The BCR-ABL oncoprotein of chronic myelogenous leukemia (CML) displays exclusive cytoplasmic localization and constitutive tyrosine kinase activity leading to the activation of different pathways that favor cell proliferation and survival. BCR-ABL induces survivin expression at both the mRNA and protein level, thus inhibiting the apoptotic machinery of CML cells and contributing to the expansion of the leukemic clone. We report that, in human CML cell lines, BCR-ABL–mediated upregulation of survivin involves the JAK2/STAT3 pathway since silencing of either protein caused a consistent reduction in survivin expression. Cell lines unresponsive to imatinib mesylate (IM) because of BCR-ABL gene amplification were not resensitized to the drug after survivin downregulation. However, cells insensitive to IM because of point mutations in the BCR-ABL kinase domain were highly responsive to hydroxyurea (HU) after survivin silencing. To address the possible clinical applications of our results, we used shepherdin, a cell-permeable peptidomimetic compound that downregulates survivin expression by preventing its interaction with Hsp90. Incubation with shepherdin of immortalized cell lines both sensitive and resistant to IM enhanced cell death induced by HU and doxorubicin. Similarly, the combination of shepherdin with first- and second-generation tyrosine kinase inhibitors reduced the colony-forming potential of human progenitors derived from both patients with IM-sensitive and IM-resistant CML. These results suggest that strategies aimed at reducing survivin levels may represent a potential therapeutic option for patients with CML unresponsive to IM. Mol Cancer Ther; 12(6); 1085–98. ©2013 AACR.

A reciprocal translocation between chromosomes 9 and 22 generates the abnormal Philadelphia (Ph) chromosome that, in turn, encodes for the chimaeric BCR-ABL oncogene (1). The ensuing BCR-ABL oncoprotein displays constitutive tyrosine kinase activity thereby activating multiple signaling pathways that lead to the development of chronic myeloid leukemia (CML; ref. 2). BCR-ABL–dependent activation of the phosphoinositide 3-kinase and the upregulation of Bcl-xL (3, 4) contribute to block cytochrome C release from the mitochondria and inhibit caspase activity, thus hampering the apoptotic machinery of CML cells and favoring the survival of leukemic myeloid progenitors. Treatment of CML cells with the ABL inhibitor imatinib mesylate (IM) suppresses BCR-ABL kinase activity reverting the antiapoptotic phenotype of the Ph-positive cells and resulting in the induction of cell death (5).

Survivin is a member of the inhibitor of apoptosis proteins (IAP) that is undetectable in normal tissues but highly expressed in most forms of human cancer and functions as both an inhibitor of cell death and a mitotic regulator (6–9). It has been reported that survivin overexpression reduces the sensitivity of neoplastic cells to death stimuli induced by gamma radiation (10) or chemotherapeutic agents (11, 12). The design of shepherdin, a cell-permeable peptidomimetic molecule that reduces survivin half-life by disrupting its binding with the molecular chaperone Hsp90, represents an attempt to increase the killing of cancer cells while preserving the viability of their normal counterpart (13).

We previously reported a significant correlation between BCR-ABL levels and the expression of survivin in a series of 44 patients with CML (14), suggesting that BCR-ABL could directly regulate survivin expression. Indeed, subsequent evidence indicated that BCR-ABL upregulates survivin at the mRNA and protein levels (15). A further study by Carter and colleagues suggested that the RAS/RAF/MAPK cascade was involved in BCR-ABL–dependent induction of survivin in a human CML cell line and that suppression of survivin expression increased responsiveness to IM therapy (16).

The aims of this study were 3-fold. We initially wanted to ascertain the signaling pathway(s) triggered by the BCR-ABL kinase to induce survivin expression. We then wanted to establish whether survivin overexpression contributed to the failure of IM treatment in a representative panel of CML cell lines with well-established mechanisms of IM resistance. Finally, we wished to determine whether reduced survivin levels increased the antiproliferative activity of different cytotoxic compounds.

We found that JAK2/STAT3 signaling modulates BCR-ABL–dependent induction of survivin and that survivin downregulation enhances the apoptotic effect of IM on CML cells that are sensitive to this compound. Lowering survivin expression increased the cytotoxic effect of hydroxyurea (HU) and doxorubicin (Doxo) on multiple cell lines unresponsive to IM. Likewise, exposure of IM-resistant cells to shepherdin reduced survivin expression and induced higher cell killing by both cytotoxic drugs and other ABL tyrosine kinase inhibitors (TKI).

Cell lines, primary cells, drugs, and shepherdin treatments

Human CML cell lines K562, KCL22, KYO-1, LAMA84, K562 S, K562 R, KCL22 S, KCL22 R, LAMA84 S, LAMA84 R, and murine Ba/F3 cells expressing either wild-type BCR-ABL (Ba/F3p210) or different BCR-ABL mutants (Ba/F3p210Y253F and Ba/F3p210T315I) were grown in RPMI-1640 (Sigma-Aldrich Corp) supplemented with 10% heat-inactivated FBS (Lonza), 2 mmol/L l-glutamine (Sigma), and penicillin/streptomycin (100 μg/mL each, also from Sigma). Human cell lines were obtained from the German Collection of Microorganisms and Cell Cultures DSMZ and used for less than 6 months after receipt with the exception of K562 S and R, KCL22 S and R, LAMA84 S and R that were provided by Carlo Gambacorti-Passerini (University of Milano Bicocca, Italy). The latter cells have not been authenticated in our lab. The K562, KCL22, and LAMA 84-IM–sensitive cell lines were maintained in parallel cultures without IM for the time required to obtain the IM-resistant clone. Ba/F3BCR-ABLp210, Ba/F3p210Y253F, and Ba/F3p210T315I were generated in our lab after viral transduction of the Ba/F3 pro-B cell line (purchased from DSMZ) with lentiviral vectors encoding for the specified BCR-ABL constructs.

Primary CML cells were obtained from the peripheral blood of patients with CML after receiving written informed consent. Total leukocytes were separated by red cell lysis.

The following drug treatments were used: 1 μmol/L IM (a gift from Novartis), 12.5 nmol/L nilotinib (a gift from Novartis), 50 μmol/L PD98059 (Calbiochem), 20 μmol/L LY294002 (Sigma), 7.5 nmol/L TG101209 (Symansis), 1 mmol/L HU (Sigma), 1 μmol/L doxorubicin (Sigma), 5 nmol/L dasatinib (a gift from Bristol-Myers Squibb), 17-Allylamino-17-demethoxygeldanamycin (17-AAG, InvivoGen; Fig. 1).

Figure 1.

Chemical structures of the specified drugs.

Figure 1.

Chemical structures of the specified drugs.

Close modal

In further experiments, we used 30 μmol/L of cell-permeable shepherdin [biotin-X-KKWKMRRNQFWVKVQRLFACGSSHK-CONH2; a gift from Dario Altieri, Wistar Cancer Institute, Philadelphia, PA] or an equimolar concentration of the corresponding scrambled peptide (biotin-X-KKWKMRRNQFWVKVQRGHSFCALKS-CONH2). The Antennapedia homeodomain sequence is underlined. X = EAHX, hexanoic acid spacer.

Immunoprecipitation and immunoblotting

Cell pellets were resuspended in isotonic buffer [25 mmol/l Trizma base (pH 8.5), 100 mmol/l NaCl, 7 mmol/l β-mercaptoethanol, 1× protease inhibitor cocktail (Roche)], sonicated and harvested by centrifugation at 14,000 rpm for 10 minutes at 4°C.

For immunoprecipitation experiments, 2 μg of antiphosphotyrosine antibody (clone 4G10, Millipore) were incubated with protein G-Sepharose (Amersham Biosciences) for 2 hours and then with 1 mg of cell lysates for an additional 2 hours. The sepharose beads were then washed in isotonic buffer and resuspended in Laemmli buffer.

For immunoblotting experiments, 40 μg of protein were separated by SDS-PAGE, transferred to nitrocellulose membranes, and blocked with 5% nonfat dry milk or with 10% bovine serum albumin (Sigma) in TBS with 0.2% Tween 20 (Sigma).

Monoclonal antibodies used were: antisurvivin (Abcam); antiphosphotyrosine (Millipore); anti-actin and anti-FLAG (Sigma). Polyclonal sera used were: anti-MAPK, anti-pMAPK, anti-AKT and anti-pAKT (Cell Signaling Technology); anti-JAK2, anti-STAT3 and anti-c-abl (clone K12; Santa Cruz Biotechnology). Appropriate horseradish peroxidase-conjugated secondary antibodies (Amersham Biosciences) were added and proteins were detected using the enhanced chemiluminescence reagent Lite Ablot (Euroclone).

Chemiluminescence images were captured and densitometric quantification of protein bands was carried out using the ImageJ 1.36b software (NIH, Bethesda, MD).

Cell proliferation and cell death assays

To evaluate cell viability, 2 × 104 cells were incubated, for 24 hours in 96-well plates with different drugs, siRNAs, or peptides and cell proliferation was subsequently determined using the luminescence ATP detection assay system ATPlite 1 step (Perkin-Elmer), following the manufacturer's instructions. To assess cell death, 5 × 105 cells were treated with different drugs or peptides, alone or in combination, for 24 hours. Cells were harvested and triplicate samples were selected to determine apoptosis using the FITC Annexin V Apoptosis Detection Kit I (BD Pharmingen) following the manufacturer's instructions. Apoptotic events were recorded as a combination of Annexin V+/PI− (early apoptosis) and Annexin V+/PI+ (late apoptosis) events and results were expressed as percentage of Annexin V+ cells.

Transfection and lentiviral transduction

For the production of the lentiviral supernatant, transfer vectors pGIPZ nonsilencing shRNA (Cat n° RHS4346) or pGIPZ shRNA anti-JAK2 (NM_004972, Cat n° RHS4430) were cotransfected with the packaging plasmids in the HEK293T cell line, using the calcium phosphate transfection method (Trans-Lentiviral shRNA Packaging Kit, Open Biosystems). Viral supernatants were harvested 48 hours after transfection. CML cells were then transduced by a double round of spin-infection. Cells were centrifuged at 1,200 × g for 90 minutes at 32°C in the presence of 1 mL of viral supernatant and 8 μg/mL polybrene (Sigma-Aldrich). Transduced cells were finally selected with 6 μg/mL puromycin (Sigma-Aldrich) for 72 hours.

Lentiviruses containing the pLEX empty vector or pLEX STAT3C were produced using the calcium phosphate transfection method (Trans-Lentiviral ORF Packaging Kit, Open Biosystems) as previously described.

RNA silencing experiments

For antisense oligonucleotides, 1 × 106 cells were resuspended in 0.8 mL of culture media mixed with 500 nmol/L antisense oligonucleotides against STAT3 (AS STAT3) or with a control oligonucleotide (AS Scr) and then electroporated in a 0.4 cm cuvette using the Gene Pulser Electroporation apparatus (Bio-Rad) with a double pulse protocol (25 μF, 500 V). Both the STAT3 antisense (5′-GCTCCAGCATCTGCTGCTTC-3′), and the control mismatch oligonucleotide (5′-GCTCCAATACCCGTTGCTTC-3′) were synthesized with a phosphorothioate backbone and with a 2′-O-methoxyethyl modification of the first and last 5 nucleotides (Eurofins MWG Operon).

For siRNA electroporation of Ba/F3 cells, cells were resuspended as previously described and mixed with 100 nmol/L of si Surv or with the corresponding scrambled control (si Scr; Dharmacon). si RNA electroporation was carried out as described above using different electrical settings (960 μF, 250 V).

STAT3 construct and mutagenesis

STAT3 was amplified by PCR using the indicated forward (FLAG-tag sequence underlined) 5′-ATAAGAATGCGGGCCGCGCCACCATGGATTACAAGGATGACGACGATAAGATGGCCCAATGGAATCAGCTAC-3′ and reverse 5′-CCGCTCGAGTCACATGGGGGAGGTAGCGC-3′ primers and cloned NotI and XhoI in the expression vector pcDNA3.1 (Life Technologies). STAT3 was then mutagenized using the Quik-Change II XL Site-Direct Mutagenesis Kit (Agilent Technologies). Primers used to generate point mutations were 5′-CATGGGCTATAAGATCATGGATTGTACCTGTATCCTGGTGTCTCCACTGGTC-3′ and 5′-GACCAGTGGAGACACCAGGATACAGGTACAATCCATGATCTTATAGCCCATG-3′. Incorporation of 2 point mutations was verified by direct sequencing. STAT3C was then cloned in the NotI and XhoI unique sites of the pLEX lentiviral vector (Open Biosystems).

Colony-forming assays

To evaluate the effect of different treatments on the colony-forming potential of CML myeloid progenitors, 5 × 104 primary cells were treated for 24 hours with shepherdin alone or in combination with IM, nilotinib, or dasatinib. Drug-treated cells were then seeded in Methocult H4435 methylcellulose medium (StemCell Technologies). Formation of colonies was assessed 14 days after plating.

Statistical analysis

Statistical analysis was conducted using GraphPad Prism 5.0 (GraphPad Software Inc). Unpaired, single-tail t tests with 95% confidence intervals were used to compare cell viability in different experimental conditions. The one-way ANOVAs according to Bonferroni's posttest were used to compare the effect of the combination of shepherdin with different tyrosine kinase inhibitors.

Survivin induction by BCR-ABL tyrosine kinase activity involves JAK2

We have previously reported that, in primary hematopoietic cells isolated from patients diagnosed with CML, the amount of BCR-ABL transcript significantly correlates with the expression levels of survivin (14). Our findings suggested that BCR-ABL might directly regulate survivin expression in CML cells and indeed, subsequent manuscripts have shown that BCR-ABL upregulates survivin at both the transcriptional and protein levels (15–17). However, these reports used single immortalized cell lines and attributed BCR-ABL–dependent induction of survivin to different signaling pathways.

To clarify these contrasting observations, we conducted an antisurvivin immunoblot on a wide panel of human CML cells before and after IM or nilotinib treatment. BCR-ABL inactivation produced a striking decrease in survivin levels in CML cells, confirming that BCR-ABL constitutive tyrosine kinase activity induces survivin expression (Fig. 2A and B). We next wanted to establish the BCR-ABL–induced pathway(s) responsible for this effect. Carter and colleagues had previously suggested that the RAS/RAF/MAPK cascade was involved in BCR-ABL–dependent upregulation of survivin (16). In a different cell line, Fang and colleagues identified JAK2/PI3K/c-myc signaling as the main regulator of BCR-ABL–dependent induction of survivin (17). However, when we suppressed mitogen-activated protein kinase (MAPK) activity with PD98059 in a set of 4 human CML cells, we detected a reduction in survivin levels only in the K562 line (Fig. 2C). Likewise, suppression of phosphoinositide 3-kinase (PI3K) activity by LY294002 induced a decrease in survivin expression only in LAMA84 cells (Fig. 2D). We therefore turned our attention to JAK2, as previous evidence had shown that BCR-ABL binds to JAK2 and phosphorylates it on tyrosine 1007, thereby activating its kinase activity (18). In turn, catalitically active JAK2 phosphorylates different STAT monomers inducing their dimerization, nuclear translocation, and transcriptional activity (18, 19). After blocking JAK2 signaling with the selective inhibitor TG101209, we observed a reduction in survivin expression in all CML lines tested (Fig. 2E). Prior evidence had established that JAK2 inhibition reduced survivin levels in AML cells (20). Furthermore, Fang and colleagues proposed that survivin upregulation was largely controlled at the transcriptional level through a JAK2-dependent mechanism (17). Our findings suggested a role for JAK2 in modulating survivin levels in CML cells.

Figure 2.

JAK2 inhibition reduces survivin expression in CML cells. The indicated cell lines were treated for 24 hours with 1 μmol/L IM (A), 12.5 nmol/L nilotinib (B), 50 μmol/L PD98059 (C), 20 μmol/L LY294002 (D), 7.5 nmol/L TG101209 (E), and subsequently assessed for their proliferation rate (left). Lysates were then assayed for the expression and phosphorylation of the specified proteins (middle; a blot representative of 3 separate experiments is shown). Histograms (right) depict survivin expression in each cell line normalized to actin, MAPK, AKT, or JAK2 as quantified by densitometric analysis, with survivin levels in untreated cells arbitrarily set at 1. Columns represent average ± SD of 3 independent experiments.

Figure 2.

JAK2 inhibition reduces survivin expression in CML cells. The indicated cell lines were treated for 24 hours with 1 μmol/L IM (A), 12.5 nmol/L nilotinib (B), 50 μmol/L PD98059 (C), 20 μmol/L LY294002 (D), 7.5 nmol/L TG101209 (E), and subsequently assessed for their proliferation rate (left). Lysates were then assayed for the expression and phosphorylation of the specified proteins (middle; a blot representative of 3 separate experiments is shown). Histograms (right) depict survivin expression in each cell line normalized to actin, MAPK, AKT, or JAK2 as quantified by densitometric analysis, with survivin levels in untreated cells arbitrarily set at 1. Columns represent average ± SD of 3 independent experiments.

Close modal

JAK2/STAT3 signaling contributes to BCR-ABL–dependent induction of survivin

To confirm that the BCR-ABL–dependent induction of survivin involves JAK2, we stably transduced CML cell lines with lentiviral vectors expressing a JAK2-specific shRNA or a nonsilencing control (Fig. 3A) and detected a reduction in survivin in all cell lines. Interestingly, a constitutive activation of the JAK2/STAT3 pathway by the BCR-ABL tyrosine kinase has been previously described in CML progenitors (21) and several reports have shown that STAT3 induces survivin expression in both solid and hematologic malignancies (22–24). Moreover, Bewry and colleagues have suggested that STAT3 might contribute to the development of IM resistance (25). To determine whether STAT3 was the pivotal downstream mediator of the BCR-ABL/JAK2 pathway, we electroporated STAT3-targeted antisense oligonucleotides or their scrambled counterpart in 4 CML lines. Immunoblot analyses showed that STAT3 downregulation reduced survivin levels in each leukemic cell line, whereas this was not the case with the scrambled control (Fig. 3B). To further confirm the role of STAT3 in the regulation of survivin, we generated an activated STAT3 (STAT3C) as previously reported by Bromberg and colleagues (Fig. 3C; ref. 26). We found that STAT3C promoted an increase in survivin levels in all 4 CML cells tested, implying that the JAK2/STAT3 cascade is a major regulator of survivin expression in CML cells.

Figure 3.

JAK2/STAT3 signaling contributes to BCR-ABL–mediated induction of survivin. A, protein extracts from the specified cell lines lentivirally transduced with either control (sh NS) or JAK2-specific (sh JAK2) shRNAs were blotted for survivin or JAK2 to verify the efficacy of the silencing procedure. B, to study the contribution of STAT3, the same cells were electroporated with antisense oligonucleotides against STAT3 (AS STAT3) or with a scrambled control (AS Scr). After 48 hours, lysates were blotted for survivin or STAT3 to confirm the decrease of the latter protein. Histograms on the right of each immunoblot measure survivin expression in the indicated conditions, with densitometric analysis conducted as detailed in the previous figure. Columns show average ± SD of 3 separate experiments. C, to further confirm the role of STAT3 in the regulation of survivin, cell lysates from CML cells lentivirally transduced with pLEX STAT3C or pLEX empty vector were blotted using antisurvivin (top), anti-FLAG (middle), or antiactin (bottom) antibodies. Panels show a blot representative of 3 independent experiments. **, P < 0.01; ***, P < 0.001; ns, not significant.

Figure 3.

JAK2/STAT3 signaling contributes to BCR-ABL–mediated induction of survivin. A, protein extracts from the specified cell lines lentivirally transduced with either control (sh NS) or JAK2-specific (sh JAK2) shRNAs were blotted for survivin or JAK2 to verify the efficacy of the silencing procedure. B, to study the contribution of STAT3, the same cells were electroporated with antisense oligonucleotides against STAT3 (AS STAT3) or with a scrambled control (AS Scr). After 48 hours, lysates were blotted for survivin or STAT3 to confirm the decrease of the latter protein. Histograms on the right of each immunoblot measure survivin expression in the indicated conditions, with densitometric analysis conducted as detailed in the previous figure. Columns show average ± SD of 3 separate experiments. C, to further confirm the role of STAT3 in the regulation of survivin, cell lysates from CML cells lentivirally transduced with pLEX STAT3C or pLEX empty vector were blotted using antisurvivin (top), anti-FLAG (middle), or antiactin (bottom) antibodies. Panels show a blot representative of 3 independent experiments. **, P < 0.01; ***, P < 0.001; ns, not significant.

Close modal

Survivin silencing sensitizes IM-resistant cells expressing BCR-ABL mutants to hydroxyurea

Resistance to IM has emerged as a major caveat in the long-term treatment of CML (27). BCR-ABL amplification or point mutations in the oncoprotein kinase domain represent 2 major mechanisms underlying IM failure. We wanted to establish whether survivin downregulation by specifically targeted siRNAs would increase the apoptotic effect of HU on CML cell lines displaying different mechanisms of IM resistance. To this end, we initially used LAMA84 R cells that are unresponsive to IM because of BCR-ABL gene amplification (28). Suppression of survivin in LAMA84 R failed to sensitize them to either HU or IM (Fig. 4B), indicating that resistance caused by BCR-ABL amplification is independent from survivin expression. However, survivin silencing in IM-responsive LAMA84 S increased the cytotoxic effect of both drugs (Fig. 4A). We next repeated these experiments using Ba/F3p210 (murine Ba/F3 cells lentivirally transduced with wild-type p210 BCR-ABL), Ba/F3p210Y253F, and Ba/F3p210T315I cell lines that are completely unresponsive to IM (>10-fold increase in the IC50 for the drug; refs. 28–30). In Ba/F3p210, survivin silencing caused a significant increase in both IM- and HU-induced death (Fig. 4C). Likewise, suppression of survivin in Ba/F3p210 cells expressing the Y253F and T315I mutants significantly increased the response to HU (Fig. 4D and E, middle). As anticipated, a reduction in survivin levels failed to restore responsiveness to IM (Fig. 4D and E, right), as point mutations induce conformational changes of the BCR-ABL kinase domain that prevent IM binding. These findings support the notion that lowering survivin expression can increase HU efficacy on CML cells unresponsive to IM because of BCR-ABL point mutations. These results also confirm that survivin does not affect the structural modifications of the BCR-ABL kinase domain induced by different amino acidic substitutions. Hence, reducing survivin levels fail to resensitize BCR-ABL mutants to the effect of IM.

Figure 4.

Survivin silencing sensitizes IM-resistant cells expressing BCR-ABL mutants to the effect of HU. The indicated IM-sensitive (A) and IM-resistant (B) cells were electroporated with siRNAs against survivin (si Surv) or with a scrambled control (si Scr) and analyzed by immunoblot to ensure survivin silencing (left panels). Twenty-four hours after electroporation, cells were treated with 1 mmol/L HU (middle panels) or 1 μmol/L IM (right) for 24 (white columns) or 48 (black columns) hours and the effect of these treatments was assessed using the ATPLite luminescence assay. The same experiments were repeated on murine Ba/F3 cells lentivirally transduced with either wild-type BCR-ABL (Ba/F3p210); C or with the indicated BCR-ABL mutants that confer IM resistance (D, E). Results shown represent average ± SD of at least 3 separate experiments conducted in triplicate with relative luminescence of untreated cells set at 100%. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ns, not significant.

Figure 4.

Survivin silencing sensitizes IM-resistant cells expressing BCR-ABL mutants to the effect of HU. The indicated IM-sensitive (A) and IM-resistant (B) cells were electroporated with siRNAs against survivin (si Surv) or with a scrambled control (si Scr) and analyzed by immunoblot to ensure survivin silencing (left panels). Twenty-four hours after electroporation, cells were treated with 1 mmol/L HU (middle panels) or 1 μmol/L IM (right) for 24 (white columns) or 48 (black columns) hours and the effect of these treatments was assessed using the ATPLite luminescence assay. The same experiments were repeated on murine Ba/F3 cells lentivirally transduced with either wild-type BCR-ABL (Ba/F3p210); C or with the indicated BCR-ABL mutants that confer IM resistance (D, E). Results shown represent average ± SD of at least 3 separate experiments conducted in triplicate with relative luminescence of untreated cells set at 100%. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ns, not significant.

Close modal

The Hsp90 peptidomimetic shepherdin sensitizes IM-resistant cells to different cytotoxic drugs

The design of shepherdin, a compound that reduces survivin expression by disrupting its interaction with Hsp90, represents an attractive opportunity to determine the possible therapeutic potential of survivin downregulation in CML cells. We therefore incubated IM-resistant cell lines K562 R, KCL22 R, LAMA84 R, and their IM-sensitive counterpart with shepherdin alone or in combination with HU or doxorubicin. As expected, addition of shepherdin, but not of the scramble control peptide, reduced survivin expression in all cell lines (Fig. 5A, D, and G). When we assayed cell proliferation and survival in the presence of shepherdin and HU or doxorubicin, we found significant reductions in the growth and viability of K562 and KCL22, regardless of their IM sensitivity (Fig. 5B, C, E, and F). Likewise, LAMA84 S cells were highly sensitive to the combination of shepherdin and different chemotherapeutic drugs (Fig. 5H). However, in agreement with previous silencing data (Fig. 4B), shepherdin failed to sensitize LAMA84 R to either HU or doxorubicin (Fig. 5I), confirming that resistance caused by BCR-ABL amplification is independent from survivin. These results were further validated by using the Ba/F3 cell model transformed by either wild-type (Ba/F3p210) or mutant BCR-ABL (Ba/F3p210Y253F, Ba/F3p210T315I). In the context of these cell lines, shepherdin efficiently suppressed survivin expression (Fig. 6A) and its association with either HU or doxorubicin significantly reduced cell proliferation and cell viability (Fig. 6B, C, and D).

Figure 5.

Treatment with the Hsp90 inhibitor shepherdin sensitizes IM-resistant cells to different cytotoxic drugs. IM-sensitive and IM-resistant cell lines were incubated for 24 hours with 30 μmol/L shepherdin (Shep) or with an equal concentration of a scrambled peptide (Scr). Alternatively, cells were exposed to 2 cytotoxic compounds [1 mmol/l HU, 1 μmol/L doxorubicin (Doxo)] in association with either Shep or Scr. Cell lysates were then examined by immunoblot to confirm survivin downregulation in the presence of shepherdin (A, D, and G). The effect of each combination was then assessed using the ATPLite luminescence assay (B, C, E, F, H, and I, left) or Annexin V-FITC/PI double staining (B, C, E, F, H, and I, right). Columns represent average ± SD of 3 independent experiments conducted in triplicate (for the ATPLite assay untreated cells were arbitrarily set at 100). *, P < 0.05; **, P < 0.01; ***, P < 0.001; ns, not significant.

Figure 5.

Treatment with the Hsp90 inhibitor shepherdin sensitizes IM-resistant cells to different cytotoxic drugs. IM-sensitive and IM-resistant cell lines were incubated for 24 hours with 30 μmol/L shepherdin (Shep) or with an equal concentration of a scrambled peptide (Scr). Alternatively, cells were exposed to 2 cytotoxic compounds [1 mmol/l HU, 1 μmol/L doxorubicin (Doxo)] in association with either Shep or Scr. Cell lysates were then examined by immunoblot to confirm survivin downregulation in the presence of shepherdin (A, D, and G). The effect of each combination was then assessed using the ATPLite luminescence assay (B, C, E, F, H, and I, left) or Annexin V-FITC/PI double staining (B, C, E, F, H, and I, right). Columns represent average ± SD of 3 independent experiments conducted in triplicate (for the ATPLite assay untreated cells were arbitrarily set at 100). *, P < 0.05; **, P < 0.01; ***, P < 0.001; ns, not significant.

Close modal
Figure 6.

Treatment with shepherdin sensitizes IM-resistant cells expressing BCR-ABL mutants to HU and doxorubicin (Doxo). Ba/F3 cells expressing either wild-type BCR-ABL or the indicated IM-resistant mutants were incubated for 24 hours with 30 μmol/L shepherdin (Shep) or with an equal concentration of a scrambled peptide (Scr). Alternatively, cells were exposed to 1 mmol/L HU or 1 μmol/L Doxo in association with either shepherdin or Scr. Cell lysates were then examined by immunoblot to confirm survivin downregulation in the presence of shepherdin (A). The effect of each combination was then assessed using the ATPLite luminescence assay (B–D, left) or Annexin V-FITC/PI double staining (B–D, right). Columns represent average ± SD of 3 independent experiments conducted in triplicate (for the ATPLite assay untreated cells were arbitrarily set at 100). *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Figure 6.

Treatment with shepherdin sensitizes IM-resistant cells expressing BCR-ABL mutants to HU and doxorubicin (Doxo). Ba/F3 cells expressing either wild-type BCR-ABL or the indicated IM-resistant mutants were incubated for 24 hours with 30 μmol/L shepherdin (Shep) or with an equal concentration of a scrambled peptide (Scr). Alternatively, cells were exposed to 1 mmol/L HU or 1 μmol/L Doxo in association with either shepherdin or Scr. Cell lysates were then examined by immunoblot to confirm survivin downregulation in the presence of shepherdin (A). The effect of each combination was then assessed using the ATPLite luminescence assay (B–D, left) or Annexin V-FITC/PI double staining (B–D, right). Columns represent average ± SD of 3 independent experiments conducted in triplicate (for the ATPLite assay untreated cells were arbitrarily set at 100). *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Close modal

Shepherdin increases the cytotoxic effect of nilotinib and dasatinib on human CML myeloid progenitors

To further ascertain the efficacy of shepherdin on BCR-ABL–expressing clones, we isolated primary cells from the peripheral blood of 3 different patients with CML: 2 individuals with newly diagnosed disease and a subject that had failed treatment with IM, nilotinib, and dasatinib. We then assessed the colony-forming potential of these cells after treatment with shepherdin alone or in combination with one of the 3 TKIs. Interestingly, we observed a significant decrease in overall colony number when shepherdin was associated with nilotinib and dasatinib, regardless of the TKI sensitivity of the primary cells (Fig. 7A, B, and C). These findings further reinforced the conclusion that downregulation of survivin expression may represent a potential therapeutic strategy to eliminate CML cells both responsive and unresponsive to the TKI-dependent inhibition of BCR-ABL kinase activity.

Figure 7.

Shepherdin sensitizes CML primary cells to different tyrosine-kinase inhibitors. Primary human cells derived from newly diagnosed (A and B) or TKI-refractory (C) patients with CML were pretreated for 24 hours with IM (1 μmol/L), nilotinib (Nil; 12.5 nmol/L), or dasatinib (Das; 5 nmol/L) either alone or in combination with shepherdin. Cells were then plated in triplicate in methylcellulose and the ensuing colonies were counted after 14 days. Colony-forming units (CFU) in each experimental condition are compared with those from untreated cells set at 100%. Values represent mean ± SD. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ns, not significant.

Figure 7.

Shepherdin sensitizes CML primary cells to different tyrosine-kinase inhibitors. Primary human cells derived from newly diagnosed (A and B) or TKI-refractory (C) patients with CML were pretreated for 24 hours with IM (1 μmol/L), nilotinib (Nil; 12.5 nmol/L), or dasatinib (Das; 5 nmol/L) either alone or in combination with shepherdin. Cells were then plated in triplicate in methylcellulose and the ensuing colonies were counted after 14 days. Colony-forming units (CFU) in each experimental condition are compared with those from untreated cells set at 100%. Values represent mean ± SD. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ns, not significant.

Close modal

Although IM has changed the natural course of CML achieving 89% overall survival rates after 8 years of follow-up (31), approximately 50% of patients with CML fail to obtain an optimal response as defined by the current European Leukemia Net (ELN) recommendations (32). To address these critical issues, second-generation TKIs (i.e., dasatinib and nilotinib) have been generated, tested, and approved for the treatment of patients with CML that fail IM (33). Despite the early success of these compounds, 50% of patients receiving either dasatinib or nilotinib will not obtain long-term benefit from these drugs and will progress to the advanced phases of the disease (34). Thus, the need for additional therapeutic strategies that will benefit this selected patient population (34, 35).

Carter and colleagues have previously shown that BCR-ABL induces the expression of the IAP survivin through the MAPK and PI3K pathways, and that reduction of survivin expression increases both spontaneous and IM-dependent death of a CML cell line (16). However, it is unclear whether these results could: (i) be extended to a more representative panel of CML cells both sensitive and resistant to IM; (ii) be used to potentiate the antiproliferative activity of other pharmacologic compounds; (iii) be exploited to devise a possible therapeutic strategy aimed at reducing survivin levels in patients with CML.

We report here that BCR-ABL kinase activity induced survivin expression in CML cell models via the JAK2/STAT3 pathway. Silencing of survivin reduced the viability of CML cells and enhanced the efficacy of IM. Moreover, survivin downregulation significantly increased the cytotoxic activity of HU, doxorubicin, nilotinib, and dasatinib on cells unresponsive to IM because of either point mutations (including T315I) in the BCR-ABL catalytic domain or other yet uncharacterized mechanisms. These results were obtained with the use of survivin-targeted siRNAs or by using shepherdin, a peptidomimetic compound that reduces survivin half-life by abrogating its interaction with the Hsp90 chaperone (13). These findings were further confirmed by experiments on human myeloid progenitors derived from both IM-sensitive and IM-resistant patients with CML.

Our observations lead to several considerations. First, and at variance with the data reported by Carter and colleagues, we found that only MAPK and PI3K signaling cascades modestly contribute to the regulation of survivin expression in CML. Conversely, we identified the JAK2/STAT3 pathway as a major contributor of BCR-ABL–mediated induction of survivin. This discrepancy could be due to the different cells used in the 2 studies. It is also possible that, in certain cell types, both JAK2 and MAPK contribute to survivin overexpression, as published evidence suggests that JAK2 and MAPK stimulate phosphorylation of STAT3, thus enhancing its biologic activity (36).

We have also found that suppression of survivin is unable to restore IM activity regardless of the mechanisms responsible for drug failure. These results are not unexpected as IM resistance attributable to BCR-ABL gene amplification is due to the enhanced signaling of a higher absolute number of BCR-ABL molecules (37). Intuitively, this phenomenon cannot be constrained by lowering survivin expression. Likewise, point mutations associated with IM failure induce alterations in the 3-dimensional structure of the BCR-ABL catalytic domain that either mimic an active conformation or affect specific amino-acidic residues required for the physical interaction with IM (38, 39). Thus, it is evident that suppression of survivin per se fails to restore sensitivity to IM, as survivin does not associate with BCR-ABL and is therefore incapable of modulating the conformation of its kinase domain.

However, suppression of survivin levels significantly increased the cytotoxic activity of different compounds on CML cells failing IM because of yet uncharacterized mechanisms or point mutations in the BCR-ABL kinase domain. In these contexts, suppression of survivin may represent a potential therapeutic strategy to reduce the viability of these leukemic clones. To this end, we have conducted several experiments using the peptidomimetic shepherdin to lower survivin expression in IM-sensitive and IM-resistant CML cells. Previous studies investigating the use of shepherdin have described strong antitumoral activity in human models of both solid and hematologic malignancies (13, 40). Our findings indicate that shepherdin-dependent reduction of survivin levels significantly increased the cytotoxic activity of HU and doxorubicin on all cell lines expressing BCR-ABL mutants. In addition, as further compounds (ISIS 23722, SPC3042, and YM155) are currently being developed as inhibitors of survivin expression (41), any pharmacologic strategy aimed at reducing survivin levels could be of significant value in patients with IM-resistant clones carrying point mutations in the BCR-ABL kinase domain.

Finally, the reduction in cell viability that we observed after shepherdin treatment was superior to that detected after siRNA-mediated silencing, suggesting that shepherdin may increase CML cell death by both survivin-dependent and survivin-independent mechanisms. Kang and colleagues have previously shown that shepherdin disables a mitochondrial chaperone network that cancer cells use to protect themselves from multiple apoptotic stimuli (42). Hence, it is possible that part of shepherdin's efficacy on CML cells depends on this mechanism. Indeed, when we treated Ba/F3 cells expressing different BCR-ABL mutants with HU or doxorubicin in association with 17-AAG (a cytosolic inhibitor of Hsp90), we found no significant increases in cell death (Fig. 8). These preliminary findings support the hypothesis that shepherdin kills CML cells through a double mechanism: (i) a reduction in survivin expression and (ii) the mithochondrial inhibition of Hsp90 chaperones.

Figure 8.

The combination of 17-AAG and HU or doxorubicin (Doxo) does not increase death rates of Ba/F3 cells expressing BCR-ABL mutants. Ba/F3 cells expressing either wild-type BCR-ABL or the indicated IM-resistant mutants were incubated for 24 hours with 0.5 μmol/L or 0.75 μmol/L 17-AAG. Cell lysates were then examined by immunoblot to ascertain changes in BCR-ABL expression (A). Alternatively, the same cells were exposed to 1 mmol/L HU or 1 μmol/L Doxo in association with 0.5 μmol/L 17-AAG. The effect of each combination on cell viability was then assessed using Annexin V-FITC/PI double staining (B–D). Columns represent average ± SD of 3 independent experiments conducted in triplicate. ns, not significant.

Figure 8.

The combination of 17-AAG and HU or doxorubicin (Doxo) does not increase death rates of Ba/F3 cells expressing BCR-ABL mutants. Ba/F3 cells expressing either wild-type BCR-ABL or the indicated IM-resistant mutants were incubated for 24 hours with 0.5 μmol/L or 0.75 μmol/L 17-AAG. Cell lysates were then examined by immunoblot to ascertain changes in BCR-ABL expression (A). Alternatively, the same cells were exposed to 1 mmol/L HU or 1 μmol/L Doxo in association with 0.5 μmol/L 17-AAG. The effect of each combination on cell viability was then assessed using Annexin V-FITC/PI double staining (B–D). Columns represent average ± SD of 3 independent experiments conducted in triplicate. ns, not significant.

Close modal

In summary, our results suggest that pharmacologic reduction of survivin expression may provide a new therapeutic approach for the molecular targeting of IM-insensitive CML cells.

No potential conflicts of interest were disclosed.

Conception and design: S. Stella, E. Tirrò, E. Conte, P. Vigneri

Development of methodology: S. Stella, E. Tirrò

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): S. Stella, E. Conte, F. Stagno, F.D. Raimondo

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): S. Stella, E. Tirrò, F. Stagno, F.D. Raimondo, P. Vigneri

Writing, review, and/or revision of the manuscript: S. Stella, E. Tirrò, E. Conte, F. Stagno, F.D. Raimondo, L. Manzella, P. Vigneri

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): S. Stella, P. Vigneri

Study supervision: S. Stella, P. Vigneri

This work was supported by a fellowship (E. Tirrò) and a national grant from Associazione Italiana per la Ricerca sul Cancro (P. Vigneri).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Nowell
PC
. 
Discovery of the Philadelphia chromosome: a personal perspective
.
J Clin Invest
2007
;
117
:
2033
5
.
2.
Ren
R
. 
Mechanisms of BCR-ABL in the pathogenesis of chronic myelogenous leukaemia
.
Nat Rev Cancer
2005
;
5
:
172
83
.
3.
Skorski
T
,
Bellacosa
A
,
Nieborowska-Skorska
M
,
Majewski
M
,
Martinez
R
,
Choi
JK
, et al
Transformation of hematopoietic cells by BCR/ABL requires activation of a PI-3k/Akt-dependent pathway
.
Embo J
1997
;
16
:
6151
61
.
4.
Amarante-Mendes
GP
,
Naekyung
Kim C
,
Liu
L
,
Huang
Y
,
Perkins
CL
,
Green
DR
, et al
Bcr-Abl exerts its antiapoptotic effect against diverse apoptotic stimuli through blockage of mitochondrial release of cytochrome C and activation of caspase-3
.
Blood
1998
;
91
:
1700
5
.
5.
Druker
BJ
,
Tamura
S
,
Buchdunger
E
,
Ohno
S
,
Segal
GM
,
Fanning
S
, et al
Effects of a selective inhibitor of the Abl tyrosine kinase on the growth of Bcr-Abl positive cells
.
Nat Med
1996
;
2
:
561
6
.
6.
Altieri
DC
. 
Validating survivin as a cancer therapeutic target
.
Nat Rev Cancer
2003
;
3
:
46
54
.
7.
Altieri
DC
. 
Survivin, cancer networks and pathway-directed drug discovery
.
Nat Rev Cancer
2008
;
8
:
61
70
.
8.
Fukuda
S
,
Pelus
LM
. 
Survivin, a cancer target with an emerging role in normal adult tissues
.
Mol Cancer Ther
2006
;
5
:
1087
98
.
9.
Lladser
A
,
Sanhueza
C
,
Kiessling
R
,
Quest
AF
. 
Is survivin the potential Achilles' heel of cancer?
Adv Cancer Res
.
111
:
1
37
.
10.
Lu
B
,
Mu
Y
,
Cao
C
,
Zeng
F
,
Schneider
S
,
Tan
J
, et al
Survivin as a therapeutic target for radiation sensitization in lung cancer
.
Cancer Res
2004
;
64
:
2840
5
.
11.
Li
F
,
Ambrosini
G
,
Chu
EY
,
Plescia
J
,
Tognin
S
,
Marchisio
PC
, et al
Control of apoptosis and mitotic spindle checkpoint by survivin
.
Nature
1998
;
396
:
580
4
.
12.
Chandele
A
,
Prasad
V
,
Jagtap
JC
,
Shukla
R
,
Shastry
PR
. 
Upregulation of survivin in G2/M cells and inhibition of caspase 9 activity enhances resistance in staurosporine-induced apoptosis
.
Neoplasia
2004
;
6
:
29
40
.
13.
Plescia
J
,
Salz
W
,
Xia
F
,
Pennati
M
,
Zaffaroni
N
,
Daidone
MG
, et al
Rational design of shepherdin, a novel anticancer agent
.
Cancer Cell
2005
;
7
:
457
68
.
14.
Conte
E
,
Stagno
F
,
Guglielmo
P
,
Scuto
A
,
Consoli
C
,
Messina
A
. 
Survivin expression in chronic myeloid leukemia
.
Cancer Lett
2005
;
225
:
105
10
.
15.
Wang
Z
,
Sampath
J
,
Fukuda
S
,
Pelus
LM
. 
Disruption of the inhibitor of apoptosis protein survivin sensitizes Bcr-abl-positive cells to STI571-induced apoptosis
.
Cancer Res
2005
;
65
:
8224
32
.
16.
Carter
BZ
,
Mak
DH
,
Schober
WD
,
Cabreira-Hansen
M
,
Beran
M
,
McQueen
T
, et al
Regulation of survivin expression through Bcr-Abl/MAPK cascade: targeting survivin overcomes imatinib resistance and increases imatinib sensitivity in imatinib-responsive CML cells
.
Blood
2006
;
107
:
1555
63
.
17.
Fang
ZH
,
Dong
CL
,
Chen
Z
,
Zhou
B
,
Liu
N
,
Lan
HF
, et al
Transcriptional regulation of survivin by c-Myc in BCR/ABL-transformed cells: implications in anti-leukaemic strategy
.
J Cell Mol Med
2009
;
13
:
2039
52
.
18.
Samanta
AK
,
Lin
H
,
Sun
T
,
Kantarjian
H
,
Arlinghaus
RB
. 
Janus kinase 2: a critical target in chronic myelogenous leukemia
.
Cancer Res
2006
;
66
:
6468
72
.
19.
Xie
S
,
Wang
Y
,
Liu
J
,
Sun
T
,
Wilson
MB
,
Smithgall
TE
, et al
Involvement of Jak2 tyrosine phosphorylation in Bcr-Abl transformation
.
Oncogene
2001
;
20
:
6188
95
.
20.
Faderl
S
,
Harris
D
,
Van
Q
,
Kantarjian
HM
,
Talpaz
M
,
Estrov
Z
. 
Granulocyte-macrophage colony-stimulating factor (GM-CSF) induces antiapoptotic and proapoptotic signals in acute myeloid leukemia
.
Blood
2003
;
102
:
630
7
.
21.
Coppo
P
,
Dusanter-Fourt
I
,
Millot
G
,
Nogueira
MM
,
Dugray
A
,
Bonnet
ML
, et al
Constitutive and specific activation of STAT3 by BCR-ABL in embryonic stem cells
.
Oncogene
2003
;
22
:
4102
10
.
22.
Aoki
Y
,
Feldman
GM
,
Tosato
G
. 
Inhibition of STAT3 signaling induces apoptosis and decreases survivin expression in primary effusion lymphoma
.
Blood
2003
;
101
:
1535
42
.
23.
Dalwadi
H
,
Krysan
K
,
Heuze-Vourc'h
N
,
Dohadwala
M
,
Elashoff
D
,
Sharma
S
, et al
Cyclooxygenase-2-dependent activation of signal transducer and activator of transcription 3 by interleukin-6 in non-small cell lung cancer
.
Clin Cancer Res
2005
;
11
:
7674
82
.
24.
Diaz
N
,
Minton
S
,
Cox
C
,
Bowman
T
,
Gritsko
T
,
Garcia
R
, et al
Activation of stat3 in primary tumors from high-risk breast cancer patients is associated with elevated levels of activated SRC and survivin expression
.
Clin Cancer Res
2006
;
12
:
20
8
.
25.
Bewry
NN
,
Nair
RR
,
Emmons
MF
,
Boulware
D
,
Pinilla-Ibarz
J
,
Hazlehurst
LA
. 
Stat3 contributes to resistance toward BCR-ABL inhibitors in a bone marrow microenvironment model of drug resistance
.
Mol Cancer Ther
2008
;
7
:
3169
75
.
26.
Bromberg
JF
,
Wrzeszczynska
MH
,
Devgan
G
,
Zhao
Y
,
Pestell
RG
,
Albanese
C
, et al
Stat3 as an oncogene
.
Cell
1999
;
98
:
295
303
.
27.
Deininger
M
. 
Resistance and relapse with imatinib in CML: causes and consequences
.
J Natl Compr Canc Netw
2008
;
6
Suppl 2
:
S11
S21
.
28.
le Coutre
P
,
Tassi
E
,
Varella-Garcia
M
,
Barni
R
,
Mologni
L
,
Cabrita
G
, et al
Induction of resistance to the Abelson inhibitor STI571 in human leukemic cells through gene amplification
.
Blood
2000
;
95
:
1758
66
.
29.
La Rosee
P
,
Johnson
K
,
Corbin
AS
,
Stoffregen
EP
,
Moseson
EM
,
Willis
S
, et al
In vitro efficacy of combined treatment depends on the underlying mechanism of resistance in imatinib-resistant Bcr-Abl-positive cell lines
.
Blood
2004
;
103
:
208
15
.
30.
Piazza
RG
,
Magistroni
V
,
Gasser
M
,
Andreoni
F
,
Galietta
A
,
Scapozza
L
, et al
Evidence for D276G and L364I Bcr-Abl mutations in Ph+ leukaemic cells obtained from patients resistant to Imatinib
.
Leukemia
2005
;
19
:
132
4
.
31.
Deininger
M
,
O'Brien
SG
,
Guilhot
F
,
Goldman
JM
,
Hochhaus
A
,
Hughes
TP
, et al
International randomized study of interferon vs STI571 (IRIS) 8-year follow up: sustained survival and low risk for progression or events in patients with newly diagnosed chronic myeloid leukemia in chronic phase (CML-CP) treated with imatinib
.
ASH Annual Meeting Abstracts
2009
;
114
:
1126
.
32.
Baccarani
M
,
Cortes
J
,
Pane
F
,
Niederwieser
D
,
Saglio
G
,
Apperley
J
, et al
Chronic myeloid leukemia: an update of concepts and management recommendations of European LeukemiaNet
.
J Clin Oncol
2009
;
27
:
6041
51
.
33.
Quintas-Cardama
A
,
Kantarjian
H
,
Cortes
J
. 
Imatinib and beyond–exploring the full potential of targeted therapy for CML
.
Nat Rev Clin Oncol
2009
;
6
:
535
43
.
34.
Jabbour
E
,
Jones
D
,
Kantarjian
HM
,
OBrien
S
,
Tam
C
,
Koller
C
, et al
Long-term outcome of patients with chronic myeloid leukemia treated with second-generation tyrosine kinase inhibitors after imatinib failure is predicted by the in vitro sensitivity of BCR-ABL kinase domain mutations
.
Blood
2009
;
114
:
2037
43
.
35.
O'Brien
S
,
Abboud
CN
,
Akhtari
M
,
Altman
J
,
Berman
E
,
DeAngelo
DJ
, et al
Chronic myelogenous leukemia. Clinical practice guidelines in oncology
.
J Natl Compr Canc Netw
2012
;
10
:
64
110
.
36.
Coppo
P
,
Flamant
S
,
De Mas
V
,
Jarrier
P
,
Guillier
M
,
Bonnet
ML
, et al
BCR-ABL activates STAT3 via JAK and MEK pathways in human cells
.
Br J Haematol
2006
;
134
:
171
9
.
37.
Shannon
KM
. 
Resistance in the land of molecular cancer therapeutics
.
Cancer Cell
2002
;
2
:
99
102
.
38.
Nagar
B
,
Hantschel
O
,
Young
MA
,
Scheffzek
K
,
Veach
D
,
Bornmann
W
, et al
Structural basis for the autoinhibition of c-Abl tyrosine kinase
.
Cell
2003
;
112
:
859
71
.
39.
Lovera
S
,
Sutto
L
,
Boubeva
R
,
Scapozza
L
,
Dolker
N
,
Gervasio
FL
. 
The different flexibility of c-Src and c-Abl kinases regulates the accessibility of a druggable inactive conformation
.
J Am Chem Soc
2012
;
134
:
2496
9
.
40.
Gyurkocza
B
,
Plescia
J
,
Raskett
CM
,
Garlick
DS
,
Lowry
PA
,
Carter
BZ
, et al
Antileukemic activity of shepherdin and molecular diversity of hsp90 inhibitors
.
J Natl Cancer Inst
2006
;
98
:
1068
77
.
41.
Ling
X
,
Cao
S
,
Cheng
Q
,
Keefe
JT
,
Rustum
YM
,
Li
F
. 
A novel small molecule FL118 that selectively inhibits survivin, Mcl-1, XIAP and cIAP2 in a p53-independent manner, shows superior antitumor activity
.
PLoS One
2012
;
7
:
e45571
.
42.
Kang
BH
,
Plescia
J
,
Dohi
T
,
Rosa
J
,
Doxsey
SJ
,
Altieri
DC
. 
Regulation of tumor cell mitochondrial homeostasis by an organelle-specific Hsp90 chaperone network
.
Cell
2007
;
131
:
257
70
.

Supplementary data