Targeting the interleukin-11 receptor α in metastatic prostate cancer: A first-in-man study

Renata Pasqualini, Randall E Millikan, Dawn R Christianson, Marina Cardó-Vila, Wouter H P Driessen, Ricardo J Giordano, Amin Hajitou, Anh G Hoang, Sijin Wen, Kirstin F Barnhart, Wallace B Baze, Valerie D Marcott, David H Hawke, Kim-Anh Do, Nora M Navone, Eleni Efstathiou, Patricia Troncoso, Roy R Lobb, Christopher J Logothetis, Wadih Arap, Renata Pasqualini, Randall E Millikan, Dawn R Christianson, Marina Cardó-Vila, Wouter H P Driessen, Ricardo J Giordano, Amin Hajitou, Anh G Hoang, Sijin Wen, Kirstin F Barnhart, Wallace B Baze, Valerie D Marcott, David H Hawke, Kim-Anh Do, Nora M Navone, Eleni Efstathiou, Patricia Troncoso, Roy R Lobb, Christopher J Logothetis, Wadih Arap

Abstract

Background: Receptors in tumor blood vessels are attractive targets for ligand-directed drug discovery and development. The authors have worked systematically to map human endothelial receptors ("vascular zip codes") within tumors through direct peptide library selection in cancer patients. Previously, they selected a ligand-binding motif to the interleukin-11 receptor alpha (IL-11Rα) in the human vasculature.

Methods: The authors generated a ligand-directed, peptidomimetic drug (bone metastasis-targeting peptidomimetic-11 [BMTP-11]) for IL-11Rα-based human tumor vascular targeting. Preclinical studies (efficacy/toxicity) included evaluating BMTP-11 in prostate cancer xenograft models, drug localization, targeted apoptotic effects, pharmacokinetic/pharmacodynamic analyses, and dose-range determination, including formal (good laboratory practice) toxicity across rodent and nonhuman primate species. The initial BMTP-11 clinical development also is reported based on a single-institution, open-label, first-in-class, first-in-man trial (National Clinical Trials number NCT00872157) in patients with metastatic, castrate-resistant prostate cancer.

Results: BMTP-11 was preclinically promising and, thus, was chosen for clinical development in patients. Limited numbers of patients who had castrate-resistant prostate cancer with osteoblastic bone metastases were enrolled into a phase 0 trial with biology-driven endpoints. The authors demonstrated biopsy-verified localization of BMTP-11 to tumors in the bone marrow and drug-induced apoptosis in all patients. Moreover, the maximum tolerated dose was identified on a weekly schedule (20-30 mg/m(2) ). Finally, a renal dose-limiting toxicity was determined, namely, dose-dependent, reversible nephrotoxicity with proteinuria and casts involving increased serum creatinine.

Conclusions: These biologic endpoints establish BMTP-11 as a targeted drug candidate in metastatic, castrate-resistant prostate cancer. Within a larger discovery context, the current findings indicate that functional tumor vascular ligand-receptor targeting systems may be identified through direct combinatorial selection of peptide libraries in cancer patients.

Keywords: bone metastasis-targeting peptidomimetic-11; clinical trial; interleukin-11 receptor α; prostate cancer; vascular targeting.

© 2015 American Cancer Society.

Figures

Figure 1
Figure 1
The efficacy of bone metastasis-targeting peptidomimetic-11 (BMTP-11) was investigated in tumor-bearing, immunodeficient mouse models of prostate cancer. (A) DU145-derived, tumor-bearing nu/nu (nude) mice were treated with either BMTP-11 (10 mg/kg; n = 10) or saline (control; n = 10) intravenously through the tail vein once weekly for 4 weeks. Tumor growth was serially monitored, and tumor volumes were measured over time. Data shown are the means ± standard error of the means (SEM). (B) In a mixed-effects model, there was a significant difference over time in the average percentage change from baseline for tumor volume in the treated animals versus the control animals (P < .0001). (C) Tumor volumes on day zero (pretreatment) and on day 28 (post-treatment) are illustrated. (D) LNCaP-derived, tumor-bearing nude mice were treated with either BMTP-11 (15 mg/kg; n = 4) or saline (control; n = 4) intravenously 3 times weekly for 2 weeks. Tumor growth was serially monitored, and tumor volumes were measured over time. Data shown are the means ± SEM. (E) In a mixed-effects model, there was a significant difference over time in the average percentage change from baseline for tumor volume in the treated animals versus the control animals (P < .0001). (F) Tumor volumes on day zero (pretreatment) and on day 28 (post-treatment) are illustrated. (G) (Top Right) The expression of interleukin-11 receptor alpha (IL-11Rα) is observed in MDA-PCA-118b–implanted tumors (T) relative to (Bottom Right) an immunoglobulin G (IgG) isotype-negative control. The asterisk indicates bone. (Top Left) Anti-CD31 (cluster of differentiation 31 [also called platelet endothelial cell adhesion molecule]) and (Bottom Left) hematoxylin and eosin (H&E) stains also are shown. (H) Severe combined immunodeficient (SCID) mice bearing MDA-PCA-118b–implanted tumors were divided into equal pretreatment cohorts using x-ray imaging, and (I) the bone-to-tumor stroma ratio was quantified in each group using relative mean gray-scale analysis before treatment was initiated. (J) MDA-PCA-118b tumor-bearing SCID mice were treated with either BMTP-11 (10 mg/kg; n = 5) or saline (control; n = 5) intravenously through the tail vein once weekly for 3 weeks. Tumor growth was serially monitored, and volumes were measured over time. Data shown are the means ± SEM. (K) In a mixed-effects model, there was a significant difference over time in the average percentage change from baseline for tumor volume in the treated animals versus the control animals (P < .0001). (L) Tumor volumes on day zero (pretreatment) and on day 21 (post-treatment) are illustrated.
Figure 2
Figure 2
Bone metastasis-targeting peptidomimetic-11 (BMTP-11) pharmacokinetics and metabolites in nonhuman primates are illustrated, including (A) the BMTP-11 standard curve in the plasma of cynomolgus monkeys and (B) the BMTP-11 plasma concentration time curves after intravenous infusion of BMTP-11 (at doses of 1 mg/kg, 3 mg/kg, and 9 mg/kg) for 2 hours. (C) This is a dose-linearity plot for the 3 different BMTP-11 doses. AUC indicates the area under the plasma concentration-time curve. (D) Observed values and model prediction fitted to a 1-compartment, open-body model (constructed using Phoenix WinNonlin [Certara LP, Princeton, NJ]) are shown. (E) Identified metabolites of BMTP-11 in plasma and their predicted amino acid sequence were based on molecular mass. The boxed top sequence indicates the parent drug, BMTP-11 (peak 2483 [p2483]). S-S indicates disulfide bonds.
Figure 3
Figure 3
Photomicrographs illustrate the expression of interleukin-11 receptor alpha (IL-11Rα) and negative control (immunoglobulin G [IgG] isotype) in human bone marrow. Each patient from the treated cohort (n = 6) underwent a bone marrow biopsy before receiving bone metastasis-targeting peptidomimetic-11.
Figure 4
Figure 4
Bone metastasis-targeting peptidomimetic-11 (BMTP-11) targets bone metastasis in patients with castrate-resistant prostate cancer. (A) This is the scheme for the first-in-man clinical trial design. GLP indicates good laboratory practice; IVPB, intravenous piggy-back infusion. (B) BMTP-11 detection by mass spectrometry is illustrated, including (Left) the cleaved proapoptotic domain and (Right) the parent BMTP-11 compound. (C) BMTP-11 immunocolocalization and (D) the terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) assay are shown before and after treatment with BMTP-11 (18 mg/m2) in representative patient samples.
Figure 5
Figure 5
Serial changes in serum tumor markers are illustrated in a patient before and after treatment with bone metastasis-targeting peptidomimetic-11 (BMTP-11). Serum prostate-specific antigen (PSA), alkaline phosphatase (Alk. Phos.), and lactate dehydrogenase (LDH) levels are indicated. 5-FU indicates infusional 5-fluorouracil plus concomitant external-beam radiotherapy; CVD, intravenous cyclophosphamide and vincristine plus oral dexamethasone; DES/Dex, oral diethylstilbestrol plus oral dexamethasone; G/S, gemcitabine intravenously plus oral sunitinib; KA/VE, oral ketoconazole plus doxorubicin intravenously alternating on a weekly basis with oral estramustine plus vinblastine intravenously; T/C, paclitaxel plus carboplatin intravenously every 3 weeks; T/S, docetaxel intravenously plus oral sunitinib; VP-16/CDDP, etoposide plus cisplatin intravenously every 3 weeks.

References

    1. Pasqualini R, Moeller BJ, Arap W. Leveraging molecular heterogeneity of the vascular endothelium for targeted drug delivery and imaging. Semin Thromb Hemost. 2010;36:343–351.
    1. Sergeeva A, Kolonin MG, Molldrem JJ, Pasqualini R, Arap W. Display technologies: application for the discovery of drug and gene delivery agents. Adv Drug Deliv Rev. 2006;58:1622–1654.
    1. Ozawa MG, Zurita AJ, Dias-Neto E, et al. Beyond receptor expression levels: the relevance of target accessibility in ligand-directed pharmacodelivery systems. Trends Cardiovasc Med. 2008;18:126–132.
    1. Arap W, Kolonin MG, Trepel M, et al. Steps toward mapping the human vasculature by phage display. Nat Med. 2002;8:121–127.
    1. Pentz RD, Flamm AL, Pasqualini R, Logothetis CJ, Arap W. Revisiting ethical guidelines for research with terminal wean and brain-dead participants. Hastings Cent Rep. 2003;33:20–26.
    1. Pentz RD, Cohen CB, Wicclair M, et al. Ethics guidelines for research with the recently dead. Nat Med. 2005;11:1145–1149.
    1. Staquicini FI, Cardó-Vila M, Kolonin MG, et al. Vascular ligand-receptor mapping by direct combinatorial selection in cancer patients. Proc Natl Acad Sci U S A. 2011;108:18637–18642.
    1. Zurita AJ, Troncoso P, Cardó-Vila M, Logothetis CJ, Pasqualini R, Arap W. Combinatorial screenings in patients: the interleukin-11 receptor alpha as a candidate target in the progression of human prostate cancer. Cancer Res. 2004;64:435–439.
    1. Cardó-Vila M, Zurita AJ, Giordano RJ, et al. A ligand peptide motif selected from a cancer patient is a receptor-interacting site within human interleukin-11 [serial online] PLoS One. 2008;3:e3452.
    1. Gu C, Liu L, He Y, Jiang J, Yang Z, Wu Q. The binding characteristics of a cyclic nonapeptide, c(CGRRAGGSC), in LNCaP human prostate cancer cells. Oncol Lett. 2012;4:443–449.
    1. Wang W, Ke S, Kwon S, et al. A new optical and nuclear dual-labeled imaging agent targeting interleukin 11 receptor alpha-chain. Bioconjug Chem. 2007;18:397–402.
    1. Ellerby HM, Arap W, Ellerby LM, et al. Anti-cancer activity of targeted pro-apoptotic peptides. Nat Med. 1999;5:1032–1038.
    1. Arap W, Haedicke W, Bernasconi M, et al. Targeting the prostate for destruction through a vascular address. Proc Natl Acad Sci USA. 2002;99:1527–1531.
    1. Arap MA, Lahdenranta J, Mintz PJ, et al. Cell surface expression of the stress response chaperone GRP78 enables tumor targeting by circulating ligands. Cancer Cell. 2004;6:275–284.
    1. Kolonin MG, Saha PK, Chan L, Pasqualini R, Arap W. Reversal of obesity by targeted ablation of adipose tissue. Nat Med. 2004;10:625–632.
    1. Lewis VO, Ozawa MG, Deavers MT, et al. The interleukin-11 receptor alpha as a candidate ligand-directed target in osteosarcoma: consistent data from cell lines, orthotopic models, and human tumor samples. Cancer Res. 2009;69:1995–1999.
    1. Barnhart KF, Christianson DR, Hanley PW, et al. A peptidomimetic targeting fat causes weight loss and improved insulin resistance in obese monkeys [serial online] Sci Transl Med. 2011;3:108ra112.
    1. Li ZG, Mathew P, Yang J, et al. Androgen receptor-negative human prostate cancer cells induce osteogenesis through FGF9-mediated mechanisms. J Clin Invest. 2008;118:2697–2710.
    1. US Food and Drug Administration. Guidance for Industry Safety Testing of Drug Metabolites. Available at: . Accessed February 26, 2015.
    1. US Food and Drug Administration. Guidance for Industry Estimating the Maximum Safe Starting Dose in Initial Clinical Trials for Therapeutics in Adult Healthy Volunteers. Available at: . Accessed February 26, 2015.
    1. Cancer Therapy Evaluation Program, National Cancer Institute. Protocol Development: CTCAE v4.0 Open Comment Period. Available at: . Accessed February 26, 2015.
    1. Scher HI, Halabi S, Tannock I, et al. Design and endpoints of clinical trials for patients with progressive prostate cancer and castrate levels of testosterone: recommendations of the Prostate Cancer Clinical Trials Working Group. J Clin Oncol. 2008;26:1148–1159.
    1. Morris MJ, Pandit-Taskar N, Carrasquillo J, et al. Phase I study of Samarium-153 lexidronam with docetaxel in castration-resistant metastatic prostate cancer. J Clin Oncol. 2009;27:2436–2442.
    1. Tu SM, Mathew P, Wong FC, Jones D, Johnson MM, Logothetis CJ. Phase I study of concurrent weekly docetaxel and repeated Samarium-153 lexidronam in patients with castration-resistant metastatic prostate cancer. J Clin Oncol. 2009;27:3319–3324.
    1. Tran C, Ouk S, Clegg NJ, et al. Development of a second-generation antiandrogen for treatment of advanced prostate cancer. Science. 2009;324:787–790.
    1. Scher HI, Fizazi K, Saad F, et al. Increased survival with enzalutamide in prostate cancer after chemotherapy. N Engl J Med. 2012;367:1187–1197.
    1. de Bono JS, Logothetis CJ, Molina A, et al. Abiraterone and increased survival in metastatic prostate cancer. N Engl J Med. 2011;364:1995–2005.
    1. Reid AH, Attard G, Danila DC, et al. Significant and sustained antitumor activity in post-docetaxel, castration-resistant prostate cancer with the CYP17 inhibitor abiraterone acetate. J Clin Oncol. 2010;28:1489–1495.
    1. Karan D, Holzbeierlein JM, Van Veldhuizen P, Thrasher JB. Cancer immunotherapy: a paradigm shift for prostate cancer treatment. Nat Rev Urol. 2012;9:376–385.
    1. de Bono JS, Oudard S, Ozguroglu M, et al. Prednisone plus cabazitaxel or mitoxantrone for metastatic castration-resistant prostate cancer progressing after docetaxel treatment: a randomised open-label trial. Lancet. 2010;376:1147–1154.

Source: PubMed

3
Iratkozz fel