10 research outputs found

    The HSP90 Inhibitor Ganetespib Alleviates Disease Progression and Augments Intermittent Cyclophosphamide Therapy in the MRL/lpr Mouse Model of Systemic Lupus Erythematosus

    No full text
    <div><p>Systemic lupus erythematosus (SLE) is a complex, systemic autoimmune disease with a diverse range of immunological and clinical manifestations. The introduction of broad spectrum immunosuppressive therapies and better management of acute disease exacerbations have improved outcomes for lupus patients over recent years. However, these regimens are burdened by substantial toxicities and confer significantly higher risks of infection, thus there remains a significant and unmet medical need for alternative treatment options, particularly those with improved safety profiles. Heat shock protein 90 (HSP90) is a ubiquitously expressed molecular chaperone that acts as an important modulator of multiple innate and adaptive inflammatory processes. Of note, accumulating clinical and experimental evidence has implicated a role for HSP90 in the pathogenesis of SLE. Here we evaluated the potential of HSP90 as a therapeutic target for this disease using the selective small molecule inhibitor ganetespib in the well-characterized MRL/lpr autoimmune mouse model. In both the prophylactic and therapeutic dosing settings, ganetespib treatment promoted dramatic symptomatic improvements in multiple disease parameters, including suppression of autoantibody production and the preservation of renal tissue integrity and function. In addition, ganetespib exerted profound inhibitory effects on disease-related lymphadenopathy and splenomegaly, and reduced pathogenic T and B cell lineage populations in the spleen. Ganetespib monotherapy was found to be equally efficacious and tolerable when compared to an effective weekly dosing regimen of the standard-of-care immunosuppressive agent cyclophosphamide. Importantly, co-treatment of ganetespib with a sub-optimal, intermittent dosing schedule of cyclophosphamide resulted in superior therapeutic indices and maximal disease control. These findings highlight the potential of HSP90 inhibition as an alternative, and potentially complementary, strategy for therapeutic intervention in SLE. Such approaches may have important implications for disease management, particularly for limiting or preventing treatment-related toxicities, a major confounding factor in current SLE therapy.</p></div

    Ganetespib prevents the development of renal damage in a pilot prophylactic dosing study.

    No full text
    <p>(A) Mean pharmacokinetic plasma concentration-time profile after i.v. administration of ganetespib to MRL/lpr mice. Ganetespib was dosed at 50 mg/kg twice weekly and samples were taken after the second dose. Data represent mean ± SD (n = 3). (B) Average proteinuria scores (±SE) are plotted for vehicle and ganetespib-treated mice over the time course of the study. Average proteinuria scores for non-lupus parental strain (MRL/mp) mice from 8–15 weeks of age are included for comparison. (C) HSP70 induction as a pharmacodynamic marker of ganetespib activity in kidneys from drug-treated animals. Kidneys were harvested at 6 and 24 hours following the second intra-weekly dose of ganetespib (or vehicle) and lysates immunoblotted for HSP70 expression. GAPDH included as a loading control. Each lane represents renal tissue from an individual animal. (D) Representative H&E stained renal histopathology showing extensive chronic inflammation and thickening of the basement membrane with crescent formation in the glomerulus of a vehicle-treated mouse (<i>left panel</i>). In contrast, the kidneys of a ganetespib-treated animal showed no evident pathologic changes (<i>right panel</i>). Original magnification, 40X; scale bar represents 50 μm. (E) To evaluate total lupus-related renal damage 7 primary characteristics of GN (glomerular capillary deposits, hypercellularity, necrosis, tubular atrophy, intratubular casts, interstitial chronic inflammation, and fibrosis) were scored for each animal and summed for a composite disease score. The average summed pathology scores for each treatment group are plotted. (E) Representative IgG immunohistochemical staining of renal tissues harvested from vehicle- and ganetespib-treated animals at 22 weeks of age. Red arrows highlight the marked reduction in glomerular IgG deposition seen following HSP90 inhibitor treatment. Original magnification 20X: scale bar represents 100 μm. (F) Quantification of IgG glomerular immunoreactivity observed in vehicle and drug treated animals.</p

    Absence of lymphoproliferation in combination-treated MRL/lpr mice.

    No full text
    <p>(A) Lymph nodes were harvested from mice upon completion of the individual therapeutic dosing regimens. Data are expressed as median pooled lymph node weights for each treatment (± SD). *p<0.05. <i>Gpib</i>, <i>ganetespib; Comb</i>, <i>combination</i>. (B) Representative images of regional lymph nodes harvested from vehicle- and combination (ganetespib + CTX/2)-treated animals at the end of the study. (C) Spleens were harvested at necropsy and weighed. Data are expressed as median spleen weights per treatment group (± SD).*p<0.05. (D) Representative images of spleens taken from mice from all treatment groups.</p

    Ganetespib prevents the development of renal damage in a pilot prophylactic dosing study.

    No full text
    <p>(A) Mean pharmacokinetic plasma concentration-time profile after i.v. administration of ganetespib to MRL/lpr mice. Ganetespib was dosed at 50 mg/kg twice weekly and samples were taken after the second dose. Data represent mean ± SD (n = 3). (B) Average proteinuria scores (±SE) are plotted for vehicle and ganetespib-treated mice over the time course of the study. Average proteinuria scores for non-lupus parental strain (MRL/mp) mice from 8–15 weeks of age are included for comparison. (C) HSP70 induction as a pharmacodynamic marker of ganetespib activity in kidneys from drug-treated animals. Kidneys were harvested at 6 and 24 hours following the second intra-weekly dose of ganetespib (or vehicle) and lysates immunoblotted for HSP70 expression. GAPDH included as a loading control. Each lane represents renal tissue from an individual animal. (D) Representative H&E stained renal histopathology showing extensive chronic inflammation and thickening of the basement membrane with crescent formation in the glomerulus of a vehicle-treated mouse (<i>left panel</i>). In contrast, the kidneys of a ganetespib-treated animal showed no evident pathologic changes (<i>right panel</i>). Original magnification, 40X; scale bar represents 50 μm. (E) To evaluate total lupus-related renal damage 7 primary characteristics of GN (glomerular capillary deposits, hypercellularity, necrosis, tubular atrophy, intratubular casts, interstitial chronic inflammation, and fibrosis) were scored for each animal and summed for a composite disease score. The average summed pathology scores for each treatment group are plotted. (E) Representative IgG immunohistochemical staining of renal tissues harvested from vehicle- and ganetespib-treated animals at 22 weeks of age. Red arrows highlight the marked reduction in glomerular IgG deposition seen following HSP90 inhibitor treatment. Original magnification 20X: scale bar represents 100 μm. (F) Quantification of IgG glomerular immunoreactivity observed in vehicle and drug treated animals.</p

    Ganetespib-based therapies suppress splenic expansion of activated T cells and abnormal plasma cells.

    No full text
    <p>(A) Plot shows the total number of splenocytes. Data are shown for 7–12 mice per treatment group. (*p<0.007; **p<0.0037; ***p<0.0012 versus vehicle-treated animals). (B) Splenocytes were isolated, counted and analyzed for the total number of CD3<sup>+</sup>CD4<sup>-</sup>CD8<sup>-</sup> T cells (<i>left panel</i>; *p<0.0001, **p<0.0004) or the number of mature phenotype CD3<sup>+</sup>CD4<sup>+</sup> and CD3<sup>+</sup>CD8<sup>+</sup> T lymphocytes (<i>right panel</i>; *p = 0.04, **p = 0.002). (C) Numbers of B220<sup>-</sup>CD19<sup>-</sup>CD38<sup>+</sup>CD138<sup>+</sup> plasma cells (<i>left panel</i>; *p = 0.03, **p = 0.007, ***p = 0.004) and B220<sup>+</sup>CD19<sup>+</sup> B cells (<i>right panel</i>).</p

    Ganetespib degrades kinases associated with T cell activation and disease pathology.

    No full text
    <p>(A) Splenocytes from MRL/mp mice were incubated for 24 hours with or without activation by plate-bound CD3 + CD28 in the presence or absence of 100nM ganetespib. Cells were lysed and levels of the indicated kinases were determined by Western blot. (B) Relative expression levels for each protein were normalized to GAPDH using densitomery. Culture conditions: <i>rest</i>, resting (unactivated); <i>act</i>, activated; <i>act + g</i>, activated plus ganetespib.</p

    Elesclomol-induced ROS generation and cytoxicity in yeast is dependent on the presence of copper.

    No full text
    <p>(A) Chemical structure of the elesclomol-Cu complex. (B) The parental BY4743 yeast strain was grown in the presence of the indicated concentrations of elesclomol, preformed elesclomol-Cu, and/or copper for 21.5 h at 30°C. Absorbance at 600 nm was used to determine cell density. (C) Flow cytometric analysis measuring ROS in <i>S. cerevisiae</i>. ROS induction, as measured by Dihydrorhodamine 123 fluorescence, was only observed with preformed elesclomol-Cu complex at a concentration (500 nM) above the MIC but not below (100 nM), nor with free elesclomol at either concentration (<i>upper panel</i>). The addition of supplementary copper (via CuCl<sub>2</sub>) to elesclomol was sufficient to induce ROS, again only at the higher concentration (<i>lower panel</i>). (D) Elesclomol is cidal to yeast cells within an hour of treatment. Logarithmically growing cells were incubated with the indicated doses of elesclomol for 1, 2 or 4 h and then plated onto media without elesclomol. 5 µM and 22.5 µM elesclomol rendered cells unviable within 1 h, and lower doses (1.25 µM) killed cells within 4 h.</p

    Biological processes and protein complexes associated with sensitivity to elesclomol.

    No full text
    <p>(A) Each node represents a significantly enriched biological process/protein complex in the elesclomol chemogenomic profile as determined by GSEA. The size of a node corresponds to the number of genes annotated to the functional category. The width of an edge corresponds to the level of gene overlap between two interconnected categories (i.e. gene sets). Edges are not shown where the overlap coefficient is less than 0.5 (see <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0029798#s4" target="_blank">Materials and Methods</a>). The color of a node shows the cluster membership where clustering is based on the level of overlap between categories. (B) Each bar plot corresponds to the cluster indicated by its border color, and shows the individual sensitivity scores (X-axis) of the genes that contributed to the functional enrichments of the cluster. For clusters with more than 10 genes contributing to the enrichments, only the top 10 associated with the most categories are shown. The percentage of times the particular genes occur in that gene set is indicated by a color key, with black indicating the gene is present every time that function appears enriched, graded to white for those genes in the leading edge that appear less frequently within the gene set.</p

    Combinations of elesclomol-Cu and ETC complex inhibitors in melanoma cells.

    No full text
    <p>(A) Single agent viability assays in Hs249T human melanoma cells using graded concentrations of elesclomol-Cu, antimycin A or rotenone for 72 h. The IC<sub>50</sub> values obtained were 11 nM, 240 nM and 77 nM, respectively. Combination treatment of elesclomol-Cu with antimycin A (B) or rotenone (C) at IC<sub>20</sub> or IC<sub>50</sub> doses resulted in significantly enhanced cytotoxicity, showing that direct modulation of the ETC and mitochondrial respiration in mammalian cells enhances the cellular activity of elesclomol.</p

    Sensitivity of <i>S. cerevisiae</i> mutant strains to elesclomol-Cu.

    No full text
    <p>(A) A genome-wide readout of heterozygous strain sensitivity. In the plot, the X-axis orders all genes by their systematic name (hence, chromosome position) while the Y-axis is a measure of the ‘fitness’ of the strain deleted for the indicated gene grown in sub-lethal doses of elesclomol-Cu. The value on the Y-axis corresponds to −log<sub>2</sub> (ratio of normalized strain signal in treatment to DMSO control). Hence, the zero line represents equivalent growth in both conditions, while each unit above the line represents a 2-fold reduction in strain fitness. (B) Genome-wide profile of homozygous strain sensitivity. The data are presented as in <i>(A)</i>. Among the 150 most sensitive strains, the 137 having mitochondrial roles are highlighted color-coded: red (7 genes, mitochondrial genome maintenance), dark brown (8 genes, metal ion homeostasis), bright green (26 genes, mitochondrial localization), aqua (4 genes, mitochondrial, uncharacterized), blue (36 genes, ox-phos and respiration) mauve (5 genes, mitochondrial splicing), light gray (9 genes, response to stress) dark brown (26 genes, mitochondrial translation), dark gray (7 genes, mitochondrial import/export) and dark green (9 genes, mitochondrial tRNA).</p
    corecore