Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Nov 9.
Published in final edited form as: Pediatr Blood Cancer. 2013 Jan 9;60(7):E42–E45. doi: 10.1002/pbc.24451

Initial Testing (Stage 1) of Ganetespib, an Hsp90 Inhibitor, by the Pediatric Preclinical Testing Program

Richard B Lock 1,*, Hernan Carol 1, John M Maris 2, Min H Kang 3, C Patrick Reynolds 3, E Anders Kolb 4, Richard Gorlick 5, Stephen T Keir 6, Catherine A Billups 7, Raushan T Kurmasheva 8, Peter J Houghton 8, Malcolm A Smith 9
PMCID: PMC4225043  NIHMSID: NIHMS639063  PMID: 23303741

Abstract

Ganetespib, an Hsp90 inhibitor, was tested against the PPTP in vitro cell line panel and selected xenografts in vivo, including JAK2- and BRAF-mutated models. Ganetespib demonstrated potent in vitro cytotoxic activity (median rIC50 8.8 nM, range 4.4–27.1 nM). In vivo, ganetespib induced significant differences in EFS distribution for 4 of 11 xenografts. Intermediate activity (EFS T/C > 2) was noted only for the MV4;11 xenograft, and there were no objective responses. Administered as single agents, Hsp90 inhibitors examined by the PPTP have shown limited evidence for a therapeutic window against both solid tumor and leukemia pediatric preclinical models.

Keywords: Developmental therapeutics, Hsp90 inhibitors, preclinical testing

INTRODUCTION

Heat shock protein 90 (Hsp90) is an essential molecular chaperone that functions as part of a multi-protein complex in the post-translational stabilization of its protein substrates (client proteins) [1]. Hsp90 has emerged as an attractive target for the development of novel anti-cancer therapeutics, since many of its client proteins are implicated in the etiology of human cancer. Hsp90 substrates include protein kinases (e.g., BRAF, JAK) transcription factors (e.g., HIF-1α) and chimeric signaling proteins (e.g., EML4-ALK) [15]. Thus, inhibition of Hsp90 may result in simultaneous blockade of many oncogenic signaling pathways, leading to tumor cell death or enhanced sensitivity to chemotherapeutic drugs.

Ganetespib (formerly known as STA-9090) is a resorcinolic triazolone Hsp90 inhibitor currently in clinical trials for several adult human cancers [6]. Ganetespib, in contrast to some other Hsp90 inhibitors that have entered clinical evaluation, appears to lack ocular toxicities, an effect that is likely related to its more favorable retinal distribution and elimination [7]. Ganetespib induced objective responses in non-small cell lung cancer (NSCLC) patients with ALK translocations [8]. It is under evaluation as monotherapy for NSCLC patients with ALK gene rearrangement (NCT01562015), and it is being studied in a phase 2B/3 clinical trial in combination with docetaxel in patients with advanced NSCLC (NCT01348126).

The Pediatric Preclinical Testing Program (PPTP) utilizes well-characterized panels of in vitro cell lines and in vivo xenografts derived from a broad spectrum of pediatric malignancies to evaluate novel drugs for potential inclusion in pediatric cancer clinical trials [9]. Therefore, it was of interest to test ganetespib against the PPTP cell lines and a focused panel of xenografts with biological characteristics suggestive of susceptibility to Hsp90 inhibition.

MATERIALS AND METHODS

In Vitro Testing

In vitro testing was performed using DIMSCAN, as previously described [10]. Cells were incubated in the presence of ganetespib for 96 hours at concentrations from 0.1 nM to 1 μM and analyzed as previously described [11].

In Vivo Tumor Growth Inhibition Studies

CB17SC scid−/− mice (Taconic Farms, Germantown, NY), were used to propagate subcutaneously implanted neuroblastoma, astrocytoma, and MV4;11 tumors, as previously described [9]. Human leukemia cells were propagated by intravenous inoculation in non-obese diabetic (NOD)/scid−/− mice as described previously [12]. Responses were determined using three activity measures as previously described [9]. An in-depth description of the analysis methods is included in the Supplemental Response Definitions section.

Statistical Methods

The exact log-rank test, as implemented using Proc StatXact for SAS®, was used to compare event-free survival (EFS) distributions between treatment and control groups. P-values were two-sided and were not adjusted for multiple comparisons given the exploratory nature of the studies.

Drugs and Formulation

Ganetespib was provided to the PPTP by Synta Pharmaceuticals Corp., through the Cancer Therapy Evaluation Program (NCI). Powder was stored at 4°C, protected from light. Drug for in vivo treatments was formulated in DMSO/cremophor RH40/5% dextrose in sterile water (at 10:18:72 parts), and administered immediately. Ganetespib was administered I.V. at 100 mg/kg (ALL xenografts in NOD-SCID mice) and 125 mg/kg (solid tumor xenografts and MV4;11 in SCID mice) using a weekly 3× schedule followed by 3 weeks observation. Ganetespib was provided to each consortium investigator in coded vials for blinded testing.

RESULTS

In Vitro Testing

Ganetespib was tested against the PPTP’s in vitro cell line panel at concentrations ranging from 0.1 nM to 1.0 μM using a 96-hour exposure period. Ganetespib demonstrated potent cytotoxic activity, with T/C% values approaching 0% for most of the cell lines at the highest concentration tested and with median Relative In/Out value of −87% (Table I). The median relative IC50 (rIC50) value for the PPTP cell lines was 8.8 nM (range 4.4–27.1 nM; Table I).

TABLE I.

In Vitro Activity for Ganetespib

Cell line Histotype rIC50 (nM) Panel rIC50/line rIC50 Ymin (observed) Relative in/out (observed Ymin) (%)
RD Rhabdomyosarcoma 8.0 1.10 3.9 −28
Rh41 Rhabdomyosarcoma 10.4 0.85 2.2 −90
Rh18 Rhabdomyosarcoma 6.2 1.41 6.0 −87
Rh30 Rhabdomyosarcoma 5.6 1.58 3.2 −81
BT-12 Rhabdoid 14.3 0.61 2.2 −74
CHLA-266 Rhabdoid 27.1 0.32 2.5 −90
TC-71 Ewing sarcoma 4.5 1.97 0.9 −30
CHLA-9 Ewing sarcoma 4.6 1.91 0.2 −94
CHLA-10 Ewing sarcoma 5.7 1.55 6.1 −3
CHLA-258 Ewing sarcoma 6.4 1.38 0.0 −100
SJ-GBM2 Glioblastoma 12.9 0.68 2.0 −80
NB-1643 Neuroblastoma 7.4 1.19 7.5 −64
NB-EBc1 Neuroblastoma 16.8 0.52 4.8 −79
CHLA-90 Neuroblastoma 22.3 0.39 6.7 −76
CHLA-136 Neuroblastoma 23.2 0.38 3.0 −90
NALM-6 ALL 11.7 0.75 0.2 −94
COG-LL-317 ALL 4.4 2.02 0.0 −99
RS4;11 ALL 13.5 0.65 6.4 −58
MOLT-4 ALL 10.6 0.83 0.1 −99
CCRF-CEM (1) ALL 12.5 0.71 1.2 −81
CCRF-CEM (2) ALL 7.2 1.22 0.8 −87
Kasumi-1 AML 5.8 1.52 0.5 −98
Karpas-299 ALCL 9.6 0.92 0.3 −96
Ramos-RA1 NHL 7.4 1.20 0.0 −100
Median 8.8 1.01 2.1 −87
Minimum 4.4 0.32 0.0 −100
Maximum 27.1 2.02 7.5 −3

In Vivo Testing

The in vivo testing panel for ganetespib included selected translocation-positive sarcomas as well as the following models that were tested as subcutaneous xenografts: NB-1643, a neuroblastoma xenograft with an activating ALK mutation (R1275Q); BT-40, a juvenile pilocytic astrocytoma model with a BRAF V600E mutation; and MV4;11, an MLL-rearranged AML cell line with FLT3-ITD, reported to be responsive to ganetespib [6]. Additionally, four JAK-mutated ALL xenografts were tested using a systemic disease protocol. Ganetespib was tolerated at the doses employed, with a 4.9% toxicity rate for treated animals.

Ganetespib induced a significant difference in EFS distribution compared to control for MV4;11 TC-71, Rh41 and NB-1643, but not for the other seven tested xenografts (Table II, Supplemental Table I). Ganetespib induced tumor growth inhibition meeting criteria for intermediate EFS T/C activity (EFS T/C > 2) for only MV4;11, which had an EFS T/C value of 2.3. Objective responses were not noted for the 11 tested xenografts.

TABLE II.

Summary of In Vivo Activity of Ganetespib

Xenograft Line Histology Median time to event (days) P-value EFS T/C Median final RTV T/Ca T/C activity EFS activityb Response activityc
BT-40 BRAF mutated astrocytoma 14.9 0.791 0.9 >4 0.96 Low Low Low
NB-1643 Neuroblastoma 12.8 0.006 1.6 >4 0.5 Low Low Int
TC-71 Ewing sarcoma 16.4 <0.001 1.7 >4 0.41 Int Low Low
CHLA-258 Ewing sarcoma 14.7 0.445 1.0 >4 0.88 Low Low Low
Rh10 ALV rhabdomyosarcoma 20.2 0.125 1.2 4 0.74 Low Low Low
Rh41 ALV rhabdomyosarcoma 17.6 0.048 1.1 >4 0.73 Low Low Low
MV4;11 MLL-rearranged AML 26.7 <0.001 2.3 >4 0.24 Int Int Int
ALL-10 ALL JAK1 V658 5 0.200 1.0 >25 Low Low
TGT_020 ALL JAK2 R867Q 8.8 0.892 1.1 >25 Low Low
TGT_047 ALL JAK2 R683G 5.1 0.433 1.2 >25 Low Low
TGT_174 ALL JAK2 P933R 8.4 0.055 1.4 >25 Low Low
a

Tumor volume T/C value: Relative tumor volumes (RTV) for control (C) and treatment (T) mice were calculated at day 21 or when all mice in the control and treated groups still had measurable tumor volumes (if <21 days). The T/C value is the mean RTV for the treatment group divided by the mean RTV for the control group. High activity = T/C ≤ 0.15; Intermediate activity = T/C ≤ 0.45 but >0.15; and low activity = T/C > 0.45;

b

EFS T/C values: The ratio of the median time to event of the treatment group and the median time to event of the respective control group. High activity requires: (a) an EFS T/C > 2; (b) a significant difference in EFS distributions; and (c) a net reduction in median tumor volume for animals in the treated group at the end of treatment as compared to at treatment initiation. Intermediate activity = criteria (a) and (b) above, but not having a net reduction in median tumor volume for treated animals at the end of the study. Low activity = EFS T/C < 2;

c

Objective response measures are described in detail in the Supplemental Response Definitions. PD1 = progressive disease with EFS T/C ≤ 1.5, and PD2 = progressive disease with EFS T/C > 1.5.

DISCUSSION

The median rIC50 value of 8.8 nM observed for the PPTP cell lines is somewhat lower than a previous report using cell lines from adult tumors [6]. Ewing sarcoma cell lines were more sensitive to ganetespib than other PPTP cell lines, a finding that was also noted for the Hsp90 inhibitor AT13387 [13]. However, the difference in rIC50 for the Ewing cell lines and the non-Ewing cell lines is only twofold (5.1 nM vs. 10.5 nM, respectively).

MV4;11 was included for in vivo testing as this cell line was noted in a prior report to be especially responsive to ganetespib when grown as a subcutaneous xenograft [6]. In our study, although ganetespib showed a significant treatment effect against MV4;11 with EFS T/C > 2, tumor regression was not observed.

Ganetespib did not show activity against the BRAF-mutated astrocytoma xenograft, BT-40, a tumor responsive to the MEK inhibitor AZD6244 [14]. Mutated BRAF has been reported to form an Hsp90–cdc37 complex that is required for its stability and function, and treatment of BRAF-mutant melanoma cells with an Hsp90 inhibitor resulted in degradation of mutant BRAF and induction of apoptosis [2]. However, the in vivo preclinical activity of 17-AAG against BRAF-mutant melanoma xenografts was modest [2], as was its clinical activity [15].

Point mutations activating the ALK tyrosine kinase domain are reported to occur in most cases of familial neuroblastoma and 10% of sporadic cases [16,17]. One hotspot mutation (F1174L) has been shown to confer in vitro resistance to the ALK inhibitor crizotinib, but not to 17-AAG [18]. Therefore, it was of interest to test the in vivo efficacy of ganetespib against the NB-1643 neuroblastoma xenograft, which harbors another ALK hotspot mutation, R1275Q. Although ganetespib significantly delayed time to event for NB-1643, the treatment effect was not large (EFS T/C = 1.6). Modest activity was also observed for ganetespib against translocation positive Ewing sarcoma and rhabdomyosarcoma xenografts.

Ganetespib was evaluated against a panel of JAK-mutated ALL xenografts. JAK mutations are noted in 5–10% of B-precursor high-risk ALL and almost always occur with concurrent over-expression of CRLF2 resulting from genomic alterations fusing CRLF2 with either the IGH locus or with P2RY8 [1921]. Ganetespib showed no evidence of activity against the JAK-mutated xenografts, a result similar to that obtained by the PPTP for the JAK inhibitor AZD1480 [22]. Weigert et al. [5] have described results that were interpreted as representing promising activity for an Hsp90 inhibitor (AUY922) against a JAK2-mutated ALL xenograft. However, the methods employed in that report do not allow an assessment of remission status, and the extension in time to event for treated versus control animals (EFS T/C using PPTP terminology) was less than twofold. This level of activity would not be considered promising using the stringent activity assessments mployed by the PPTP.

The results presented here as well as prior reports by the PPTP provide limited evidence for an in vivo therapeutic window for Hsp90 inhibitors against solid tumor and ALL pediatric preclinical models [13,23]. These results contrast with the clinical activity observed for ganetespib for non-small cell lung cancer (NSCLC) patients with EML4-ALK translocations [8], and point to the possibility that future research may identify a pediatric cancer(s) with biological features that result in comparable responsiveness to Hsp90 inhibition.

Supplementary Material

Supp Material
Supp TableS1

Acknowledgments

This work was supported by NO1-CM-42216, CA21765, and CA108786 from the National Cancer Institute. Ganetespib was provided by Synta Pharmaceuticals Corp. In addition to the authors represents work contributed by the following: Sherry Ansher, Joshua Courtright, Kathryn Evans, Edward Favours, Henry S. Friedman, Charles Stopford, Chandra Tucker, Jianrong Wu, Joe Zeidner, Ellen Zhang, and Jian Zhang. JAK-mutated ALL xenografts were developed in collaboration with the Children’s Oncology Group. Children’s Cancer Institute Australia for Medical Research is affiliated with the University of New South Wales and Sydney Children’s Hospitals Network.

Grant sponsor: National Cancer Institute; Grant numbers: NO1-CM-42216, CA21765, CA108786.

Footnotes

Conflict of interest: Nothing to declare.

Additional Supporting Information may be found in the online version of this article.

References

  • 1.Cullinan SB, Whitesell L. Heat shock protein 90: A unique chemotherapeutic target. Semin Oncol. 2006;33:457–465. doi: 10.1053/j.seminoncol.2006.04.001. [DOI] [PubMed] [Google Scholar]
  • 2.Grbovic OM, Basso AD, Sawai A, et al. V600E B-Raf requires the Hsp90 chaperone for stability and is degraded in response to Hsp90 inhibitors. Proc Natl Acad Sci USA. 2006;103:57–62. doi: 10.1073/pnas.0609973103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Kim HL, Cassone M, Otvos L, Jr, et al. HIF-1alpha and STAT3 client proteins interacting with the cancer chaperone Hsp90: Therapeutic considerations. Cancer Biol Ther. 2008;7:10–14. doi: 10.4161/cbt.7.1.5458. [DOI] [PubMed] [Google Scholar]
  • 4.Normant E, Paez G, West KA, et al. The Hsp90 inhibitor IPI-504 rapidly lowers EML4-ALK levels and induces tumor regression in ALK-driven NSCLC models. Oncogene. 2011;30:2581–2586. doi: 10.1038/onc.2010.625. [DOI] [PubMed] [Google Scholar]
  • 5.Weigert O, Lane AA, Bird L, et al. Genetic resistance to JAK2 enzymatic inhibitors is overcome by HSP90 inhibition. J Exp Med. 2012;209:259–273. doi: 10.1084/jem.20111694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Ying W, Du Z, Sun L, et al. Ganetespib, a unique triazolone-containing Hsp90 inhibitor, exhibits potent antitumor activity and a superior safety profile for cancer therapy. Mol Cancer Ther. 2012;11:475–484. doi: 10.1158/1535-7163.MCT-11-0755. [DOI] [PubMed] [Google Scholar]
  • 7.Zhou D, Teofilovici F, Liu Y, et al. Associating retinal drug exposure and retention with the ocular toxicity profiles of Hsp90 inhibitors. J Clin Oncol. 2012;30(suppl):abstr 3086. [Google Scholar]
  • 8.Wong K, Koczywas M, Goldman JW, et al. An open-label phase II study of the Hsp90 inhibitor ganetespib (STA-9090) as monotherapy in patients with advanced non-small cell lung cancer (NSCLC) J Clin Oncol. 2011;29(suppl):abstr 7500. [Google Scholar]
  • 9.Houghton PJ, Morton CL, Tucker C, et al. The pediatric preclinical testing program: Description of models and early testing results. Pediatr Blood Cancer. 2006;49:928–940. doi: 10.1002/pbc.21078. [DOI] [PubMed] [Google Scholar]
  • 10.Frgala T, Kalous O, Proffitt RT, et al. A fluorescence microplate cytotoxicity assay with a 4-log dynamic range that identifies synergistic drug combinations. Mol Cancer Ther. 2007;6:886–897. doi: 10.1158/1535-7163.MCT-04-0331. [DOI] [PubMed] [Google Scholar]
  • 11.Houghton PJ, Morton CL, Kolb EA, et al. Initial testing (stage 1) of the proteasome inhibitor bortezomib by the pediatric preclinical testing program. Pediatr Blood Cancer. 2007;50:37–45. doi: 10.1002/pbc.21214. [DOI] [PubMed] [Google Scholar]
  • 12.Liem NL, Papa RA, Milross CG, et al. Characterization of childhood acute lymphoblastic leukemia xenograft models for the preclinical evaluation of new therapies. Blood. 2004;103:3905–3914. doi: 10.1182/blood-2003-08-2911. [DOI] [PubMed] [Google Scholar]
  • 13.Kang MH, Reynolds CP, Houghton PJ, et al. Initial testing (stage 1) of AT13387, an HSP90 inhibitor, by the pediatric preclinical testing program. Pediatr Blood Cancer. 2011;59:185–188. doi: 10.1002/pbc.23154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Kolb EA, Gorlick R, Houghton PJ, et al. Initial testing (stage 1) of AZD6244 (ARRY-142886) by the pediatric preclinical testing program. Pediatr Blood Cancer. 2010;55:668–677. doi: 10.1002/pbc.22576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Solit DB, Osman I, Polsky D, et al. Phase II trial of 17-allylamino-17-demethoxygeldanamycin in patients with metastatic melanoma. Clin Cancer Res. 2008;14:8302–8307. doi: 10.1158/1078-0432.CCR-08-1002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.De Brouwer S, De Preter K, Kumps C, et al. Meta-analysis of neuroblastomas reveals a skewed ALK mutation spectrum in tumors with MYCN amplification. Clin Cancer Res. 2010;16:4353–4362. doi: 10.1158/1078-0432.CCR-09-2660. [DOI] [PubMed] [Google Scholar]
  • 17.Mosse YP, Laudenslager M, Longo L, et al. Identification of ALK as a major familial neuroblastoma predisposition gene. Nature. 2008;455:930–935. doi: 10.1038/nature07261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Sasaki T, Okuda K, Zheng W, et al. The neuroblastoma-associated F1174L ALK mutation causes resistance to an ALK kinase inhibitor in ALK-translocated cancers. Cancer Res. 2010;70:10038–10043. doi: 10.1158/0008-5472.CAN-10-2956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Harvey RC, Mullighan CG, Chen IM, et al. Rearrangement of CRLF2 is associated with mutation of JAK kinases, alteration of IKZF1, Hispanic/Latino ethnicity, and a poor outcome in pediatric B-progenitor acute lymphoblastic leukemia. Blood. 2010;115:5312–5321. doi: 10.1182/blood-2009-09-245944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Mullighan CG, Collins-Underwood JR, Phillips LA, et al. Rearrangement of CRLF2 in B-progenitor-and Down syndrome-associated acute lymphoblastic leukemia. Nat Genet. 2009;41:1243–1246. doi: 10.1038/ng.469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Chen IM, Harvey RC, Mullighan CG, et al. Outcome modeling with CRLF2, IKZF1, JAK and minimal residual disease in pediatric acute lymphoblastic leukemia: A Children’s Oncology Group Study. Blood. 2012;119:3512–3522. doi: 10.1182/blood-2011-11-394221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Carol H, Lock R, Maris J, et al. Pediatric Preclinical Testing Program (PPTP) evaluation of the JAK inhibitor AZD1480. Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012. p. LB-318. [Google Scholar]
  • 23.Smith MA, Morton CL, Phelps DA, et al. Stage 1 testing and pharmacodynamic evaluation of the HSP90 inhibitor alvespimycin (17-DMAG, KOS-1022) by the pediatric preclinical testing program. Pediatr Blood Cancer. 2008;51:34–41. doi: 10.1002/pbc.21508. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supp Material
Supp TableS1

RESOURCES