Antitumor activity of rapamycin in a Phase I trial for patients with recurrent PTEN-deficient glioblastoma

Tim F Cloughesy, Koji Yoshimoto, Phioanh Nghiemphu, Kevin Brown, Julie Dang, Shaojun Zhu, Teli Hsueh, Yinan Chen, Wei Wang, David Youngkin, Linda Liau, Neil Martin, Don Becker, Marvin Bergsneider, Albert Lai, Richard Green, Tom Oglesby, Michael Koleto, Jeff Trent, Steve Horvath, Paul S Mischel, Ingo K Mellinghoff, Charles L Sawyers, Tim F Cloughesy, Koji Yoshimoto, Phioanh Nghiemphu, Kevin Brown, Julie Dang, Shaojun Zhu, Teli Hsueh, Yinan Chen, Wei Wang, David Youngkin, Linda Liau, Neil Martin, Don Becker, Marvin Bergsneider, Albert Lai, Richard Green, Tom Oglesby, Michael Koleto, Jeff Trent, Steve Horvath, Paul S Mischel, Ingo K Mellinghoff, Charles L Sawyers

Abstract

Background: There is much discussion in the cancer drug development community about how to incorporate molecular tools into early-stage clinical trials to assess target modulation, measure anti-tumor activity, and enrich the clinical trial population for patients who are more likely to benefit. Small, molecularly focused clinical studies offer the promise of the early definition of optimal biologic dose and patient population.

Methods and findings: Based on preclinical evidence that phosphatase and tensin homolog deleted on Chromosome 10 (PTEN) loss sensitizes tumors to the inhibition of mammalian target of rapamycin (mTOR), we conducted a proof-of-concept Phase I neoadjuvant trial of rapamycin in patients with recurrent glioblastoma, whose tumors lacked expression of the tumor suppressor PTEN. We aimed to assess the safety profile of daily rapamycin in patients with glioma, define the dose of rapamycin required for mTOR inhibition in tumor tissue, and evaluate the antiproliferative activity of rapamycin in PTEN-deficient glioblastoma. Although intratumoral rapamycin concentrations that were sufficient to inhibit mTOR in vitro were achieved in all patients, the magnitude of mTOR inhibition in tumor cells (measured by reduced ribosomal S6 protein phosphorylation) varied substantially. Tumor cell proliferation (measured by Ki-67 staining) was dramatically reduced in seven of 14 patients after 1 wk of rapamycin treatment and was associated with the magnitude of mTOR inhibition (p = 0.0047, Fisher exact test) but not the intratumoral rapamycin concentration. Tumor cells harvested from the Ki-67 nonresponders retained sensitivity to rapamycin ex vivo, indicating that clinical resistance to biochemical mTOR inhibition was not cell-intrinsic. Rapamycin treatment led to Akt activation in seven patients, presumably due to loss of negative feedback, and this activation was associated with shorter time-to-progression during post-surgical maintenance rapamycin therapy (p < 0.05, Logrank test).

Conclusions: Rapamycin has anticancer activity in PTEN-deficient glioblastoma and warrants further clinical study alone or in combination with PI3K pathway inhibitors. The short-term treatment endpoints used in this neoadjuvant trial design identified the importance of monitoring target inhibition and negative feedback to guide future clinical development.

Trial registration: http://www.ClinicalTrials.gov (#NCT00047073).

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Clinical Trial Design
Figure 1. Clinical Trial Design
Enrollment in the Phase I clinical trial was restricted to patients whose initial tumor resection (“surgery 1”) specimen was PTEN-deficient by immunohistochemistry. Patients were enrolled after failing standard therapy with radiation and chemotherapy (i.e., “tumor recurrence”). Prior to the scheduled salvage tumor resection (“surgery 2”), patients received a short course (mean: 7.5 d) of oral rapamycin. Rapamycin was resumed after recovery from surgery until patients developed clinical and/or radiographic evidence of treatment failure. The effects of rapamycin on tumor cell proliferation and mTOR signaling in tumor tissue were determined by comparing the tumor tissue collected during salvage resection (“surgery 2”) with a sample of the same tumor collected during the initial tumor resection (“surgery 1”). Time-to-progression (TTP) was defined as the interval between start of rapamycin therapy and postoperative treatment failure.
Figure 2. Rapamycin Crosses the Blood–Brain Barrier…
Figure 2. Rapamycin Crosses the Blood–Brain Barrier and Blocks mTOR in Tumor Tissue
(A) Rapamycin concentrations in tumor tissue (filled squares) and peripheral blood (empty circles) grouped by rapamycin dose cohorts (2 mg, 5 mg, or 10 mg per os daily). Intratumoral rapamycin concentration for patient 11 could not be determined due to insufficient frozen tumor material. The last preoperative dose of rapamycin was given on the day of craniotomy and peripheral blood was collected within 24 h of surgery. (B) Quantification of mTOR activity in tumor tissue by immunohistochemistry. The cartoon to the left depicts the S6 kinase 1 branch of the mTOR signaling pathway resulting in phosphorylation of S6 ribosomal protein at serine 235/236 and serine 240/244. The panel to the right shows a comparison between immunoblotting (top) and IHC (bottom) for the determination of S6 phosphorylation in tumor tissue from rapamycin patients 1, 2, and 3. The fold change in serine 235/236 phosphorylation between S2 and S1 for patients 1, 2, and 3 were 0.45, 1.01, and 0.45, respectively (see Figure S2A). (C) Changes in S6 phosphorylation between S2 and S1 (y-axis: ratio of S6 phosphorylation in S2 sample to S6 phosphorylation in S1 sample) for all patients for whom matched S1 and S2 samples were available (14/15 rapamycin patients and 9/9 patients who did not receive rapamycin). S6 phosphorylation was determined by IHC using phosphosite-specific antibodies against serine 235/236 (left) and serine 240/244 (right). Please see Figures S1 and S2 for details regarding IHC scoring method and results for individual tumors. p-values for the difference in mean S2/S1 ratios for each group were determined using the Kruskal Wallace test.
Figure 3. Rapamycin Inhibits Tumor Cell Proliferation…
Figure 3. Rapamycin Inhibits Tumor Cell Proliferation in Subsets of PTEN-Deficient GBMs
(A) Ki-67 labeling index of GBMs before (S1) and during (S2) rapamycin therapy compared with tumor samples from patients who did not receive rapamycin (see text for detail). The horizontal line inside a box plot shows the median value. The lower and upper end of the box corresponds to the 25th and 75th percentile, respectively. The whiskers extend to the 95% range. The Wilcoxon nonparametric group comparison test was used to calculate p-values for the differences between different patient groups. (B) Changes in tumor cell proliferation between S1 and S2 for 14/15 rapamycin patients. The horizontal line inside the box indicates the median value. The lower and upper border of the box corresponds to the 25th and 75th percentile, respectively. The whiskers extend to the 95% range. Paraffin blocks from patient 14 were not sufficient for quantification. The median Ki-67 labeling index of the S2 specimen from patients 1, 3, 8, 9, 11, and 13 was 0 (indicated by asterix). N.S. indicates that the difference in Ki-67 labeling index not statistically significant at the 0.05 level according to the Wilcoxon test. (C) Relationship between the magnitude of S6 inhibition and Ki-67 response in S2 tumor samples from rapamycin-treated patients. p-values were determined by Fisher Exact test for different thresholds of pS6 inhibition. (D) Ex vivo rapamycin response of short-term cultures derived from tumors with in vivo S6 response (patients 1 and 3) or resistance (patients 2 and 12). Shown are S6 and p85 (loading) immunoblots of whole cell lysates 8 h after treatment with vehicle, 0.3 nM rapamycin, and 3 nM rapamycin.
Figure 4. Induction of Akt Signaling in…
Figure 4. Induction of Akt Signaling in a Subset of Rapamycin-Treated Tumors
(A) Determination of Akt activation in tumor tissue during mTOR inhibitor therapy. The left panel is a cartoon illustrating the mTOR/S6K1 dependent feedback loop of the PI3k-Akt pathway. The right panel shows changes in phosphorylation of the Akt-substrate PRAS40 (threonine 246) during rapamycin therapy correspond to changes in Akt phosphorylation (serine 473). Tumors from patients 2 and 5 show an increase in pAkt and pPRAS40 immunostaining during rapamycin treatment, whereas the tumor from patient 11 shows a decrease in the same markers on rapamycin. (B) Changes in PRAS40 phosphorylation between S1 and S2 for 14/15 rapamycin patients. The horizontal line inside the box indicates the median value. The lower and upper border of the box corresponds to the 25th and 75th percentile, respectively. The whiskers extend to the 95% range. Paraffin blocks from patient 14 were not sufficient for quantification. N.S. indicates that the difference in pPRAS40 staining intensity not statistically significant at the 0.05 level according to the Wilcoxon test. (C) Kaplan Meier analysis illustrating the relationship between pPRAS40 induction and time-to-tumor progression.

References

    1. Arteaga CL, Baselga J. Tyrosine kinase inhibitors: why does the current process of clinical development not apply to them? Cancer Cell. 2004;5:525–531.
    1. Sarker D, Workman P. Pharmacodynamic biomarkers for molecular cancer therapeutics. Adv Cancer Res. 2007;96:213–268.
    1. Michaelis LC, Ratain MJ. Measuring response in a post-RECIST world: from black and white to shades of grey. Nat Rev Cancer. 2006;6:409–414.
    1. Smith IE, Walsh G, Skene A, Llombart A, Mayordomo JI, et al. A phase II placebo-controlled trial of neoadjuvant anastrozole alone or with gefitinib in early breast cancer. J Clin Oncol. 2007;25:3816–3822.
    1. Dowsett M, Smith IE, Ebbs SR, Dixon JM, Skene A, et al. Prognostic value of Ki67 expression after short-term presurgical endocrine therapy for primary breast cancer. J Natl Cancer Inst. 2007;99:167–170.
    1. Bjornsti MA, Houghton PJ. The TOR pathway: a target for cancer therapy. Nat Rev Cancer. 2004;4:335–348.
    1. Hudes G, Carducci M, Tomczak P, Dutcher J, Figlin R, et al. Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N Engl J Med. 2007;356:2271–2281.
    1. Sabatini DM. mTOR and cancer: insights into a complex relationship. Nat Rev Cancer. 2006;6:729–734.
    1. Neshat MS, Mellinghoff IK, Tran C, Stiles B, Thomas G, et al. Enhanced sensitivity of PTEN-deficient tumors to inhibition of FRAP/mTOR. Proc Natl Acad Sci U S A. 2001;98:10314–10319.
    1. Podsypanina K, Lee RT, Politis C, Hennessy I, Crane A, et al. An inhibitor of mTOR reduces neoplasia and normalizes p70/S6 kinase activity in Pten+/- mice. Proc Natl Acad Sci U S A. 2001;98:10320–10325.
    1. Yu K, Toral-Barza L, Discafani C, Zhang WG, Skotnicki J, et al. mTOR, a novel target in breast cancer: the effect of CCI-779, an mTOR inhibitor, in preclinical models of breast cancer. Endocr Relat Cancer. 2001;8:249–258.
    1. Shi Y, Gera J, Hu L, Hsu JH, Bookstein R, et al. Enhanced sensitivity of multiple myeloma cells containing PTEN mutations to CCI-779. Cancer Res. 2002;62:5027–5034.
    1. Majumder PK, Febbo PG, Bikoff R, Berger R, Xue Q, et al. mTOR inhibition reverses Akt-dependent prostate intraepithelial neoplasia through regulation of apoptotic and HIF-1-dependent pathways. Nat Med. 2004;10:594–601.
    1. Wendel HG, De Stanchina E, Fridman JS, Malina A, Ray S, et al. Survival signalling by Akt and eIF4E in oncogenesis and cancer therapy. Nature. 2004;428:332–337.
    1. Yilmaz OH, Valdez R, Theisen BK, Guo W, Ferguson DO, et al. Pten dependence distinguishes haematopoietic stem cells from leukaemia-initiating cells. Nature. 2006;441:475–482.
    1. Quinn JA, Desjardins A, Weingart J, Brem H, Dolan ME, et al. Phase I trial of temozolomide plus O6-benzylguanine for patients with recurrent or progressive malignant glioma. J Clin Oncol. 2005;23:7178–7187.
    1. Choe G, Horvath S, Cloughesy TF, Crosby K, Seligson D, et al. Analysis of the phosphatidylinositol 3′-kinase signaling pathway in glioblastoma patients in vivo. Cancer Res. 2003;63:2742–2746.
    1. Mellinghoff IK, Wang MY, Vivanco I, Haas-Kogan DA, Zhu S, et al. Molecular determinants of the response of glioblastomas to EGFR kinase inhibitors. N Engl J Med. 2005;353:2012–2024.
    1. Wullschleger S, Loewith R, Hall MN. TOR signaling in growth and metabolism. Cell. 2006;124:471–484.
    1. Pende M, Um SH, Mieulet V, Sticker M, Goss VL, et al. S6K1(-/-)/S6K2(-/-) mice exhibit perinatal lethality and rapamycin-sensitive 5′-terminal oligopyrimidine mRNA translation and reveal a mitogen-activated protein kinase-dependent S6 kinase pathway. Mol Cell Biol. 2004;24:3112–3124.
    1. Roux PP, Shahbazian D, Vu H, Holz MK, Cohen MS, et al. RAS/ERK signaling promotes site-specific ribosomal protein S6 phosphorylation via RSK and stimulates cap-dependent translation. J Biol Chem. 2007;282:14056–14064.
    1. Yatscoff R, LeGatt D, Keenan R, Chackowsky P. Blood distribution of rapamycin. Transplantation. 1993;56:1202–1206.
    1. Sun XJ, Goldberg JL, Qiao LY, Mitchell JJ. Insulin-induced insulin receptor substrate-1 degradation is mediated by the proteasome degradation pathway. Diabetes. 1999;48:1359–1364.
    1. Haruta T, Uno T, Kawahara J, Takano A, Egawa K, et al. A rapamycin-sensitive pathway down-regulates insulin signaling via phosphorylation and proteasomal degradation of insulin receptor substrate-1. Mol Endocrinol. 2000;14:783–794.
    1. Tremblay F, Marette A. Amino acid and insulin signaling via the mTOR/p70 S6 kinase pathway. A negative feedback mechanism leading to insulin resistance in skeletal muscle cells. J Biol Chem. 2001;276:38052–38060.
    1. Takano A, Usui I, Haruta T, Kawahara J, Uno T, et al. Mammalian target of rapamycin pathway regulates insulin signaling via subcellular redistribution of insulin receptor substrate 1 and integrates nutritional signals and metabolic signals of insulin. Mol Cell Biol. 2001;21:5050–5062.
    1. Sun SY, Rosenberg LM, Wang X, Zhou Z, Yue P, et al. Activation of Akt and eIF4E survival pathways by rapamycin-mediated mammalian target of rapamycin inhibition. Cancer Res. 2005;65:7052–7058.
    1. O'Reilly KE, Rojo F, She QB, Solit D, Mills GB, et al. mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. Cancer Res. 2006;66:1500–1508.
    1. Kovacina KS, Park GY, Bae SS, Guzzetta AW, Schaefer E, et al. Identification of a proline-rich Akt substrate as a 14-3-3 binding partner. J Biol Chem. 2003;278:10189–10194.
    1. Vander Haar E, Lee SI, Bandhakavi S, Griffin TJ, Kim DH. Insulin signalling to mTOR mediated by the Akt/PKB substrate PRAS40. Nat Cell Biol. 2007;9:316–323.
    1. Sancak Y, Thoreen CC, Peterson TR, Lindquist RA, Kang SA, et al. PRAS40 is an insulin-regulated inhibitor of the mTORC1 protein kinase. Mol Cell. 2007;25:903–915.
    1. Sarbassov DD, Guertin DA, Ali SM, Sabatini DM. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science. 2005;307:1098–1101.
    1. Sarbassov DD, Ali SM, Sengupta S, Sheen JH, Hsu PP, et al. Prolonged rapamycin treatment inhibits mTORC2 assembly and Akt/PKB. Mol Cell. 2006;22:159–168.
    1. Ichimura K, Ohgaki H, Kleihues P, Collins VP. Molecular pathogenesis of astrocytic tumours. J Neurooncol. 2004;70:137–160.
    1. Cui X, Kim HJ, Kuiatse I, Kim H, Brown PH, et al. Epidermal growth factor induces insulin receptor substrate-2 in breast cancer cells via c-Jun NH(2)-terminal kinase/activator protein-1 signaling to regulate cell migration. Cancer Res. 2006;66:5304–5313.
    1. Zhang H, Bajraszewski N, Wu E, Wang H, Moseman AP, et al. PDGFRs are critical for PI3K/Akt activation and negatively regulated by mTOR. J Clin Invest. 2007;117:730–738.
    1. Parsons R. Human cancer, PTEN and the PI-3 kinase pathway. Semin Cell Dev Biol. 2004;15:171–176.
    1. Sawyers CL. Will mTOR inhibitors make it as cancer drugs? Cancer Cell. 2003;4:343–348.
    1. Galanis E, Buckner JC, Maurer MJ, Kreisberg JI, Ballman K, et al. Phase II trial of temsirolimus (CCI-779) in recurrent glioblastoma multiforme: a North Central Cancer Treatment Group Study. J Clin Oncol. 2005;23:5294–5304.
    1. Chang SM, Kuhn J, Wen P, Greenberg H, Schiff D, et al. Phase I/pharmacokinetic study of CCI-779 in patients with recurrent malignant glioma on enzyme-inducing antiepileptic drugs. Invest New Drugs. 2004;22:427–435.
    1. Chang SM, Wen P, Cloughesy T, Greenberg H, Schiff D, et al. Phase II study of CCI-779 in patients with recurrent glioblastoma multiforme. Invest New Drugs. 2005;23:357–361.
    1. Guertin DA, Sabatini DM. Defining the role of mTOR in cancer. Cancer Cell. 2007;12:9–22.
    1. Sawyers CL. Making progress through molecular attacks on cancer. Cold Spring Harb Symp Quant Biol. 2005;70:479–482.
    1. Vredenburgh JJ, Desjardins A, Herndon JE, 2nd, Dowell JM, Reardon DA, et al. Phase II trial of bevacizumab and irinotecan in recurrent malignant glioma. Clin Cancer Res. 2007;13:1253–1259.
    1. Waldherr C, Mellinghoff IK, Tran C, Halpern BS, Rozengurt N, et al. Monitoring antiproliferative responses to kinase inhibitor therapy in mice with 3′-deoxy-3′-18F-fluorothymidine PET. J Nucl Med. 2005;46:114–120.
    1. Bianco R, Shin I, Ritter CA, Yakes FM, Basso A, et al. Loss of PTEN/MMAC1/TEP in EGF receptor-expressing tumor cells counteracts the antitumor action of EGFR tyrosine kinase inhibitors. Oncogene. 2003;22:2812–2822.
    1. Wang MY, Lu KV, Zhu S, Dia EQ, Vivanco I, et al. Mammalian target of rapamycin inhibition promotes response to epidermal growth factor receptor kinase inhibitors in PTEN-deficient and PTEN-intact glioblastoma cells. Cancer Res. 2006;66:7864–7869.
    1. Li D, Shimamura T, Ji H, Chen L, Haringsma HJ, et al. Bronchial and peripheral murine lung carcinomas induced by T790M-L858R mutant EGFR respond to HKI-272 and rapamycin combination therapy. Cancer Cell. 2007;12:81–93.
    1. Fan QW, Knight ZA, Goldenberg DD, Yu W, Mostov KE, et al. A dual PI3 kinase/mTOR inhibitor reveals emergent efficacy in glioma. Cancer Cell. 2006;9:341–349.
    1. Faivre S, Kroemer G, Raymond E. Current development of mTOR inhibitors as anticancer agents. Nat Rev Drug Discov. 2006;5:671–688.
    1. Thomas GV, Tran C, Mellinghoff IK, Welsbie DS, Chan E, et al. Hypoxia-inducible factor determines sensitivity to inhibitors of mTOR in kidney cancer. Nat Med. 2006;12:122–127.
    1. Kummar S, Kinders R, Rubinstein L, Parchment RE, Murgo AJ, et al. Compressing drug development timelines in oncology using phase '0' trials. Nat Rev Cancer. 2007;7:131–139.

Source: PubMed

3
S'abonner