Combinatorial drug design targeting multiple cancer signaling networks controlled by mitochondrial Hsp90 (original) (raw)

Selective targeting of the mitochondrial Hsp90 network. We began this study by designing what we believe to be a new class of small molecule Hsp90 antagonists selectively targeted to mitochondria, i.e., Gamitrinibs. The chemical synthesis of Gamitrinib is described in detail in the Supplemental Data and Supplemental Figure 1 (supplemental material available online with this article; doi:10.1172/JCI37613DS1). The structure of Gamitrinib is combinatorial (Figure 1A) and contains a benzoquinone ansamycin backbone derived from the Hsp90 inhibitor 17-(allylamino)-17-demethoxy­geldanamycin (17-AAG) (14), a linker region on the C17 position, and a mitochondrial targeting moiety, either provided by 1 to 4 tandem repeats of cyclic guanidinium (27) (Gamitrinib-G1–G4) or triphenylphosphonium (28) (Gamitrinib–TPP-OH) (Figure 1A). By molecular dynamics simulation, the 17-AAG portion of Gamitrinib is predicted to make contacts with the Hsp90 ATPase pocket, whereas the mitochondriotropic guanidinium module is excluded from the binding interface, pointing outside of the ATPase pocket toward the solvent (Figure 1B, top panel). In the predicted docking structure, the binding arrangement of Gamitrinibs to Hsp90 closely follows that of GA (29), with root mean square deviation of heavy atoms of the 17-AAG region being 0.5 υ (Figure 1B, bottom panel).

Structure of Gamitrinibs.Figure 1

Structure of Gamitrinibs. (A) Combinatorial modular design. The indicated individual modules of Gamitrinibs (mitochondrial targeting, linker, and Hsp90 inhibition) are indicated. G-G1, Gamitrinib-G1; G-TPP, Gamitrinib–TPP-OH; TBDPS, tert-butyldiphenylsilyl. (B) A 3D docking model of Gamitrinib-G1 with Hsp90 N-domain (top panel). The side chains of contact sites are labeled and in color. An overlay of Gamitrinib-G1 (red) and GA (gray) in the ATPase pocket of Hsp90 (bottom panel).

We next carried out 2 experiments to test whether the addition of a mitochondriotropic moiety reduced the ability of the 17-AAG backbone to bind Hsp90 and block its chaperone function. In previous studies, modifications of the C17 position of GA or 17-AAG did not affect inhibition of the chaperone ATPase cycles. Consistent with this, increasing concentrations of GA effectively competed with GA affinity beads for binding to Hsp90 in a tumor cell lysate (Figure 2A, left panel). Similarly, Gamitrinib-G4 displaced Hsp90 from GA affinity beads, albeit less efficiently than GA in this assay (Figure 2A, right panel). Conversely, increasing concentrations of both unconjugated 17-AAG and Gamitrinib-G4 capably inhibited Hsp90 chaperone activity (Figure 2B), using a purified client protein assay, i.e., Chk1 reconstitution assay (30). We next asked whether the combinatorial design of Gamitrinibs allowed their accumulation in isolated tumor mitochondria. For these experiments, we used a spectrophotometric approach coupled to gradient density ultracentrifugation of cellular extracts for isolation of purified mitochondria (Figure 2C). In preliminary studies, we found that at a drug concentration of 0.2 M, unconjugated 17-AAG exhibited 2 peaks of absorption at 338 nm (absorption, 2.448) and 533 nm (absorption, 0.182), whereas peak absorption for Gamitrinib-G4 was at 338 nm (absorption, 2.124) and 543 nm (absorption, 0.138), respectively. Thus, an absorbance at 338 nm was chosen for all subsequent detection studies of Gamitrinibs. Using these experimental conditions, Gamitrinib-G4 was readily detected at a 1–1.5 M interface, corresponding to isolated mitochondria after gradient density ultracentrifugation (Figure 2, C and D). In contrast, 17-AAG did not localize to the mitochondrial interface as compared with untreated samples (Figure 2, C and D) in agreement with previous observations (22). Using a similar approach, all Gamitrinibs accumulated comparably in isolated tumor mitochondria, regardless of the structure or composition of their mitochondriotropic moieties (Figure 2E).

Inhibition of Hsp90 chaperone activity.Figure 2

Inhibition of Hsp90 chaperone activity. (A) Competition with GA affinity beads. GA (left panel) or Gamitrinib-G4 (right panel), at the indicated concentrations, were incubated with aliquots of SKBr3 tumor cell lysates followed by affinity purification of Hsp90 using GA affinity beads. Data are densitometric quantifications of scanning and image analysis of Hsp90 bands visualized by Western blotting. (B) Inhibition of Chk1 kinase activity. 17-AAG or Gamitrinib-G4 (1–10 μM) were analyzed for modulation of Chk1-dependent phosphorylation of Cdc25. Densitometric quantification of protein bands (right panel). (A and B) Data are representative of 2 independent experiments with identical results. (C) Accumulation of Gamitrinib in mitochondria. Cellular extracts were loaded with vehicle (None), unconjugated 17-AAG, or Gamitrinib-G4 and analyzed before (left panel) or after gradient density ultracentrifugation (right panel). The position of the 1–1.5 M interface corresponding to isolated mitochondria is indicated. (D) Quantification of mitochondrial accumulation. Mitochondria isolated from HeLa cells were incubated with 17-AAG, Gamitrinib-G4, or vehicle and analyzed using absorbance. Data are the mean ± SEM (n = 3). (E) Mitochondriotropic properties of all Gamitrinibs. Mitochondria isolated from Raji cells were incubated with the indicated Gamitrinibs or vehicle and analyzed by absorbance. Data are from a representative experiment out of 2 independent determinations.

Gamitrinibs induce sudden mitochondrial permeability transition. When added to isolated tumor mitochondria, Gamitrinibs caused immediate loss of inner mitochondrial membrane potential, all with comparable efficiency, regardless of their different mitochondriotropic moieties (Figure 3A). In contrast, unconjugated Hsp90 inhibitors, including GA, 17-AAG, or 17-(dimethylaminoethylamino)-17-demethoxygeldanamycin (DMAG), did not affect mitochondrial membrane potential (Figure 3A). Next, we asked whether this response was specific for inhibition of Hsp90 chaperone activity inside mitochondria. Consistent with the data presented above, Gamitrinibs (Gamitrinib-G4 or –TPP-OH) suddenly depolarized tumor mitochondria, and this reaction was partially reversed by cyclosporine A (CsA), an inhibitor of the immunophilin CypD (Figure 3B). In contrast, treatment of tumor mitochondria with the mitochondriotropic moieties of Gamitrinbs, tetraguanidinium (TG-OH) or TPP-OH plus unconjugated 17-AAG or GA had no effect on mitochondrial membrane potential, with or without CsA (Figure 3B).

Mitochondrial dysfunction.Figure 3

Mitochondrial dysfunction. (A) Mitochondrial membrane depolarization. Tetramethylrhodamine methyl ester–loaded (TMRM-loaded) HeLa cell mitochondria treated with Gamitrinibs or the various indicated agents (1 μM) were analyzed for changes in fluorescence emission. (B) Analysis of individual mitochondriotropic moieties. TMRM-loaded HeLa cell mitochondria were incubated with 1.5 μM 17-AAG plus tetraguanidinium (TG-OH) or Gamitrinib-G4 (left panel) or 0.7 μM GA plus TPP-OH or Gamitrinib–TPP-OH (right panel) and analyzed for changes in fluorescence emission in the presence or absence of CsA (5 μM). Arrows indicate point of addition. (C) Cytochrome c (Cyto c) release. Mitochondria isolated from HeLa cells treated with Gamitrinibs or 17-AAG (20 minutes) were analyzed for cytochrome c release in supernatants (S) or pellets (P). Cox-IV or Ran was used as a mitochondrial or cytosolic marker, respectively. Reactivity of the antibody to Ran with isolated cytosolic extracts (C) was used as a control. (D) Mitochondrial accumulation. Isolated HeLa cell mitochondria were incubated with vehicle or Gamitrinib-G4 in the presence or absence of CsA and analyzed using absorbance. Data are the mean ± SEM. (E) Analysis of nontargeted Hsp90 inhibitors. Isolated HeLa cell mitochondria were incubated with increasing concentrations of the various indicated agents for 3 hours and analyzed for cytochrome c release. Data are representative of 2 independent experiments.

Loss of mitochondrial membrane potential followed by rupture of the outer membrane and release of cytochrome c are hallmarks of mitochondrial permeability transition (21) and the molecular prerequisites for the initiation of the intrinsic pathway of apoptosis (21). To further test this model, we examined the ability of Gamitrinibs to induce cytochrome c release from isolated tumor mitochondria. In these experiments, all Gamitrinibs induced rapid (20 minute) discharge of mitochondrial cytochrome c in the supernatant, whereas unconjugated 17-AAG was ineffective (Figure 3C). Because Gamitrinib-induced loss of mitochondrial membrane potential was sensitive to CsA (Figure 3B), we next asked whether pharmacologic inhibition of CypD prevented the accumulation of the compound(s) in mitochondria. Gamitrinib-G4 readily accumulated in isolated tumor mitochondria, within the same time interval of induction of organelle permeability transition (Figure 3D), in agreement with the data presented above (Figure 2, C–E). In these experiments, preincubation of isolated mitochondria with CsA, under the same conditions used to antagonize permeability transition (Figure 3B), did not reduce Gamitrinib accumulation in mitochondria (Figure 3D).

Additional derivatives of 17-AAG as well as purine- and isoxazole resorcinol–based Hsp90 antagonists (Supplemental Figure 2) have been recently developed, with promising anticancer activity in preclinical studies (25). Therefore, it was of interest to test whether these GA-modified or non–GA-based Hsp90 antagonists affected mitochondrial integrity. Gamitrinib-G4 induced sudden and complete discharge of cytochrome c from tumor mitochondria (Figure 3E), in agreement with the data above. In contrast, increasing concentrations of 17-AAG, hydroquinone derivative of 17-AAG (IPI-504), purine analog (BIIB021), or isoxazole (NVP-AUY922) Hsp90 inhibitors (25) had no effect on cytochrome c release (Figure 3E).

Induction of mitochondrial apoptosis by Gamitrinibs. A 3-hour exposure of lung adenocarcinoma H460 cells to Gamitrinib-G3 or -G4 was sufficient to produce concentration dependent (IC50 ~0.5 μM) and complete loss of cell viability (Figure 4A, left panel) in the tumor cell population. Within this time frame, Gamitrinib-G1 or 17-AAG had no effect and Gamitrinib-G2 or Gamitrinib–TPP-OH had intermediate activity (Figure 4A, left panel). By 24 hours, all Gamitrinibs had comparably killed the entire tumor cell population, whereas 17-AAG had only partially reduced cell viability or cell proliferation (Figure 4A, right panel). As Gamitrinibs all comparably induce membrane depolarization of isolated mitochondria (Figure 3A), these data suggest that the Gamitrinib-G4 and -G3 moieties provide more efficient intracellular drug uptake, as compared with Gamitrinib-G1 and -G2 structures in vivo.

Gamitrinib-mediated anticancer activity.Figure 4

Gamitrinib-mediated anticancer activity. (A) Time course. H460 cells treated with the indicated concentrations of Gamitrinibs or 17-AAG were analyzed by a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay after 3 hours (left panel) or 24 hours (right panel). Data are the mean ± SD (n = 2). (B) Caspase-associated cell death. H460 cells, treated with Gamitrinib-G4 or vehicle for 4 hours, were labeled with JC-1 and analyzed for loss of mitochondrial membrane potential by changes in FL2/FL1 fluorescence ratio (top panel) or Asp-Glu-Val-Asp-ase (DEVDase) (caspase) activity (bottom panel) using multiparametric flow cytometry. The percentage of cells in each quadrant is indicated. PI, propidium iodide. (C) Insensitivity to Bax. Wild-type or Bax–/– HCT116 cells were incubated with the indicated increasing concentrations of Gamitrinib-G4 and analyzed after 6 hours using MTT assay. Data are representative of 2 experiments.

We next studied the cell death phenotype induced by Gamitrinibs. Treatment of H460 cells with Gamitrinib-G4 resulted in rapid loss of mitochondrial membrane potential in the whole tumor cell population, pronounced effector caspase activity, and cell death consistent with genuine mitochondrial apoptosis (Figure 4B). In contrast, vehicle alone had no effect on mitochondrial integrity or cell viability (Figure 4B). Proapoptotic Bcl-2 family proteins regulate mitochondrial cell death, in particular permeabilization of the outer membrane (21). Therefore, we next asked whether Gamitrinib-induced tumor cell killing was dependent on Bax, a multidomain proapoptotic Bcl-2 protein required for many cell death responses (31). In these experiments, Gamitrinib-G4 comparably induced concentration-dependent killing of wild-type or Bax–/– colorectal cancer HCT116 cells (Figure 4C). Consistent with these data, Gamitrinib-G4 exhibited a broad spectrum of anticancer activity, and comparably killed a molecularly and genetically heterogeneous panel of tumor cell types, representative of epithelial and hematologic malignancies (Table 1). In contrast, exposure of these cells to the mitochondriotropic moiety (TG-OH) plus unconjugated 17-AAG had no effect on cell viability (Table 1).

Table 1

Broad spectrum of Gamitrinib-G4 anticancer activity

Mitochondriotoxic mechanism of action of Gamitrinibs. Current Hsp90 inhibitors predominantly cause cell cycle arrest in most tumor cell types, followed by a variable degree of apoptosis after a prolonged drug exposure of 48–72 hours. To test whether Gamitrinibs had a similar mechanism of action, we used breast adenocarcinoma SKBr3 cells, which are considered sensitive to Hsp90 inhibition. Gamitrinib-G4, Gamitrinib–TPP-OH, or unconjugated 17-AAG all resulted in comparable reduction of metabolic activity in SKBr3 cells by 48 hours and throughout a 96-hour interval after treatment (Figure 5A). However, most SKBr3 cells treated with 17-AAG were still alive after 72 hours, as shown using Trypan blue exclusion and light microscopy (Figure 5B). In contrast, Gamitrinibs were cytotoxic, and a 24-hour treatment was sufficient to cause nearly complete killing of the entire tumor cell population (Figure 5B). When analyzed in a model of long-term tumorigenicity, a brief (4-hour) exposure of H460 cells to Gamitrinib-G4 completely abolished colony formation in soft agar after a 2-week incubation (Figure 5C). In contrast, 17-AAG had no effect on tumorigenicity under the same conditions, and vehicle alone did not decrease H460 colony formation in soft agar (Figure 5C).

Gamitrinibs induction of mitochondrial apoptosis.Figure 5

Gamitrinibs induction of mitochondrial apoptosis. (A) Comparison with 17-AAG. SKBr3 cells were treated with Gamitrinibs or 17-AAG (10 μM) for the indicated time intervals and analyzed using MTT assay. Data are representative of at least 2 independent experiments. (B) Tumor cell killing. SKBr3 cells treated with vehicle, Gamitrinib-G4, Gamitrinib–TPP-OH, or 17-AAG (10 μM), for the indicated time intervals, were analyzed by Trypan blue exclusion. Data are the mean ± SEM (n = 3). (C) Colony formation. H460 cells treated with vehicle, 17-AAG (50 μM), or Gamitrinib-G4 (50 μM) for 4 hours were analyzed for colony formation in soft agar after 2 weeks. Representative microscopy fields are shown. Original magnification, ×40. (D) Client protein modulation. HeLa cells treated with the indicated Gamitrinibs, 17-AAG (5 μM), or vehicle were analyzed for modulation of Hsp90 client proteins Akt and Chk1 in the cytosol or changes in expression of Hsp70 after 24 hours by Western blotting. (E) Requirement for CypD in Gamitrinib anticancer activity. H460 cells transfected with control (closed symbols) or CypD (open symbols) siRNA were treated with 17-AAG (circles) or Gamitrinib-G4 (squares) and analyzed using MTT assay after 6 hours. Data are the mean ± SEM (n = 3). The inset shows Western blotting of CypD knockdown by siRNA.

A hallmark of Hsp90 inhibition by conventional small molecule antagonists is the degradation of client proteins in the cytosol, with concomitant upregulation of Hsp70 chaperone levels. Consistent with this, treatment of cervical carcinoma HeLa cells with 17-AAG resulted in efficient loss of Hsp90 client proteins, Akt and Chk1 (14), and increased expression of Hsp70 (32), as analyzed by Western blotting (Figure 5D). At variance with this phenotype, incubation of HeLa cells with Gamitrinib-G4 had no effect on the levels of Hsp90 client proteins in the cytosol and did not result in upregulation of Hsp70 levels (Figure 5D).

Previous studies have shown that mitochondrial Hsp90 chaperones physically associate with the permeability pore component, CypD (21), and prevent its opening by a protein folding mechanism that antagonizes the initiation of cell death. Consistent with this model, siRNA silencing of CypD in H460 cells (Figure 5E, inset) markedly attenuated Gamitrinib-G4–induced tumor cell killing (Figure 5E). In contrast, a nontargeted siRNA was without effect (Figure 5E, inset) and 17-AAG did not reduce H460 cell viability within the same time interval in the presence or absence of CypD (Figure 5E). Collectively, these data demonstrate that Gamitrinibs exhibit what we believe to be a unique, mitochondriotoxic mechanism of action, which involves CypD induction of organelle permeability transition, without affecting global Hsp90 homeostasis outside of mitochondria.

Selectivity of Gamitrinib anticancer activity. Because mitochondria of most normal tissues do not contain Hsp90 chaperones, it was of interest to test whether Gamitrinibs were selective anticancer agents. Exposure of normal mitochondria isolated from WS-1 human fibroblasts to Gamitrinib-G4 or 17-AAG induced only a modest decrease in mitochondrial membrane potential in the presence or absence of CsA (Figure 6A). Similarly, Gamitrinibs did not induce discharge of cytochrome c from normal mitochondria (Figure 6B). As control, mitochondria isolated from HeLa cells were nearly completely depleted of cytochrome c after treatment with Gamitrinib-G1 but not 17-AAG (Figure 6B). Validating the specificity of these results, Gamitrinib-G4 readily accumulated in isolated normal mouse liver mitochondria, and this response was also unaffected by CypD inhibition with CsA (Figure 6C). Similar to the phenotype observed with tumor mitochondria, nontargeted 17-AAG did not accumulate in normal mitochondria (Figure 6C).

Selectivity of Gamitrinib anticancer activity.Figure 6

Selectivity of Gamitrinib anticancer activity. (A) Mitochondrial membrane potential. TMRM-loaded mitochondria isolated from WS-1 normal human fibroblasts were incubated with Gamitrinib-G4 or 17-AAG plus the uncoupled mitochondriotropic moiety TG-OH and analyzed for changes in inner membrane potential in the presence or absence of CsA. (B) Cytochrome c release. Mitochondria isolated from normal HFF fibroblasts were treated with Gamitrinibs or 17-AAG and analyzed by Western blotting. HeLa cells were used as control. Cox-IV or Ran was used as a mitochondrial or cytosolic marker, respectively. Reactivity of the antibody to Ran with isolated cytosolic extracts from HeLa or HFF cells was used as a control. (C) Mitochondrial accumulation. Isolated normal mouse liver mitochondria were incubated with vehicle, 17-AAG, or Gamitrinib-G4 in the presence or absence of CsA and analyzed using absorbance. Data are the mean ± SEM. (D) Analysis of cell viability. Human fibroblasts (HFF, black line), bovine aortic endothelial cells (brown line), intestinal epithelial cells (red line), or human umbilical vein endothelial cells (green line) were treated with Gamitrinib-G4 (solid lines) or 17-AAG (dashed lines) and analyzed using MTT assay after 24 hours. Data are representative of 2 experiments.

Consistent with this inability to induce mitochondrial permeability transition in normal cells, concentrations of Gamitrinib that readily killed tumor cell types did not cause cell death of normal primary cell types, including bovine aortic endothelial cells or intestinal epithelial cells, and only modestly reduced the viability of normal human foreskin fibroblasts or human umbilical vein endothelial cells (Figure 6D). Similarly, comparable concentrations of 17-AAG had no effect on the viability of the various normal human cells tested (Figure 6D).

Gamitrinib inhibition of tumor growth in vivo. Systemic administration of Gamitrinib-G4 to mice inhibited the growth of established human leukemia (Supplemental Figure 3A), breast (Supplemental Figure 3B), and lung (Figure 7A) xenograft tumors in vivo. In contrast, comparable concentrations of 17-AAG had no effect on human lung cancer growth in mice (Figure 7A, top panel). Gamitrinibs carrying different mitochondriotropic moieties, i.e., Gamitrinib-G1 or –TPP-OH, also comparably inhibited lung cancer growth in vivo (Figure 7A, bottom panel). Tumors harvested from Gamitrinib-treated animals exhibited extensive apoptosis in situ (Figure 7B) and discharge of cytochrome c in the cytosol (Figure 7C), suggestive of treatment-related mitochondrial dysfunction in vivo. At the concentrations used, the various Gamitrinibs did not cause significant animal weight loss throughout the treatment (Figure 7D). For preliminary toxicology experiments, organs, including brain, small and large intestine, heart, spleen, liver, pancreas, stomach, lung and kidneys, were collected from mice in the various treatment groups and analyzed histologically. In these studies, organs harvested from Gamitrinib-treated animals were histologically unremarkable compared with the vehicle group, with no changes in general architecture and no evidence of inflammation or liver steatosis (Supplemental Figure 4).

Gamitrinibs anticancer activity in vivo.Figure 7

Gamitrinibs anticancer activity in vivo. (A) Kinetics of xenograft tumor growth. SCID/beige mice carrying H460 lung adenocarcinoma xenograft tumors (100–150 mm3) were treated with Gamitrinib-G4 or 17-AAG (top panel), with a dose escalation regimen as described in Supplemental Data, or with Gamitrinib-G1 or Gamitrinib–TPP-OH (bottom panel). Tumor volume was measured with a caliper. (B) Induction of apoptosis in vivo. Tumor specimens from vehicle- or Gamitrinib-treated tumors were analyzed for internucleosomal DNA fragmentation in situ (using TUNEL staining), and positive cells were quantified (bottom panel). Original magnification, ×400. ***P < 0.0001. Data are the mean ± SEM (A and B). (C) Mitochondrial dysfunction in vivo. Cytosolic fractions from H460 xenograft tumors harvested from vehicle- or Gamitrinib-G4–treated animals were analyzed by Western blotting. Two mice per group were tested and the animal number is shown. (D) Animal weight. Mice treated with the various Gamitrinibs, 17-AAG, or vehicle were analyzed for percentage weight change at the end of the experiment. Data are the mean ± SEM.