Improvement of Pharmacokinetic Profile of TRAIL via Trimer-Tag Enhances its Antitumor Activity in vivo

Haipeng Liu, Danmei Su, Jinlong Zhang, Shuaishuai Ge, Youwei Li, Fei Wang, Michel Gravel, Anne Roulston, Qin Song, Wei Xu, Joshua G Liang, Gordon Shore, Xiaodong Wang, Peng Liang, Haipeng Liu, Danmei Su, Jinlong Zhang, Shuaishuai Ge, Youwei Li, Fei Wang, Michel Gravel, Anne Roulston, Qin Song, Wei Xu, Joshua G Liang, Gordon Shore, Xiaodong Wang, Peng Liang

Abstract

TNF-related apoptosis-inducing ligand (TRAIL/Apo2L) has long been considered a tantalizing target for cancer therapy because it mediates activation of the extrinsic apoptosis pathway in a tumor-specific manner by binding to and trimerizing its functional receptors DR4 or DR5. Despite initial promise, both recombinant human TRAIL (native TRAIL) and dimeric DR4/DR5 agonist monoclonal antibodies (mAbs) failed in multiple human clinical trials. Here we show that in-frame fusion of human C-propeptide of α1(I) collagen (Trimer-Tag) to the C-terminus of mature human TRAIL leads to a disulfide bond-linked homotrimer which can be expressed at high levels as a secreted protein from CHO cells. The resulting TRAIL-Trimer not only retains similar bioactivity and receptor binding kinetics as native TRAIL in vitro which are 4-5 orders of magnitude superior to that of dimeric TRAIL-Fc, but also manifests more favorable pharmacokinetic and antitumor pharmacodynamic profiles in vivo than that of native TRAIL. Taken together, this work provides direct evidence for the in vivo antitumor efficacy of TRAIL being proportional to systemic drug exposure and suggests that the previous clinical failures may have been due to rapid systemic clearance of native TRAIL and poor apoptosis-inducing potency of dimeric agonist mAbs despite their long serum half-lives.

Conflict of interest statement

P. Liang, X. Wang and J.G. Liang have ownership interest in Clover Biopharmaceuticals. P. Liang also has ownership interest in GenHunter Corporation. The other authors disclose no financial conflicts of interest.

Figures

Figure 1
Figure 1
Structures of different TRAIL polypeptides. (a) Three recombinant soluble forms of human TRAIL have been used in this study: a native TRAIL comprised of the extracellular domain of TRAIL, a dimeric TRAIL-Fc comprised of extracellular TRAIL domain fused to human IgG1 Fc domain, and a TRAIL-Trimer comprised of extracellular TRAIL domain fused to Trimer-Tag. Amino acid sequence lengths are shown for each protein and domain respectively. (b) Theoretical molecular weights (kDa) of both monomeric and multimeric forms of native TRAIL, TRAIL-Fc and TRAIL-Trimer, respectively. Native TRAIL associates into a non-covalently-linked homotrimer, TRAIL-Fc forms a covalently-linked homodimer, and TRAIL-Trimer forms a covalently-linked homotrimer.
Figure 2
Figure 2
High-level expression and purification of TRAIL-Trimer fusion protein. (a) 10% SDS-PAGE analysis of TRAIL-Trimer expression from a fed-batch serum-free cell culture in the bioreactor. 10 µL of cell-free conditioned medium from Day 6 to Day 13 were analyzed under non-reducing condition followed by Coomassie Blue staining. (b) Bioassay analysis of TRAIL-Trimer production in conditioned medium from Day 5 to Day 13. (c) SDS-PAGE and western blot analysis of purified TRAIL-Trimer under either non-reducing or reducing conditions. 2 µg of purified protein was analyzed by a 10% SDS-PAGE and stained with Coomassie Blue. 0.2 µg of purified protein was analyzed by western blot using monoclonal antibody against TRAIL-domain and Trimer-domain, respectively.
Figure 3
Figure 3
Purity evaluation of TRAIL-Trimer, TRAIL-Fc and native TRAIL. SDS-PAGE analysis of purified TRAIL-Trimer, TRAIL-Fc and native TRAIL under either non-reducing or reducing conditions. 2 µg of purified protein was analyzed by 10% or 15% SDS-PAGE, respectively, and stained with Coomassie Blue.
Figure 4
Figure 4
Bioactivity and affinity with DR5-Fc comparison of TRAIL-Trimer, TRAIL-Fc and native TRAIL. (a) Bioactivity detection of TRAIL-Trimer, TRAIL-Fc and native TRAIL. The IC50 value of TRAIL-Trimer, TRAIL-Fc and native TRAIL were assessed using a TRAIL sensitive cell line COLO205 on a Real-Time Cell Analysis (RTCA) system. The IC50 value was obtained according to the dose-response cell index (CI) curve. The IC50 value of TRAIL-Trimer, TRAIL-Fc and native TRAIL is 23.2 ng/mL, 260.8 µg/mL, and 6.7 ng/mL, respectively. The bottom graph displays IC50 values when molar ratio-adjusted based on theoretical molecular weights for each protein. (b) Kinetic parameters of TRAIL-Trimer, TRAIL-Fc and native TRAIL binding to the soluble DR5-Fc fusion protein was assessed by biolayer interferometry measurements. The Super Streptavidin biosensor tips of the ForteBio Octet RED 96 were coated with biotinylated DR5-Fc. The biosensor tips were dipped in increasing concentrations gradient of the same molarity (15.4 nM–123.5 nM) to measure binding of TRAIL-Trimer, TRAIL-Fc and native TRAIL to DR5-Fc and subsequently moved to wells containing buffer (PBS) to measure dissociation rates. Detailed results are shown in Table 1.
Figure 5
Figure 5
Pharmacokinetic profile of TRAIL-Trimer and native TRAIL determined by western blot. Mice were injected intravenously with equimolar amounts of TRAIL-Trimer (40 mg/kg) or native TRAIL (15 mg/kg) (n = 3 for each protein). (a,c) Relative serum concentrations of both TRAIL-Trimer (a) and native TRAIL (c) were evaluated at periodic intervals by western blotting. TRAIL proteins were immunoblotted with an antibody against human TRAIL, and visualized by chemiluminescence. The images were acquired using the ChemiDoc Touch Imaging System (Bio-Rad) and the quantification of the amount of TRAIL proteins present in each sample was done using Image Lab software (Bio-Rad). (b,d) Estimated half-lives of TRAIL-Trimer (b) and native TRAIL (d) were calculated using GraphPad Prism v5.04. Error bars represent ± SEM.
Figure 6
Figure 6
In vivo antitumor activities of TRAIL-Trimer and native TRAIL. (a) Nude mice with established COLO205 xenografts were given TRAIL-Trimer (30, 50, or 80 mg/kg/day) or vehicle as an i.v. bolus for 5 consecutive days (n = 6/group). Results shown are group mean (±S.D.). (b) Nude mice with established COLO205 xenografts were given the same molar concentration of TRAIL-Trimer and native TRAIL. Results shown are group mean (±S.D.).

References

    1. Ashkenazi A, Dixit VM. Death receptors: signaling and modulation. Science. 1998;281:1305–1308. doi: 10.1126/science.281.5381.1305.
    1. Wang S, El-Deiry W. S. TRAIL and apoptosis induction by TNF-family death receptors. Oncogene. 2003;22:8628–8633. doi: 10.1038/sj.onc.1207232.
    1. Younes M, Georgakis GV, Rahmani M, Beer D, Younes A. Functional expression of TRAIL receptors TRAIL-R1 and TRAIL-R2 in esophageal adenocarcinoma. Eur J Cancer. 2006;42:542–547. doi: 10.1016/j.ejca.2005.11.013.
    1. Koornstra JJ, et al. Expression of TRAIL (TNF-related apoptosis-inducing ligand) and its receptors in normal colonic mucosa, adenomas and carcinomas. J Pathol. 2003;200:327–335. doi: 10.1002/path.1364.
    1. Wiley SR, et al. Identification and characterization of a new member of the TNF family that induces apoptosis. Immunity. 1995;3:673–682. doi: 10.1016/1074-7613(95)90057-8.
    1. Pitti RM, et al. Induction of apoptosis by Apo-2 Ligand, a new member of the tumor necrosis factor receptor family. J Biol Chem. 1996;271:12687–12690. doi: 10.1074/jbc.271.22.12687.
    1. Walczak H, Krammer PH. The CD95 (APO-1/Fas) and the TRAIL (APO-2L) apoptosis systems. Ex Cell Res. 2000;256:58–66. doi: 10.1006/excr.2000.4840.
    1. Dickens LS, et al. A death effector domain chain DISC model reveals a crucial role for caspase-8 chain assembly in mediating apoptotic cell death. Mol Cell. 2012;47:291–305. doi: 10.1016/j.molcel.2012.05.004.
    1. Lemke J, von Karstedt S, Zinngrebe J, Walczak H. Getting TRAIL back on track for cancer therapy. Cell Death Differ. 2014;21:1350–1364. doi: 10.1038/cdd.2014.81.
    1. Ravi R, et al. Regulation of death receptor expression and TRAIL/Apo2L-induced apoptosis by NF-κB. Nat Cell Biol. 2001;3:409–416. doi: 10.1038/35070096.
    1. Fulda S, Debatin KM. Extrinsic versus intµrinsic apoptosis pathways in anticancer chemotherapy. Oncogene. 2006;25:4798–4811. doi: 10.1038/sj.onc.1209608.
    1. Pukac L, et al. HGS-ETR1, a fully human TRAIL-receptor 1 monoclonal antibody, induces cell death in multiple tumour types in vitro and in vivo. Br J Cancer. 2005;92:1430–1441. doi: 10.1038/sj.bjc.6602487.
    1. Marini P, et al. Combined treatment of colorectal tumours with agonistic TRAIL receptor antibodies HGS-ETR1 and HGS-ETR2 and radiotherapy: enhanced effects in vitro and dose-dependent growth delay in vivo. Oncogene. 2006;25:5145–5154. doi: 10.1038/sj.onc.1209516.
    1. Wilson NS, et al. An Fcgamma receptor-dependent mechanism drives antibody-mediated target-receptor signaling in cancer cells. Cancer Cell. 2011;19:101–113. doi: 10.1016/j.ccr.2010.11.012.
    1. Kelley SK, et al. Preclinical studies to predict the disposition of Apo2L/Tumor necrosis factor-related apoptosis-inducing ligand in humans: characterization of in vivo efficacy, pharmacokinetics, and safety. JPET. 2001;299:31–38.
    1. Lawrence D, et al. Differential hepatocyte toxicity of recombinant Apo2L/TRAIL versions. Nat Med. 2001;7:383–385. doi: 10.1038/86397.
    1. Walczak K, et al. Tumoricidal activity of tumor necrosis factor-related apoptosis-inducing ligand in vivo. Nat Med. 1999;5:157–163. doi: 10.1038/5517.
    1. Wang Y, et al. Synthetic lethal targeting of MYC by activation of the DR5 death receptor pathway. Cancer Cell. 2004;5:501–512. doi: 10.1016/S1535-6108(04)00113-8.
    1. Ashley DM, et al. In vitro sensitivity testing of minimally passaged and uncultured gliomas with TRAIL and/or chemotherapy drugs. Br J Cancer. 2008;99:294–304. doi: 10.1038/sj.bjc.6604459.
    1. Bae S, et al. Doxorubicin-loaded human serum albumin nanoparticles surface-modified with TNF-related apoptosis-inducing ligand and transferrin for targeting multiple tumor types. Biomaterials. 2012;33:1536–1546. doi: 10.1016/j.biomaterials.2011.10.050.
    1. Mitchell MJ, Wayne E, Rana K, Schaffer CB, King MR. TRAIL-coated leukocytes that kill cancer cells in the circulation. Proc Natl Acad Sci USA. 2014;111:930–935. doi: 10.1073/pnas.1316312111.
    1. Xu H, Wang KQ, Deng YH, Chen DW. Effects of cleavable PEG-cholesterol derivatives on the accelerated blood clearance of PEGylated liposomes. Biomaterials. 2010;31:4757–4763. doi: 10.1016/j.biomaterials.2010.02.049.
    1. Kim TH, et al. PEGylated TNF-related apoptosis-inducing ligand (TRAIL)-loaded sustained release PLGA microspheres for enhanced stability and antitumor activity. J Control Release. 2011;150:63–69. doi: 10.1016/j.jconrel.2010.10.037.
    1. Lim SM, et al. Improved biological half-life and anti-tumor activity of TNF-related apoptosis-inducing ligand (TRAIL) using PEGexposed nanoparticles. Biomaterials. 2011;32:3538–3546. doi: 10.1016/j.biomaterials.2011.01.054.
    1. Won EY, et al. The structure of the trimer of human 4-1BB ligand is unique among members of the tumor necrosis factor superfamily. J Biol Chem. 2010;285:9202–9210. doi: 10.1074/jbc.M109.084442.
    1. Xiang H, Nguyen CB, Kelley SK, Dybdal N, Escandón E. Tissue distribution, stability, and pharmacokinetics of Apo2 ligand/tumor necrosis factor-related apoptosis-inducing ligand in human colon carcinoma COLO205 tumor-bearing nude mice. Drug Metab Dispos. 2004;32:1230–1238. doi: 10.1124/dmd.104.000323.
    1. Melkko J, Niemi S, Risteli L, Risteli J. Radioimmunoassay of the carboxyterminal propeptide of human type I procollagen. Clin Chem. 1990;36:1328–1332.
    1. Sorva A, et al. Familial high serum concentrations of the carboxyl-terminal propeptide of type I procollagen. Clin Chem. 1994;40:1591–1593.
    1. Bernocco S, et al. Biophysical characterization of the C-propeptide trimer from human procollagen III reveals a tri-lobed structure. J Biol Chem. 2001;276:48930–48936. doi: 10.1074/jbc.M108611200.
    1. Bourhis JM, et al. Structural Basis of Fibrillar Collagen Trimerization and Related Genetic Disorders. Nat Struct Mol Biol. 2012;19:1031–1036. doi: 10.1038/nsmb.2389.
    1. Yang X, Farzan M, Wyatt R, Sodroski J. Characterization of stable, soluble trimers containing complete ectodomains of human immunodeficiency virus type 1 envelope glycoproteins. J Virol. 2000;74:5716–5725. doi: 10.1128/JVI.74.12.5716-5725.2000.
    1. Frank S, et al. Stabilization of short collagen-like triple helices by protein engineering. J Mol Biol. 2001;308:1081–1089. doi: 10.1006/jmbi.2001.4644.
    1. Chen B, et al. A chimeric protein of simian immunodeficiency virus envelope glycoprotein gp140 and Escherichia coli aspartate transcarbamoylase. J Virol. 2004;78:4508–4516. doi: 10.1128/JVI.78.9.4508-4516.2004.
    1. Harding FA, Stickler MM, Razo J, DuBridge RB. The immunogenicity of humanized and fully human antibodies. MAbs. 2010;2:256–265. doi: 10.4161/mabs.2.3.11641.
    1. Holash J, et al. VEGF-trap: a VEGF blocker with potent antitumor effects. Proc Natl Acad Sci USA. 2002;99:11393–11398. doi: 10.1073/pnas.172398299.
    1. Genovese MC, et al. Abatacept for rheumatoid arthritis refractory to tumor necrosis factor α inhibition. N Engl J Med. 2005;353:1114–1123. doi: 10.1056/NEJMoa050524.
    1. Kareskog L, et al. Therapeutic effect of the combination of etanercept and methotrexate compared with each treatment alone in patients with rheumatoid arthritis: double-blind randomised controlled trial. The Lancet. 2004;363:675–681. doi: 10.1016/S0140-6736(04)15640-7.
    1. Haynes NM, et al. CD11c+ dendritic cells and B cells contribute to the tumoricidal activity of anti-DR5 antibody therapy in established tumors. J Immunol. 2010;185:532–541. doi: 10.4049/jimmunol.0903624.
    1. Clark R, et al. Long-acting growth hormones produced by conjugation with polyethylene glycol. J Biol Chem. 1996;271:21969–21977. doi: 10.1074/jbc.271.36.21969.
    1. Zhang L, Fang B. Mechanisms of resistance to TRAIL-induced apoptosis in cancer. Cancer Gene Ther. 2005;12:228–237. doi: 10.1038/sj.cgt.7700792.
    1. Pan G, et al. An antagonist decoy receptor and a death domain-containing receptor for TRAIL. Science. 1997;277:815–818. doi: 10.1126/science.277.5327.815.
    1. Sheridan JP, et al. Control of TRAIL-induced apoptosis by a family of signaling and decoy receptors. Science. 1997;277:818–821. doi: 10.1126/science.277.5327.818.
    1. Ashkenazi A, Dixit VM. Apoptosis control by death and decoy receptors. Curr Opin Cell Biol. 1999;11:255–260. doi: 10.1016/S0955-0674(99)80034-9.
    1. Merino D, et al. Differential inhibition of TRAIL-mediated DR5-DISC formation by decoy receptors 1 and 2. Mol Cell Biol. 2006;26:7046–7055. doi: 10.1128/MCB.00520-06.
    1. Morizot A, et al. Chemotherapy overcomes TRAIL-R4-mediated TRAIL resistance at the DISC level. Cell Death Differ. 2011;18:700–711. doi: 10.1038/cdd.2010.144.
    1. LeBlanc H, et al. Tumor-cell resistance to death receptor-induced apoptosis through mutational inactivation of the proapoptotic Bcl-2 homolog Bax. Nat Med. 2002;8:274–281. doi: 10.1038/nm0302-274.
    1. Roy N, Deveraux QL, Takahashi R, Salvesen GS, Reed JC. The c-IAP-1 and c-IAP-2 proteins are direct inhibitors of specific caspases. EMBO J. 1997;16:6914–6925. doi: 10.1093/emboj/16.23.6914.
    1. Deveraux QL, Reed JC. IAP family proteins–suppressors of apoptosis. Genes Dev. 1999;13:239–252. doi: 10.1101/gad.13.3.239.
    1. Seymour JF, et al. Bcl-2 inhibitor ABT-199 (GDC-0199) monotherapy shows anti-tumor activity including complete remissions in high-risk relapsed/refractory (R/R) chronic lymphocytic leukemia (CLL) and small lymphocytic lymphoma (SLL) Blood. 2013;122:872. doi: 10.1182/blood-2013-05-501569.
    1. Bai L, Smith DC, Wang S. Small-molecule SMAC mimetics as new cancer therapeutics. Pharmacol Ther. 2014;144:82–95. doi: 10.1016/j.pharmthera.2014.05.007.
    1. Li L, et al. A small molecule Smac mimic potentiates TRAIL- and TNFalpha-mediated cell death. Science. 2004;305:1471–1474. doi: 10.1126/science.1098231.
    1. Fulda S, Meyer E, Debatin KM. Inhibition of TRAIL-induced apoptosis by Bcl-2 overexpression. Oncogene. 2002;21:2283–2294. doi: 10.1038/sj.onc.1205258.
    1. Kim KJ, et al. Two-promoter vector is highly efficient for overproduction of protein complexes. Protein Sci. 2004;13:1698–1703. doi: 10.1110/ps.04644504.
    1. Ke N, Wang X, Xu X, Abassi YA. The xCELLigence system for real-time and label-free monitoring of cell viability. Methods Mol Biol. 2011;740:33–43. doi: 10.1007/978-1-61779-108-6_6.
    1. Atienza JM, Zhu J, Wang X, Xu X, Abassi Y. Dynamic monitoring of cell adhesion and spreading on microelectronic sensor arrays. J Biomol Screen. 2005;10:795–805. doi: 10.1177/1087057105279635.
    1. Ceriotti L, et al. Real-time assessment of cytotoxicity by impedance measurement on a 96-well plate. Sens Actuators B. 2007;123:769–778. doi: 10.1016/j.snb.2006.10.024.
    1. Corbert, T. et al. In vivo methods for screening and preclinical testing: use of rodent solid tumors for drug discovery in Anticancer Drug Development Guide: Preclinical Screening, Clinical Trials and Approval (ed. Teicher, B.) 75–99 (Humana Press, 1997).

Source: PubMed

3
Abonneren