Pharmacokinetics and Pharmacodynamics of a 13-mer LNA-inhibitor-miR-221 in Mice and Non-human Primates

Maria Eugenia Gallo Cantafio, Boye Schnack Nielsen, Chiara Mignogna, Mariamena Arbitrio, Cirino Botta, Niels M Frandsen, Christian Rolfo, Pierosandro Tagliaferri, Pierfrancesco Tassone, Maria Teresa Di Martino, Maria Eugenia Gallo Cantafio, Boye Schnack Nielsen, Chiara Mignogna, Mariamena Arbitrio, Cirino Botta, Niels M Frandsen, Christian Rolfo, Pierosandro Tagliaferri, Pierfrancesco Tassone, Maria Teresa Di Martino

Abstract

Locked nucleic acid (LNA) oligonucleotides have been successfully used to efficiently inhibit endogenous small noncoding RNAs in vitro and in vivo. We previously demonstrated that the direct miR-221 inhibition by the novel 13-mer LNA-i-miR-221 induces significant antimyeloma activity and upregulates canonical miR-221 targets in vitro and in vivo. To evaluate the LNA-i-miR-221 pharmacokinetics and pharmacodynamics, novel assays for oligonucleotides quantification in NOD.SCID mice and Cynomolgus monkeys (Macaca fascicularis) plasma, urine and tissues were developed. To this aim, a liquid chromatography/mass spectrometry method, after solid-phase extraction, was used for the detection of LNA-i-miR-221 in plasma and urine, while a specific in situ hybridization assay for tissue uptake analysis was designed. Our analysis revealed short half-life, optimal tissue biovailability and minimal urine excretion of LNA-i-miR-221 in mice and monkeys. Up to 3 weeks, LNA-i-miR-221 was still detectable in mice vital organs and in xenografted tumors, together with p27 target upregulation. Importantly, no toxicity in the pilot monkey study was observed. Overall, our findings indicate the suitability of LNA-i-miR-221 for clinical use and we provide here pilot data for safety analysis and further development of LNA-miRNA-based therapeutics for human cancer.

Figures

Figure 1
Figure 1
Toxicity evaluation of Locked Nucleic Acid (LNA) LNA-i-miR-221 in mouse organs. Hematoxylin and eosin (H&E) staining of heart, liver, and kidney retrieved from mouse 1 week after one cycle (day 1, 4, 8, 15, and 22) of treatment. a, b, c, show representative areas of heart retrieved after saline (a), 25 mg/kg LNA-i-miR-221 (b) or 100 mg/kg course of treatments. d, e, f show representative areas of liver, while g, h, i kidney areas, after saline (d,g), 25 mg/kg (e,h) or 100 mg/kg (f,i) LNA-i-miR-221 treatment. Frames a, b, c, d, e, f are 100× while g, h, i are 200× magnification.
Figure 2
Figure 2
In situ hybridization (ISH) analysis of LNA-i-miR-221. Serial sections from formalin-fixed paraffin-embedded (FFPE) tissue samples of NOD.SCID mouse kidney after a single i.p. (intraperitoneal) injection of 25 mg/kg LNA-i-miR-221 (ad) or saline (e), processed by ISH with Locked Nucleic Acid (LNA) probes for LNA-i-miR-221 (a, b, e), miR-126 (c), and miR-221 scramble (d). Intense LNA-i-miR-221 ISH signal is detectable in tubules at both low (5 nmol/l) and high (20 nmol/l) probe concentrations (a, b), whereas the probe results in no ISH signal in saline-treated kidney. The positive control probe, miR-126, stains endothelial cells including capillaries in the glomeruli (c), and only background staining is seen by the use of the scramble negative control probe (d).
Figure 3
Figure 3
In situ hybridization (ISH) detection of LNA-i-miR-221 in mouse tissues. LNA-i-miR-221 ISH on sections from organs retrieved from mice after 2 (I) and 7 (II) days of a single intraperitoneal (i.p.) injection of the LNA-i-miR-221 at therapeutic dose of 25 mg/kg or saline (III). Tissue sections from LNA-i-miR-221-treated (I and II) or saline-treated (III) liver (a), kidney (b), heart (c), bone marrow (d), and NCI-H929 xenograft tumor (e) samples after miR-221i probe hybridization. LNA-i-miR-221 staining is detectable in macrophages and/or vessels (liver and tumor), tubules (kidney), probably vascular cells (heart), subpopulation of hematopoietic cells (bone marrow). Bars: Liver, kidney, and heart: 100 µm; bone marrow: 12 µm; tumor: 50 µm.
Figure 4
Figure 4
Long-lasting LNA-i-miR-221 signal in mouse tissues. LNA-i-miR-221 in situ hybridization (ISH) in NOD.SCID mouse organs after injection of therapeutic doses of LNA-i-miR-221 at 25 mg/kg at days 1, 4, 8, 15, and 22. The organs were harvested at 1 week (I) or 3 weeks (II) after last injection. The staining intensity is high both after 1 and 3 weeks: (a) liver and (b) kidney maintain an intense signal throughout the period, while a slightly weaker signal is detectable in heart (c), bone marrow (d) and tumor tissue (e) 3 weeks after last dose. No ISH signal is seen in the organs from saline-treated animals (III). Bars: row I, II, and III (bone marrow and tumor) 50 µm, row III (liver, kidney, and heart) 25 µm.
Figure 5
Figure 5
LNA-i-miR-221 in situ hybridization (ISH) signal in mouse brain. LNA-i-miR-221 ISH in brain samples retrieved from mice after 2 (a) or 7 days (b) of a single intraperitoneal (i.p.) injection of the LNA-i-miR-221 at the therapeutic dose of 25 mg/kg or saline (c). Brain sections from LNA-i-miR-221 (a, b) or saline treated (c) animals using miR-221i probe (a, b, c), scramble negative control probe, (d) or positive control probe against miR-126 (e). Intense staining is seen in the liver (f) from the same mouse as in a. No nervous parenchyma showed detectable LNA-i-miR-221 ISH signal.
Figure 6
Figure 6
miR-221 inhibition by LNA-i-miR-221 induces p27 upregulation in mouse tissues. Immunohistochemistry (IHC) of p27 in liver and kidney mouse tissue from untreated mice (a, d) and from mice that underwent a course of 25 mg/kg LNA-i-miR-221 treatment retrieved after 1 (b,e) or 3 (c,f) weeks from last treatment. Increased p27 expression is showed in representative image of liver and kidney (80 and 90% of positive cells, respectively) from treated animals, compared with the same control tissue (60 and 50%) in liver and kidney, respectively. Heart tissue of untreated (g) and treated animals showing the same low expression of p27 (30%) at both time-points evaluated (h,i). MM xenograft tumors showed an increase of positive cells form 20% in the saline treated animals (l) to 40% after 1 week (m) and 60% after 3 weeks (n) from last injection. Positive cells were counted in 10 high-magnification random fields of the most representative areas; the degree of immunostaining for each antibody was expressed as the percentage of positive cells among the total number of cells. Representative image of different tissues are shown as 400× magnification.
Figure 7
Figure 7
Pharmacokinetics (PK) profile of LNA-i-miR-221 in mice. (a) Plot of plasma concentrations at different time-points for LNA-i-miR-221 indicates a rapid plasmatic clearance from 1.5 to 6 hours after single dose injection (25 mg/kg), followed by a distribution phase, till to almost no detectability 6 hours after injection; (b) plasma LNA-i-miR-221 concentration (ng/ml) at each time point from 1.5 to 24 hours; (cd) ISH for LNA-i-miR-221 in xenograft tumors of untreated (c) and treated animals (d) retrieved after 12 hours of single injection of LNA-i-miR-221 (25 mg/kg). **: BLQ ≤ LLQ (LLQ = 25 ng/ml).
Figure 8
Figure 8
Pharmacokinetics (PK) profile of LNA-i-miR-221 in monkey study. (a) Plot of plasma concentration at different time-points for LNA-i-miR-221 indicates a rapid plasmatic clearance from 0.5 to 48 hours after single dose injection (875 mg/kg), followed by distribution phase, till to almost no detectablility after 48 hours; (b) plot of urine concentration versus four range of urine collection spanning from 2 hours to 48 hours, showing that most LNA-i-miR-221 excretion occurred within 6 hours; (c) plasma LNA-i-miR-221 concentration (ng/ml) at each time points (hours); (d) urine LNA-i-miR-221 concentration (ng/ml) at each time point. **: BLQ ≤ LLQ (LLQ = 50 ng/ml).

References

    1. Amodio, N, Di Martino, MT, Neri, A, Tagliaferri, P and Tassone, P (2013). Non-coding RNA: a novel opportunity for the personalized treatment of multiple myeloma. Expert Opin Biol Ther 13 (Suppl. 1): S125–S137.
    1. Esteller, M (2011). Non-coding RNAs in human disease. Nat Rev Genet 12: 861–874.
    1. Calin, GA, Sevignani, C, Dumitru, CD, Hyslop, T, Noch, E, Yendamuri, S et al. (2004). Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. Proc Natl Acad Sci USA 101: 2999–3004.
    1. Lin, C, Li, X, Zhang, Y, Guo, Y, Zhou, J, Gao, K et al. (2015). The microRNA feedback regulation of p63 in cancer progression. Oncotarget 6: 8434–8453.
    1. Di Martino, MT, Guzzi, PH, Caracciolo, D, Agnelli, L, Neri, A, Walker, BA et al. (2015). Integrated analysis of microRNAs, transcription factors and target genes expression discloses a specific molecular architecture of hyperdiploid multiple myeloma. Oncotarget 6: 19132–19147.
    1. Lionetti, M, Musto, P, Di Martino, MT, Fabris, S, Agnelli, L, Todoerti, K et al. (2013). Biological and clinical relevance of miRNA expression signatures in primary plasma cell leukemia. Clin Cancer Res 19: 3130–3142.
    1. Tagliaferri, P, Rossi, M, Di Martino, MT, Amodio, N, Leone, E, Gulla, A et al. (2012). Promises and challenges of MicroRNA-based treatment of multiple myeloma. Curr Cancer Drug Targets 12: 838–846.
    1. Amodio, N, Rossi, M, Raimondi, L, Pitari, MR, Botta, C, Tagliaferri, P et al. (2015). miR-29s: a family of epi-miRNAs with therapeutic implications in hematologic malignancies. Oncotarget 6: 12837–12861.
    1. Rossi, M, Amodio, N, Di Martino, MT, Caracciolo, D, Tagliaferri, P and Tassone, P (2013). From target therapy to miRNA therapeutics of human multiple myeloma: theoretical and technological issues in the evolving scenario. Curr Drug Targets 14: 1144–1149.
    1. Thorsen, SB, Obad, S, Jensen, NF, Stenvang, J and Kauppinen, S (2012). The therapeutic potential of microRNAs in cancer. Cancer J 18: 275–284.
    1. Amodio, N, Bellizzi, D, Leotta, M, Raimondi, L, Biamonte, L, D'Aquila, P et al. (2013). miR-29b induces SOCS-1 expression by promoter demethylation and negatively regulates migration of multiple myeloma and endothelial cells. Cell Cycle 12: 3650–3662.
    1. Di Martino, MT, Gullà, A, Gallo Cantafio, ME, Altomare, E, Amodio, N, Leone, E et al. (2014). In vitro and in vivo activity of a novel locked nucleic acid (LNA)-inhibitor-miR-221 against multiple myeloma cells. PLoS One 9: e89659.
    1. Raimondi, L, Amodio, N, Di Martino, MT, Altomare, E, Leotta, M, Caracciolo, D et al. (2014). Targeting of multiple myeloma-related angiogenesis by miR-199a-5p mimics: in vitro and in vivo anti-tumor activity. Oncotarget 5: 3039–3054.
    1. Rossi, M, Amodio, N, Di Martino, MT, Tagliaferri, P, Tassone, P and Cho, WC (2014). MicroRNA and multiple myeloma: from laboratory findings to translational therapeutic approaches. Curr Pharm Biotechnol 15: 459–467.
    1. Leotta, M, Biamonte, L, Raimondi, L, Ronchetti, D, Di Martino, MT, Botta, C et al. (2014). A p53-dependent tumor suppressor network is induced by selective miR-125a-5p inhibition in multiple myeloma cells. J Cell Physiol 229: 2106–2116.
    1. Leone, E, Morelli, E, Di Martino, MT, Amodio, N, Foresta, U, Gullà, A et al. (2013). Targeting miR-21 inhibits in vitro and in vivo multiple myeloma cell growth. Clin Cancer Res 19: 2096–2106.
    1. Scognamiglio, I, Di Martino, MT, Campani, V, Virgilio, A, Galeone, A, Gullà, A et al. (2014). Transferrin-conjugated SNALPs encapsulating 2'-O-methylated miR-34a for the treatment of multiple myeloma. Biomed Res Int 2014: 217–365.
    1. Amodio, N, Leotta, M, Bellizzi, D, Di Martino, MT, D'Aquila, P, Lionetti, M et al. (2012). DNA-demethylating and anti-tumor activity of synthetic miR-29b mimics in multiple myeloma. Oncotarget 3: 1246–1258.
    1. Di Martino, MT, Campani, V, Misso, G, Gallo Cantafio, ME, Gullà, A, Foresta, U et al. (2014). In vivo activity of miR-34a mimics delivered by stable nucleic acid lipid particles (SNALPs) against multiple myeloma. PLoS One 9: e90005.
    1. Di Martino, MT, Gullà, A, Cantafio, ME, Lionetti, M, Leone, E, Amodio, N et al. (2013). In vitro and in vivo anti-tumor activity of miR-221/222 inhibitors in multiple myeloma. Oncotarget 4: 242–255.
    1. Tassone, P, Neri, P, Burger, R, Di Martino, MT, Leone, E, Amodio, N et al. (2012). Mouse models as a translational platform for the development of new therapeutic agents in multiple myeloma. Curr Cancer Drug Targets 12: 814–822.
    1. Bader, AG (2012). miR-34 - a microRNA replacement therapy is headed to the clinic. Front Genet 3: 120.
    1. Amodio, N, Di Martino, MT, Foresta, U, Leone, E, Lionetti, M, Leotta, M et al. (2012). miR-29b sensitizes multiple myeloma cells to bortezomib-induced apoptosis through the activation of a feedback loop with the transcription factor Sp1. Cell Death Dis 3: e436.
    1. Di Martino, MT, Leone, E, Amodio, N, Foresta, U, Lionetti, M, Pitari, MR et al. (2012). Synthetic miR-34a mimics as a novel therapeutic agent for multiple myeloma: in vitro and in vivo evidence. Clin Cancer Res 18: 6260–6270.
    1. Morelli, E, Leone, E, Cantafio, ME, Di Martino, MT, Amodio, N, Biamonte, L et al. (2015). Selective targeting of IRF4 by synthetic microRNA-125b-5p mimics induces anti-multiple myeloma activity in vitro and in vivo. Leukemia 29: 2173–2183.
    1. Han, Z, Zhang, Y, Yang, Q, Liu, B, Wu, J, Zhang, Y et al. (2015). miR-497 and miR-34a retard lung cancer growth by co-inhibiting cyclin E1 (CCNE1). Oncotarget 6: 13149–13163.
    1. Liu, Y, Li, X, Zhu, S, Zhang, JG, Yang, M, Qin, Q et al. (2015). Ectopic expression of miR-494 inhibited the proliferation, invasion and chemoresistance of pancreatic cancer by regulating SIRT1 and c-Myc. Gene Ther 22: 729–738.
    1. Tréhoux, S, Lahdaoui, F, Delpu, Y, Renaud, F, Leteurtre, E, Torrisani, J et al. (2015). Micro-RNAs miR-29a and miR-330-5p function as tumor suppressors by targeting the MUC1 mucin in pancreatic cancer cells. Biochim Biophys Acta 1853(10 Pt A): 2392–2403.
    1. Zheng, F, Liao, YJ, Cai, MY, Liu, TH, Chen, SP, Wu, PH et al. (2015). Systemic delivery of microRNA-101 potently inhibits hepatocellular carcinoma in vivo by repressing multiple targets. PLoS Genet 11: e1004873.
    1. Guzman-Aranguez, A, Loma, P and Pintor, J (2013). Small-interfering RNAs (siRNAs) as a promising tool for ocular therapy. Br J Pharmacol 170: 730–747.
    1. Moreno, PM and Pêgo, AP (2014). Therapeutic antisense oligonucleotides against cancer: hurdling to the clinic. Front Chem 2: 87.
    1. Lundin, KE, Højland, T, Hansen, BR, Persson, R, Bramsen, JB, Kjems, J et al. (2013). Biological activity and biotechnological aspects of locked nucleic acids. Adv Genet 82: 47–107.
    1. Yu, RZ, Grundy, JS and Geary, RS (2013). Clinical pharmacokinetics of second generation antisense oligonucleotides. Expert Opin Drug Metab Toxicol 9: 169–182.
    1. Elmén, J, Lindow, M, Schütz, S, Lawrence, M, Petri, A, Obad, S et al. (2008). LNA-mediated microRNA silencing in non-human primates. Nature 452: 896–899.
    1. Straarup, EM, Fisker, N, Hedtjärn, M, Lindholm, MW, Rosenbohm, C, Aarup, V et al. (2010). Short locked nucleic acid antisense oligonucleotides potently reduce apolipoprotein B mRNA and serum cholesterol in mice and non-human primates. Nucleic Acids Res 38: 7100–7111.
    1. Lanford, RE, Hildebrandt-Eriksen, ES, Petri, A, Persson, R, Lindow, M, Munk, ME et al. (2010). Therapeutic silencing of microRNA-122 in primates with chronic hepatitis C virus infection. Science 327: 198–201.
    1. Hansen, JB, Fisker, N, Westergaard, M, Kjaerulff, LS, Hansen, HF, Thrue, CA et al. (2008). SPC3042: a proapoptotic survivin inhibitor. Mol Cancer Ther 7: 2736–2745.
    1. Obad, S, dos Santos, CO, Petri, A, Heidenblad, M, Broom, O, Ruse, C et al. (2011). Silencing of microRNA families by seed-targeting tiny LNAs. Nat Genet 43: 371–378.
    1. Bianchini, D, Omlin, A, Pezaro, C, Lorente, D, Ferraldeschi, R, Mukherji, D et al. (2013). First-in-human phase I study of EZN-4176, a locked nucleic acid antisense oligonucleotide to exon 4 of the androgen receptor mRNA in patients with castration-resistant prostate cancer. Br J Cancer 109: 2579–2586.
    1. Janssen, HL, Reesink, HW, Lawitz, EJ, Zeuzem, S, Rodriguez-Torres, M, Patel, K et al. (2013). Treatment of HCV infection by targeting microRNA. N Engl J Med 368: 1685–1694.
    1. van Dongen, WD and Niessen, WM (2011). Bioanalytical LC-MS of therapeutic oligonucleotides. Bioanalysis 3: 541–564.
    1. Yu, RZ, Kim, TW, Hong, A, Watanabe, TA, Gaus, HJ and Geary, RS (2007). Cross-species pharmacokinetic comparison from mouse to man of a second-generation antisense oligonucleotide, ISIS 301012, targeting human apolipoprotein B-100. Drug Metab Dispos 35: 460–468.
    1. Koller, E, Vincent, TM, Chappell, A, De, S, Manoharan, M and Bennett, CF (2011). Mechanisms of single-stranded phosphorothioate modified antisense oligonucleotide accumulation in hepatocytes. Nucleic Acids Res 39: 4795–4807.
    1. Geary, RS, Yu, RZ and Levin, AA (2001). Pharmacokinetics of phosphorothioate antisense oligodeoxynucleotides. Curr Opin Investig Drugs 2: 562–573.
    1. Erickson, MA, Niehoff, ML, Farr, SA, Morley, JE, Dillman, LA, Lynch, KM et al. (2012). Peripheral administration of antisense oligonucleotides targeting the amyloid-β protein precursor reverses AβPP and LRP-1 overexpression in the aged SAMP8 mouse brain. J Alzheimers Dis 28: 951–960.
    1. Banks, WA (2011). Measurement of phosphorothioate oligodeoxynucleotide antisense transport across the blood-brain barrier. Methods Mol Biol 789: 337–342.
    1. Banks, WA, Farr, SA, Butt, W, Kumar, VB, Franko, MW and Morley, JE (2001). Delivery across the blood-brain barrier of antisense directed against amyloid beta: reversal of learning and memory deficits in mice overexpressing amyloid precursor protein. J Pharmacol Exp Ther 297: 1113–1121.
    1. Evers, MM, Toonen, LJ and van Roon-Mom, WM (2015). Antisense oligonucleotides in therapy for neurodegenerative disorders. Adv Drug Deliv Rev 87: 90–103.
    1. Sands, H, Gorey-Feret, LJ, Cocuzza, AJ, Hobbs, FW, Chidester, D and Trainor, GL (1994). Biodistribution and metabolism of internally 3H-labeled oligonucleotides. I. Comparison of a phosphodiester and a phosphorothioate. Mol Pharmacol 45: 932–943.
    1. Kole, R, Krainer, AR and Altman, S (2012). RNA therapeutics: beyond RNA interference and antisense oligonucleotides. Nat Rev Drug Discov 11: 125–140.
    1. Jørgensen, S, Baker, A, Møller, S and Nielsen, BS (2010). Robust one-day in situ hybridization protocol for detection of microRNAs in paraffin samples using LNA probes. Methods 52: 375–381.
    1. Nielsen, BS, Jørgensen, S, Fog, JU, Søkilde, R, Christensen, IJ, Hansen, U et al. (2011). High levels of microRNA-21 in the stroma of colorectal cancers predict short disease-free survival in stage II colon cancer patients. Clin Exp Metastasis 28: 27–38.
    1. Nielsen, BS, Møller, T and Holmstrøm, K (2014). Chromogen detection of microRNA in frozen clinical tissue samples using LNA™ probe technology. Methods Mol Biol 1211: 77–84.

Source: PubMed

3
Suscribir