsp90 function is required for interferon-γ (IFN-γ) and interleukin 17 (IL-17) signaling in these cell types. NF-κB is one of the main transcription factors responsible for expression and secretion of proinflammatory IFN-γ, TNF-α, and IL-17. 17-DMAG through degradation of NF-κB suppresses of IFN-γ and IL-17 expression on CD4+ T cells and decreases percentages of Th1 and Th17 cells [93]. 17-DMAG also by inhibiting NF-κB activation and subsequent NO/iNOS pathway and TNF-α release, limits hemorrhage-induced injury in small intestine [94]. Hsp90 also plays a crucial role in production of iNOS, interleukin-6 (IL-6) and IL-12, as well as activation of Toll-like receptor (TLR). Inhibition of Hsp90 by 17-DMAG leads to the enhancement of CD8+T cells, and reduction of double-negative T cells, CD4+/CD8+ ratio and follicular B cells in the mice suffering from systemic lupus erythematosus (SLE). Thus, the results also suggest that 17-DMAG hold an anti-inflammatory effect in lupus [95]. The other mechanism is related to reduction of proinflammatory kinase Akt and IKK expression. Lipopolysaccharide (LPS) and IFN-γ stimulate inflammatory mediator production in macrophages and lead to signal transduction through activation of signaling pathways including Akt/mTOR and IKK/NF-κB. The signal transduction through these pathways results in the production of TNF-α, IL-6, and NO. 17DMAG by suppressing the Akt/NF-κB pathway and decreasing expression of IL-6 and NO, reduces inflammation in LPS/IFN- γ stimulated macrophages [96]. The next mechanism is the induction of HSP70 and heme oxygenase (HO)-1 (an antioxidant protein) production. This will prevent the LPSinduced multiple organ dysfunction syndrome (MODS) in sepsis. LPS binds to toll-like receptor 4 on the membrane of macrophage/neutrophils, and activates the NF-κB pathway. The expression of TNF-α, IL6 and iNOS, and an increase in the production of NO will ensue. The release of superoxide onions, initiated by the activation of macrophages and neutrophils, induce the onset of MODS. Meanwhile, HSP70 plays a substantial role in the protection of organs from bacterial infection and acute inflammation-induced damages. In the context of sepsis, 17DMAG activates HSF-1 which induces the expression of HSP-70, this will tones-down the oxidative stress due to LPS [97]. EphA2 is a receptor tyrosine kinases (RTK) that has key responsibilities including cell survival, proliferation, migration, and differentiation. This protein is found to be overexpressed in tumor cells, facilitated by Hsp90. Rao et al. have reported that 17-DMAG functions as an immune adjuvant upon administration anti-EphA2 vaccines. 17DMAG functions as an immune adjuvant via reduction of myeloid-derived suppressor cells and regulatory T cells in the tumor microenvironment. It also promotes the activation of tumor-associated vasculature and promotion of production of chemokines that recruit Type1 tumor-infiltrating lymphocytes, and enhancement of proteasome degradation of EphA2 and recognition of tumor cells through EphA2specific CD8+ T cells [98].

Neutropenic fever, fatigue, nausea and diarrhea [103] Gastrointestinal, liver function changes, and ocular [105]

Biomedicine & Pharmacotherapy 102 (2018) 608–617

H. Mellatyar et al.

Biomedicine & Pharmacotherapy 102 (2018) 608–617

H. Mellatyar et al.

Table 3 NIH Funded Terminated and Completed Trials. Disease

Phase

Drugs

Status

Her2 Positive Breast Cancer Lymphoma Small Intestine Cancer Unspecified Adult Solid Tumor, Protocol Specific Metastatic Solid Tumors or Tumors That Cannot Be Removed by Surgery Solid Tumor Breast Cancer Metastatic or Unresectable Solid Tumors Metastatic or Unresectable Solid Tumors or Lymphomas Relapsed Chronic Lymphocytic Leukemia, Small Lymphocytic Lymphoma, or B-Cell Prolymphocytic Leukemia

II I I I

Alvespimycin(17-DMAG) 17-DMAG 17-DMAG Alvespimycin Trastuzumab Paclitaxel Alvespimycin Hydrochloride 17-DMAG Alvespimycin Hydrochloride

Terminated Completed Unknown Completed

or 5 d, every 3 weeks’ schedule with dosages from 1.5 to 46 mg/m2, maximum plasma concentration (Cmax) for 17-DMAG were reported as 0.071 to 1.7 μg/mL [102]. Also, on a twice weekly schedule with 21 mg/m2 and 24 mg/m2, Cmax were as 499 ± 274 ng/mL and 475 ng/ ml, respectively [103,104]. The Cmax for patients treated with a dose 80 mg/m2 of 17-DMAG as weekly reported 2680 nmol/L [105]. Urinary excretion of 17-DMAG accounted for approximately 20% of a dose and rapidly cleared through the hepatobiliary system [102]. The terminal half-life of 17-DMAG is 18–24 hours and its clearance averaged is 17 L/hour [102–106].

I I

Completed Completed Terminated

reported prolonged stable disease in patients with carcinoid, melanoma, NSCLC, breast cancer, and salivary gland tumor [102,104,106]. NIH-funded phase I and II clinical trials of 17-DMAG summarize in Table 3. 3. Conclusion and future prospects This review attempted to summarize the anticancer, antiangiogenic, antioxidant, as well as anti-inflammatory effects of 17-DMAG in several signaling pathways. It also studies the interactions of 17-DMAG with the proteins located in these pathways. The pre-clinical data and clinical trial data show us the 17-DMAG when combined with ATO, vorinostat, radiation, and trastuzumab is more clinically efficient. This suggests that 17-DMAG in combination with anticancer drugs, may be suitable to achieve a clinically significant anti-cancer response. Also, to prevent the development of drug resistance in cancer therapy, 17DMAG can be combined with other potent anticancer agents. The role of 17-DMAG in cancer therapy in present time is in a state of evolution. 17-DMAG needs to be studied in the phase II and III clinical trials as part of combination therapy for solid tumors. There are efforts to gain new formulations of 17-DMAG with approved pharmacokinetics and pharmacodynamics properties. More over when it is in conjunction with NPs as opposed to single use, it also exerts more efficacy. The nanomaterials, also have the potential to lessen the side effects, facilitate intracellular administration and ameliorate the clinical efficacy of 17-DMAG. It is expected that advance in progress of nanotechnologybased anti-cancer materials will provide modern, individualized anticancer therapies based on 17-DMAG ensuring reduction in cancer morbidity and mortality. Despite the abundant advancements made in the discovery and development of Hsp90 inhibitors, none of these inhibitors have yet successfully reached the market. Therefore, it is hoped that safe, effective and approved 17-DMAG formulations will become available for cancer therapy in near future.

2.2.2. Toxicity The most common dose-limiting toxicities (DLTs) of administration of 17-DMAG were fatigue, nausea, vomiting, diarrhea, anorexia and liver enzyme disturbances [102–106]. Ocular toxicity mentioned as a concern for further development of 17-DMAG [106] and patients with ocular adverse events including blurry vision, dry eye, keratitis, and conjunctivitis, or ocular surface disease reported by Pacey et al. All ocular adverse events were Grade 2 or less [105]. Cardiac DLTs including acute myocardial infarction, and rise in troponin were also observed upon the 17-DMAG administration [103,107]. DLTs of peripheral neuropathy and renal dysfunction were reversible upon the discontinuation of 17-DMAG. The grade 1 musculoskeletal pain associated with 17-DMAG administration was frequently observed in patients [104]. 2.2.3. Pharmacodynamics Peripheral blood mononuclear cells (PBMCs) were collected pretreatment and following the administration of 17-DMAG to assess the inhibition which was imposed on Hsp90. Pacey et al. have investigated the Hsp72 induction and client proteins, LCK and Cdk4, depletion 24 h after the 17-DMAG administration in PBMCs. The PBMCs and tumor biopsy samples obtained confirmed the notion that the function of Hsp90 was indeed inhibited [105]. Further, the induction of Hsp70 and the depletion of Akt and pAkt client proteins in a time dependent manner was reported by Jhaveri et al [106]. The study in different phase I trials attest upon the wide variability in levels of Hsp70 proteins, and Hsp90′s client proteins in PBMCs. This variability implies the insufficiency of evidence whether they can be considered as a substitute when the inhibition Hsp90 was assessed [102,104].

Conflict of interest The authors declare no conflict of interest. Acknowledgement This study was financially supported by grant No: 960205 of the Biotechnology Development Council of the Islamic Republic of Iran.

2.2.4. Clinical efficacy In the phase I trial of advanced solid tumors, 17-DMAG was administered intravenously at doses between 2.5–106 mg/m2 weekly. A patient with castration refractory prostate cancer (CRPC) completely responded. Also a patient with metastatic melanoma showed partial response and three patients with chondrosarcoma, CRPC, and renal cancer showed stable disease [105]. Also, in the phase I trials on AML and CLL, 17-DMAG was given at a dosage of 24 mg/m2 twice a week. Antileukemic activity was observed in 4 of 17 patients evaluated. Three patients showed complete remission with incomplete blood count recovery and one patient showed > 50% bone marrow blast reduction [103,107]. Other phase I trials

References [1] A. Singh, et al., Topically applied Hsp90 inhibitor 17AAG inhibits UVR-induced cutaneous squamous cell carcinomas, J. Invest. Dermatol. 135 (4) (2015) 1098–1107. [2] Z.A. Wainberg, et al., Inhibition of HSP90 with AUY922 induces synergy in HER2amplified trastuzumab-resistant breast and gastric cancer, Mol. Cancer Ther. 12 (4) (2013) 509–519. [3] R. Garcia-Carbonero, A. Carnero, L. Paz-Ares, Inhibition of HSP90 molecular chaperones: moving into the clinic, Lancet Oncol. 14 (9) (2013) e358–e369. [4] V. Saxena, Y. Naguib, M.D. Hussain, Folate receptor targeted 17-allylamino-17demethoxygeldanamycin (17-AAG) loaded polymeric nanoparticles for breast cancer, Colloids Surf. B: Biointerfaces 94 (2012) 274–280.

614

Biomedicine & Pharmacotherapy 102 (2018) 608–617

H. Mellatyar et al.

Res. 61 (10) (2001) 4003–4009. [41] G.P. Krishnamoorthy, et al., Molecular mechanism of 17-allylamino-17-demethoxygeldanamycin (17-AAG)-induced AXL receptor tyrosine kinase degradation, J. Biol. Chem. 288 (24) (2013) 17481–17494. [42] J. Page, et al., Comparison of geldanamycin (NSC-122750) and 17-allylaminogeldanamycin (NSC-330507D) toxicity in rats, Proc. Am. Assoc. Cancer Res, (1997). [43] R.K. Ramanathan, et al., 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. 11 (9) (2005) 3385–3391. [44] R. Bagatell, L. Whitesell, Altered Hsp90 function in cancer: a unique therapeutic opportunity, Mol. Cancer Ther. 3 (8) (2004) 1021–1030. [45] W.B. Pratt, D.O. Toft, Regulation of signaling protein function and trafficking by the hsp90/hsp70-based chaperone machinery 1, Exp. Biol. Med. 228 (2) (2003) 111–133. [46] A.G. Ortiz, J.M. Salcedo, Heat shock proteins as targets in oncology, Clin. Transl. Oncol. 12 (3) (2010) 166–173. [47] M.A. Biamonte, et al., Heat shock protein 90: inhibitors in clinical trials, J. Med. Chem. 53 (1) (2010) 3–17. [48] H.A. Burris, et al., Tanespimycin pharmacokinetics: a randomized dose-escalation crossover phase 1 study of two formulations, Cancer Chemother. Pharm. 67 (5) (2011) 1045–1054. [49] A.M. Leng, et al., The apoptotic effect and associated signalling Of HSP90 inhibitor 17‐DMAG in hepatocellular carcinoma cells, Cell Biol. Int. 36 (10) (2012) 893–899. [50] J. Moreno‐Farre, et al., Development and validation of a liquid chromatography/ tandem mass spectrometry method for the determination of the novel anticancer agent 17‐DMAG in human plasma, Rapid Commun. Mass Spectrom. 20 (19) (2006) 2845–2850. [51] J.M. Jez, et al., Crystal structure and molecular modeling of 17-DMAG in complex with human Hsp90, Chem. Biol. 10 (4) (2003) 361–368. [52] R. Fukumoto, J.G. Kiang, Geldanamycin analog 17-DMAG limits apoptosis in human peripheral blood cells by inhibition of p53 activation and its interaction with heat-shock protein 90 kDa after exposure to ionizing radiation, Radiat. Res. 176 (3) (2011) 333–345. [53] A.E. Kabakov, V.A. Kudryavtsev, V.L. Gabai, Hsp90 inhibitors as promising agents for radiotherapy, J. Mol. Med. 88 (3) (2010) 241–247. [54] M.J. Egorin, et al., Pharmacokinetics, tissue distribution, and metabolism of 17(dimethylaminoethylamino)-17-demethoxygeldanamycin (NSC 707545) in CD 2 F 1 mice and fischer 344 rats, Cancer Chemother. Pharm. 49 (1) (2002) 7–19. [55] L. Neckers, Heat shock protein 90: the cancer chaperone, Heat Shock Proteins in Cancer, Springer, 2007, pp. 231–252. [56] C. Prodromou, et al., Identification and structural characterization of the ATP/ ADP-binding site in the Hsp90 molecular chaperone, Cell 90 (1) (1997) 65–75. [57] O. Ayrault, et al., Inhibition of Hsp90 via 17-DMAG induces apoptosis in a p53dependent manner to prevent medulloblastoma, Proc. Natl. Acad. Sci. 106 (40) (2009) 17037–17042. [58] J. Madrigal-Matute, et al., Heat shock protein 90 inhibitors attenuate inflammatory responses in atherosclerosis, Cardiovasc. Res. 86 (2) (2010) 330–337. [59] G. Kaur, et al., Antiangiogenic properties of 17-(dimethylaminoethylamino)-17demethoxygeldanamycin, Clin. Cancer Res. 10 (14) (2004) 4813–4821. [60] R. Panarsky, F. Reichert, Z. Ben-Ishay, Effectiveness of 17DMAG, a geldanamycin derivative, in murine acute myeloid leukemia, Acta Haematologica 121 (1) (2009) 32–36. [61] S. Ghoshal, et al., Down-regulation of heat shock protein 70 improves arsenic trioxide and 17-DMAG effects on constitutive signal transducer and activator of transcription 3 activity, Cancer Chemother. Pharm. 66 (4) (2010) 681–689. [62] E. Hertlein, et al., 17-DMAG targets the nuclear factor-κB family of proteins to induce apoptosis in chronic lymphocytic leukemia: clinical implications of HSP90 inhibition, Blood 116 (1) (2010) 45–53. [63] F. Gao, et al., Proliferation and apoptosis of cell line K562 treated with HSP90 inhibitor 17-DMAG, Zhongguo shi yan xue ye xue za zhi 25 (4) (2017) 998–1002. [64] F. Ge, et al., Effect of heat shock protein 90 inhibitor 17-DMAG on proliferation and apoptosis of acute lymphocytic leukemia cell line Jurkat, Zhongguo shi yan xue ye xue za zhi 25 (4) (2017) 1011–1015. [65] M. Hollingshead, et al., In vivo antitumor efficacy of 17-DMAG (17-dimethylaminoethylamino-17-demethoxygeldanamycin hydrochloride), a water-soluble geldanamycin derivative, Cancer Chemother. Pharm. 56 (2) (2005) 115–125. [66] G. Niu, et al., Monitoring therapeutic response of human ovarian cancer to 17DMAG by noninvasive PET imaging with 64Cu-DOTA-trastuzumab, Eur. J. Nucl. Med. Mol. Imag. 36 (9) (2009) 1510. [67] J. Schwock, et al., Targeting focal adhesion kinase with dominant-negative FRNK or Hsp90 inhibitor 17-DMAG suppresses tumor growth and metastasis of SiHa cervical xenografts, Cancer Res. 69 (11) (2009) 4750–4759. [68] C. Palacios, et al., Autophagy inhibition sensitizes multiple myeloma cells to 17dimethylaminoethylamino-17-demethoxygeldanamycin-induced apoptosis, Leuk. Res. 34 (11) (2010) 1533–1538. [69] N. Kobayashi, et al., The anti-proliferative effect of heat shock protein 90 inhibitor, 17-DMAG, on non-small-cell lung cancers being resistant to EGFR tyrosine kinase inhibitor, Lung Cancer 75 (2) (2012) 161–166. [70] H.J. Kim, et al., P-glycoprotein confers acquired resistance to 17-DMAG in lung cancers with an ALK rearrangement, BMC Cancer 15 (1) (2015) 553. [71] Z. Qu, et al., Combined effects of 17-DMAG and TNF on cells through a mechanism related to the NF-kappaB pathway, Diagn. Pathol. 8 (1) (2013) 70. [72] J. Zhang, et al., Hypoxia attenuates Hsp90 inhibitor 17-DMAG-induced cyclin B1

[5] K.S. Pedersen, et al., Phase II trial of gemcitabine and tanespimycin (17AAG) in metastatic pancreatic cancer: a Mayo clinic phase II consortium study, Invest. New Drugs 33 (4) (2015) 963–968. [6] P. Workman, et al., Drugging the cancer chaperone HSP90, Ann. N.Y. Acad. Sci. 1113 (1) (2007) 202–216. [7] G. Chiosis, Heat shock proteins in disease–from molecular mechanisms to therapeutics, Curr. Top. Med. Chem. 16 (25) (2016) 2727. [8] M. Waza, et al., 17-AAG, an Hsp90 inhibitor, ameliorates polyglutamine-mediated motor neuron degeneration, Nat. Med. 11 (10) (2005) 1088–1095. [9] K.B. Kaplan, R. Li, A prescription for ‘stress’–the role of Hsp90 in genome stability and cellular adaptation, Trends Cell Biol. 22 (11) (2012) 576–583. [10] R. Nimmanapalli, et al., Regulation of 17-aag—induced apoptosis: role of bcl-2, Bcl-x L, and Bax downstream of 17-aag—mediated down-regulation of Akt, Raf-1, and Src kinases, Blood 102 (1) (2003) 269–275. [11] T.-y. Lin, et al., HSP90 inhibitor encapsulated photo-theranostic nanoparticles for synergistic combination cancer therapy, Theranostics 6 (9) (2016) 1324. [12] J. Trepel, et al., Targeting the dynamic HSP90 complex in cancer, Nat. Rev. Cancer 10 (8) (2010) 537–549. [13] M.P. Goetz, et al., The Hsp90 chaperone complex as a novel target for cancer therapy, Ann. Oncol. 14 (8) (2003) 1169–1176. [14] M. Stope, et al., Jump in the fire—heat shock proteins and their impact on ovarian cancer therapy, Crit. Rev. Oncol. Hematol. 97 (2016) 152–156. [15] S. Pacey, et al., A phase II trial of 17-allylamino, 17-demethoxygeldanamycin (17AAG, tanespimycin) in patients with metastatic melanoma, Invest. New Drugs 30 (1) (2012) 341–349. [16] J. Imai, et al., The molecular chaperone Hsp90 plays a role in the assembly and maintenance of the 26S proteasome, EMBO J. 22 (14) (2003) 3557–3567. [17] C. Wang, et al., Heat shock proteins in hepatocellular carcinoma: molecular mechanism and therapeutic potential, Int. J. Cancer 138 (8) (2016) 1824–1834. [18] V. Smith, et al., Comparison of 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17DMAG) and 17-allylamino-17-demethoxygeldanamycin (17AAG) in vitro: effects on Hsp90 and client proteins in melanoma models, Cancer Chemother. Pharm. 56 (2) (2005) 126–137. [19] D.A. Proia, G.F. Kaufmann, Targeting heat-shock protein 90 (HSP90) as a complementary strategy to immune checkpoint blockade for cancer therapy, Cancer Immunol. Res. 3 (6) (2015) 583–589. [20] S. Sulthana, et al., Combination therapy of NSCLC using Hsp90 inhibitor and doxorubicin carrying functional nanoceria, Mol. Pharm. 14 (3) (2017) 875–884. [21] M. Saif, et al., Open-label, dose-escalation, safety, pharmacokinetic, and pharmacodynamic study of intravenously administered CNF1010 (17-(allylamino)-17demethoxygeldanamycin [17-AAG]) in patients with solid tumors, Cancer Chemother. Pharm. 71 (5) (2013) 1345–1355. [22] J.C. Young, I. Moarefi, F.U. Hartl, Hsp90, J. Cell Biol. 154 (2) (2001) 267–274. [23] K. Richter, J. Buchner, Hsp90: chaperoning signal transduction, J. Cell. Physiol. 188 (3) (2001) 281–290. [24] S. Tukaj, et al., Topically applied Hsp90 blocker 17AAG inhibits autoantibodymediated blister-inducing cutaneous inflammation, J. Invest. Dermatol. 137 (2) (2017) 341–349. [25] A. Taiyab, A.S. Sreedhar, C.M. Rao, Hsp90 inhibitors, GA and 17AAG, lead to ER stress-induced apoptosis in rat histiocytoma, Biochem. Pharmacol. 78 (2) (2009) 142–152. [26] C. Jolly, R.I. Morimoto, Role of the heat shock response and molecular chaperones in oncogenesis and cell death, J. Natl. Cancer Inst. 92 (19) (2000) 1564–1572. [27] D. Parsell, S. Lindquist, The function of heat-shock proteins in stress tolerance: degradation and reactivation of damaged proteins, Annu. Rev. Genet. 27 (1) (1993) 437–496. [28] G. Lurje, H.-J. Lenz, EGFR signaling and drug discovery, Oncology 77 (6) (2009) 400–410. [29] S.K. Calderwood, et al., Heat shock proteins in cancer: chaperones of tumorigenesis, Trends Biochem. Sci 31 (3) (2006) 164–172. [30] A. Maloney, P. Workman, HSP90 as a new therapeutic target for cancer therapy: The story unfolds, Expert Opin. Biol. Ther. 2 (1) (2002) 3–24. [31] Y. Miyata, H. Nakamoto, L. Neckers, The therapeutic target Hsp90 and cancer hallmarks, Curr. Pharm. Des. 19 (3) (2013) 347–365. [32] S.Z. Usmani, R. Bona, Z. Li, 17 AAG for HSP90 inhibition in cancer-from bench to bedside, Curr. Mol. Med. 9 (5) (2009) 654–664. [33] U. Banerji, et al., Phase I pharmacokinetic and pharmacodynamic study of 17allylamino, 17-demethoxygeldanamycin in patients with advanced malignancies, J. Clin. Oncol. 23 (18) (2005) 4152–4161. [34] E.V. Batrakova, et al., Effects of pluronic P85 unimers and micelles on drug permeability in polarized BBMEC and caco-2 cells, Pharm. Res. 15 (10) (1998) 1525–1532. [35] Y.-C. Chen, et al., Effects of surface modification of PLGA-PEG-PLGA nanoparticles on loperamide delivery efficiency across the blood–brain barrier, J. Biomater. Appl. 27 (7) (2013) 909–922. [36] J.G. Supko, et al., Preclinical pharmacologic evaluation of geldanamycin as an antitumor agent, Cancer Chemother. Pharm. 36 (4) (1995) 305–315. [37] D.G. Savage, K.H. Antman, Imatinib mesylate—a new oral targeted therapy, New Engl. J. Med. 346 (9) (2002) 683–693. [38] J.P. Grenert, et al., The amino-terminal domain of heat shock protein 90 (hsp90) that binds geldanamycin is an ATP/ADP switch domain that regulates hsp90 conformation, J. Biol. Chem. 272 (38) (1997) 23843–23850. [39] J. Kreuter, Influence of the surface properties on nanoparticle-mediated transport of drugs to the brain, J. Nanosci. Nanotechnol. 4 (5) (2004) 484–488. [40] I. Hostein, et al., Inhibition of signal transduction by the Hsp90 inhibitor 17-allylamino-17-demethoxygeldanamycin results in cytostasis and apoptosis, Cancer

615

Biomedicine & Pharmacotherapy 102 (2018) 608–617

H. Mellatyar et al.

[73]

[74] [75] [76]

[77]

[78]

[79]

[80]

[81]

[82]

[83]

[84]

[85]

[86]

[87] [88]

[89] [90]

[91]

[92] [93]

[94]

[95]

[96]

[97]

[98]

[99]

[100]

[101]

accumulation in hepatocellular carcinoma cells, Cell Stress Chaperones 21 (2) (2016) 339–348. B. Yi, J. Yang, L. Wang, The growth inhibitory effect of 17-DMAG on ALK and MYCN double-positive neuroblastoma cell line, Tumor Biol. 35 (4) (2014) 3229–3235. F. Ma, et al., Proteomic analysis of 17-DMAG effects on colon cancer HT-29 Cells. (2015). Y.-J. Chang, et al., Glucose-regulated protein 78 mediates the therapeutic efficacy of 17-DMAG in colon cancer cells, Tumor Biol. 36 (6) (2015) 4367–4376. J.G. Kim, et al., HSP90 inhibitor 17-DMAG exerts anticancer effects against gastric cancer cells principally by altering oxidant-antioxidant balance, Oncotarget 8 (34) (2017) 56473. R. Rao, et al., Co-treatment with heat shock protein 90 inhibitor 17-dimethylaminoethylamino-17-demethoxygeldanamycin (DMAG) and vorinostat: a highly active combination against human mantle cell lymphoma (MCL) cells, Cancer Biol. Therapy 8 (13) (2009) 1273–1280. E.E. Bull, et al., Enhanced tumor cell radiosensitivity and abrogation of G2 and S phase arrest by the Hsp90 inhibitor 17-(dimethylaminoethylamino)-17-demethoxygeldanamycin, Clin. Cancer Res. 10 (23) (2004) 8077–8084. J.G. Kiang, J.T. Smith, N.G. Agravante, Geldanamycin analog 17-DMAG inhibits iNOS and caspases in gamma-irradiated human T cells, Radiat. Res. 172 (3) (2009) 321–330. M. Montazeri, et al., Antiproliferative and apoptotic effect of dendrosomal curcumin nanoformulation in P53 mutant and wde-type cancer cell lines, Anti-Cancer Agents Med. Chem. (Former. Curr. Med. Chem.-Anti-Cancer Agents) 17 (5) (2017) 662–673. S. Amirsaadat, et al., Silibinin-loaded magnetic nanoparticles inhibit hTERT gene expression and proliferation of lung cancer cells, Artif. Cells Nanomed., Biotechnol. 45 (8) (2017) 1649–1656. H. Sadeghzadeh, et al., The effects of nanoencapsulated curcumin-Fe3O4 on proliferation and hTERT gene expression in lung cancer cells, Anti-Cancer Agents Med. Chem. (Former. Curr. Med. Chem.-Anti-Cancer Agents) 17 (10) (2017) 1363–1373. R. Farajzadeh, et al., Macrophage repolarization using CD44-targeting hyaluronic acid–polylactide nanoparticles containing curcumin, Artif. Cells Nanomed. Biotechnol. (2017) 1–9. F. Badrzadeh, et al., Comparison between effects of free curcumin and curcumin loaded NIPAAm-MAA nanoparticles on telomerase and PinX1 gene expression in lung cancer cells, Asian Pac. J. Cancer Prev. 15 (20) (2014) 8931–8936. A. Eatemadi, et al., Comparison, synthesis and evaluation of anticancer drugloaded polymeric nanoparticles on breast cancer cell lines, Artif. Cells Nanomed., Biotechnol. 44 (3) (2016) 1008–1017. R. Farajzadeh, et al., Nano-encapsulated metformin-curcumin in PLGA/PEG inhibits synergistically growth and hTERT gene expression in human breast cancer cells, Artif. Cells Nanomed. Biotechnol. (2017) 1–9. Y.H. Alimohammadi, S.W. Joo, PLGA-based nanoparticles as cancer drug delivery systems, Asian Pac. J. Cancer Prev. 15 (2014) 517–535. H. Mellatyar, et al., Comparison of inhibitory effect of 17-DMAG nanoparticles and free 17-DMAG in HSP90 gene expression in lung cancer, Asian Pac. J. Cancer Prev. 15 (20) (2014) 8693–8698. H. Mellatyar, et al., Targeting HSP90 Gene expression with 17-DMAG nanoparticles breast cancer cells, Asian Pac. J. Cancer Prev. 17 (5) (2016) 2453–2457. A. Herzog, et al., Hsp90 and angiogenesis in bone disorders—lessons from the avian growth plate, Am. J. Physiol.-Regul. Integr. Comp. Physiol. 301 (1) (2011) R140–R147. M. Shahzad, et al., Hsp-90 inhibitor geldanamycin attenuates liver oxidative stress and toxicity in thiram-induced tibial dyschondroplasia, Pak. Vet. J. 34 (4) (2014) 545–547. J. Madrigal-Matute, et al., HSP90 inhibition by 17-DMAG attenuates oxidative stress in experimental atherosclerosis, Cardiovasc. Res. 95 (1) (2012) 116–123. S. Tukaj, D. Zillikens, M. Kasperkiewicz, Inhibitory effects of heat shock protein 90 blockade on proinflammatory human Th1 and Th17 cell subpopulations, J. Inflamm. 11 (1) (2014) 10. J.G. Kiang, et al., 17-DMAG diminishes hemorrhage-induced small intestine injury by elevating bcl-2 protein and inhibiting iNOS pathway, TNF-α increase, and caspase-3 activation, Cell Biosci. 1 (1) (2011) 21. S.K. Shimp, et al., Heat shock protein 90 inhibition by 17-DMAG lessens disease in the MRL/lpr mouse model of systemic lupus erythematosus, Cell. Mol. Immunol. 9 (3) (2012) 255–266. S.K. Shimp, et al., HSP90 inhibition by 17-DMAG reduces inflammation in J774 macrophages through suppression of Akt And nuclear factor-κB pathways, Inflamm. Res. 61 (5) (2012) 521–533. Y.-L. Wang, et al., 17-DMAG, an HSP90 inhibitor, ameliorates multiple organ dysfunction syndrome via induction of HSP70 in endotoxemic rats, PloS One 11 (5) (2016) e0155583. A. Rao, et al., Combination therapy with HSP90 inhibitor 17-DMAG reconditions the tumor microenvironment to improve recruitment of therapeutic T cells, Cancer Res. 72 (13) (2012) 3196–3206. J.L. Eiseman, et al., Pharmacokinetics and pharmacodynamics of 17-demethoxy 17-[[(2-dimethylamino) ethyl] amino] geldanamycin (17DMAG, NSC 707545) in CB-17 SCID mice bearing MDA-MB-231 human breast cancer xenografts, Cancer Chemother. Pharm. 55 (1) (2005) 21–32. E.R. Glaze, et al., Preclinical toxicity of a geldanamycin analog, 17-(dimethylaminoethylamino)-17-demethoxygeldanamycin (17-DMAG), in rats and dogs: potential clinical relevance, Cancer Chemother. Pharm. 56 (6) (2005) 637–647. M.A. Smith, et al., Stage 1 testing and pharmacodynamic evaluation of the HSP90

[102]

[103]

[104]

[105]

[106]

[107]

[108]

[109] [110]

[111] [112] [113]

[114]

[115]

[116]

[117]

[118] [119]

[120]

[121]

[122]

[123] [124]

[125] [126]

[127] [128]

[129]

[130]

616

inhibitor alvespimycin (17‐DMAG, KOS‐1022) by the pediatric preclinical testing program, Pediatr. Blood Cancer 51 (1) (2008) 34–41. R.K. Ramanathan, et al., Phase I pharmacokinetic and pharmacodynamic study of 17-dimethylaminoethylamino-17-demethoxygeldanamycin, an inhibitor of heatshock protein 90, in patients with advanced solid tumors, J. Clin. Oncol. 28 (9) (2010) 1520–1526. J. Lancet, et al., Phase I study of the heat shock protein 90 inhibitor alvespimycin (KOS-1022, 17-DMAG) administered intravenously twice weekly to patients with acute myeloid leukemia, Leukemia 24 (4) (2010) 699–705. S. Kummar, et al., Phase I trial of 17-dimethylaminoethylamino-17-demethoxygeldanamycin (17-DMAG), a heat shock protein inhibitor, administered twice weekly in patients with advanced malignancies, Eur. J. Cancer 46 (2) (2010) 340–347. S. Pacey, 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. 17 (6) (2011) 1561–1570 p. clincanres. 1927. 2010. K. Jhaveri, et al., A phase I dose-escalation trial of trastuzumab And alvespimycin hydrochloride (KOS-1022; 17 DMAG) in the treatment of advanced solid tumors, Clin. Cancer Res. 18 (18) (2012) 5090–5098. K. Maddocks, et al., A phase I trial of the intravenous Hsp90 inhibitor alvespimycin (17-DMAG) in patients with relapsed chronic lymphocytic leukemia/small lymphocytic lymphoma, Leukemia Lymphoma 57 (9) (2016) 2212–2215. D.B. Solit, et al., 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. 8 (5) (2002) 986–993. Y. Fang, et al., Hsp90 regulates androgen receptor hormone binding affinity in vivo, J. Biol. Chem. 271 (45) (1996) 28697–28702. J. Beliakoff, et al., Hormone-refractory breast cancer remains sensitive to the antitumor activity of heat shock protein 90 inhibitors, Clin. Cancer Res. 9 (13) (2003) 4961–4971. A.E. Fliss, et al., Control of estrogen receptor ligand binding by Hsp90, J. Steroid Biochem. Mol. Biol. 72 (5) (2000) 223–230. D.F. Smith, et al., Progesterone receptor structure and function altered by geldanamycin, an hsp90-binding agent, Mol. Cell. Biol. 15 (12) (1995) 6804–6812. P.N. Münster, et al., Degradation of HER2 by ansamycins induces growth arrest and apoptosis in cells with HER2 overexpression via a HER3, phosphatidylinositol 3′-kinase-AKT-dependent pathway, Cancer Res. 62 (11) (2002) 3132–3137. W. Xu, et al., Sensitivity of mature Erbb2 to geldanamycin is conferred by its kinase domain and is mediated by the chaperone protein Hsp90, J. Biol. Chem. 276 (5) (2001) 3702–3708. T. Shimamura, et al., 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. 65 (14) (2005) 6401–6408. T.W. Schulte, et al., Destabilization of raf-1 by geldanamycin leads to disruption of the raf-1-MEK-mitogen-activated protein kinase signalling pathway, Mol. Cell. Biol. 16 (10) (1996) 5839–5845. T.W. Schulte, et al., Disruption of the raf-1-Hsp90 molecular complex results in destabilization of raf-1 and loss of raf-1-ras association, J. Biol. Chem. 270 (41) (1995) 24585–24588. S.J. Arlander, et al., Hsp90 inhibition depletes Chk1 and sensitizes tumor cells to replication stress, J. Biol. Chem. 278 (52) (2003) 52572–52577. L. Sepp-Lorenzino, et al., Herbimycin A induces the 20 S proteasome-and ubiquitindependent degradation of receptor tyrosine kinases, J. Biol. Chem. 270 (28) (1995) 16580–16587. H.-J. Ochel, et al., The benzoquinone ansamycin geldanamycin stimulates proteolytic degradation of focal adhesion kinase, Mol. Genet. Metab. 66 (1) (1999) 24–30. F.S. Goes, J. Martin, Hsp90 chaperone complexes are required for the activity and stability of yeast protein kinases Mik1, Wee1 and Swe1, FEBS J. 268 (8) (2001) 2281–2289. N.T. Archie, T.N. Sheikh, G.K. Schwartz, The Hsp90 Inhibitor, 17-Allylamino-17Demethoxygeldanamycin (17AAG) Abrogates the G2/M DNA Damage Checkpoint and Induces Apoptosis Selectively in p53-defective Colon Cancer Cells by Downregulating Both Chk1 and Wee1, AACR, 2005. G. de Cárcer, et al., Requirement of Hsp90 for centrosomal function reflects its regulation of polo kinase stability, EMBO J. 20 (11) (2001) 2878–2884. J.E. Castro, 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 106 (7) (2005) 2506–2512. S. Sato, N. Fujita, T. Tsuruo, Modulation of Akt kinase activity by binding to Hsp90, Proc. Natl. Acad. Sci. 97 (20) (2000) 10832–10837. A.D. Basso, et al., Akt forms an intracellular complex with heat shock protein 90 (Hsp90) and Cdc37 and is destabilized by inhibitors of Hsp90 function, J. Biol. Chem. 277 (42) (2002) 39858–39866. L. Stepanova, et al., Mammalian p50Cdc is a protein kinase-targeting subunit of Hsp90 that binds and stabilizes Cdk4, Gene Dev. 10 (12) (1996) 1491–1502. R. Nimmanapalli, E. O’Bryan, K. Bhalla, Geldanamycin And its analogue 17-allylamino-17-demethoxygeldanamycin lowers Bcr-abl levels and induces apoptosis and differentiation of Bcr-Abl-positive human leukemic blasts, Cancer Res. 61 (5) (2001) 1799–1804. M.E. Gorre, et al., BCR-ABL point mutants isolated from patients with imatinib mesylate–resistant chronic myeloid leukemia remain sensitive to inhibitors of the BCR-ABL chaperone heat shock protein 90, Blood 100 (8) (2002) 3041–3044. C.P. Webb, et al., The geldanamycins are potent inhibitors of the hepatocyte growth factor/scatter factor-met-urokinase plasminogen activator-plasmin proteolytic network, Cancer Res. 60 (2) (2000) 342–349.

Biomedicine & Pharmacotherapy 102 (2018) 608–617

H. Mellatyar et al.

[135] N.J. Mabjeesh, et al., Geldanamycin induces degradation of hypoxia-inducible factor 1α protein via the proteosome pathway in prostate cancer cells, Cancer Res. 62 (9) (2002) 2478–2482. [136] H.L. Forsythe, et al., Stable association of hsp90 and p23, but not hsp70, with active human telomerase, J. Biol. Chem. 276 (19) (2001) 15571–15574. [137] R. Villa, et al., Inhibition of telomerase activity by geldanamycin and 17-allylamino, 17-demethoxygeldanamycin in human melanoma cells, Carcinogenesis 24 (5) (2003) 851–859. [138] P. Fortugno, et al., Regulation of survivin function by Hsp90, Proc. Natl. Acad. Sci. 100 (24) (2003) 13791–13796.

[131] Q. Yao, et al., FLT3 expressing leukemias are selectively sensitive to inhibitors of the molecular chaperone heat shock protein 90 through destabilization of signal transduction-associated kinases, Clin. Cancer Res. 9 (12) (2003) 4483–4493. [132] L. Whitesell, et al., The physical association of multiple molecular chaperone proteins with mutant p53 is altered by geldanamycin, an hsp90-binding agent, Mol. Cell. Biol. 18 (3) (1998) 1517–1524. [133] E. Minet, et al., Hypoxia‐induced activation of HIF‐1: role of HIF‐1α‐Hsp90 interaction, FEBS Lett. 460 (2) (1999) 251–256. [134] J.S. Isaacs, et al., Hsp90 regulates a von hippel lindau-independent hypoxia-inducible factor-1α-degradative pathway, J. Biol. Chem. 277 (33) (2002) 29936–29944.

617