In vitro and in vivo activity of a novel locked nucleic acid (LNA)-inhibitor-miR-221 against multiple myeloma cells

Maria Teresa Di Martino, Annamaria Gullà, Maria Eugenia Gallo Cantafio, Emanuela Altomare, Nicola Amodio, Emanuela Leone, Eugenio Morelli, Santo Giovanni Lio, Daniele Caracciolo, Marco Rossi, Niels M Frandsen, Pierosandro Tagliaferri, Pierfrancesco Tassone, Maria Teresa Di Martino, Annamaria Gullà, Maria Eugenia Gallo Cantafio, Emanuela Altomare, Nicola Amodio, Emanuela Leone, Eugenio Morelli, Santo Giovanni Lio, Daniele Caracciolo, Marco Rossi, Niels M Frandsen, Pierosandro Tagliaferri, Pierfrancesco Tassone

Abstract

Background & aim: The miR-221/222 cluster is upregulated in malignant plasma cells from multiple myeloma (MM) patients harboring the t(4;14) translocation. We previously reported that silencing of miR-221/222 by an antisense oligonucleotide induces anti-MM activity and upregulates canonical miR-221/222 targets. The in vivo anti-tumor activity occurred when miR-221/222 inhibitors were delivered directly into MM xenografts. The aim of the present study was to evaluate the anti-MM activity of a novel phosphorothioate modified backbone 13-mer locked nucleic acid (LNA)-Inhibitor-miR-221 (LNA-i-miR-221) specifically designed for systemic delivery.

Methods: In vitro anti-MM activity of LNA-i-miR-221 was evaluated by cell proliferation and BrdU uptake assays. In vivo studies were performed with non-obese diabetic/severe combined immunodeficient (NOD.SCID) mice bearing t(4;14) MM xenografts, which were intraperitoneally or intravenously treated with naked LNA-i-miR-221. RNA extracts from retrieved tumors were analyzed for miR-221 levels and modulation of canonical targets expression. H&E staining and immunohistochemistry were performed on retrieved tumors and mouse vital organs.

Results: In vitro, LNA-i-miR-221 exerted strong antagonistic activity against miR-221 and induced upregulation of the endogenous target p27Kip1. It had a marked anti-proliferative effect on t(4;14)-translocated MM cells but not on MM cells not carrying the translocation and not overexpressing miR-221. In vivo, systemic treatment with LNA-i-miR-221 triggered significant anti-tumor activity against t(4;14) MM xenografts; it also induced miR-221 downregulation, upregulated p27Kip1 and reduced Ki-67. No behavioral changes or organ-related toxicity were observed in mice as a consequence of treatments.

Conclusions: LNA-i-miR-221 is a highly stable, effective agent against t(4;14) MM cells, and is suitable for systemic use. These data provide the rationale for the clinical development of LNA-i-miR-221 for the treatment of MM.

Conflict of interest statement

Competing Interests: Dr. Niels Montano Frandsen is employed by Exiqon A/S, Vedbaek, Denmark. This does not alter our adherence to PLOS ONE policies on sharing data and materials.

Figures

Figure 1. LNA-i-miR-221 specifically recognizes the miR-221…
Figure 1. LNA-i-miR-221 specifically recognizes the miR-221 complementary sequence.
Luciferase reporter assay of NCI-H929 cells co-transfected with pLightSwitch_3′UTR Reporter Vector containing the miR-221-3p synthetic sequence cloned downstream of the Renilla luminescent reporter gene (RenSP) and miR-221/222 mimics (A) or LNA-i-miR-221 (B) or scrambled sequences as control. The firefly luciferase activity was normalized to Renilla luciferase activity. The data are shown as relative luciferase activity of miR-221/222-transfected cells versus the control (miR-NC) or LNA-i-miR-221-transfected cells versus the scrambled control (LNA-i-miR-NC). C) Dual-luciferase assay of NCI-H929 cells co-transfected with firefly luciferase constructs containing the 3′UTR of p27Kip1 and LNA-i-miR-221 or scrambled oligonucleotides (NC) as indicated. Firefly luciferase activity was normalized to r Renilla luciferase activity. The data are shown as relative luciferase activity of LNA-i-miR-221-transfected cells versus the control (NC).
Figure 2. Antiproliferative effects induced by transient…
Figure 2. Antiproliferative effects induced by transient expression of LNA-i-miR-221 in MM cell lines.
Effects on proliferation (A) and BrdU uptake (B) in NCI-H929 cells induced by transient LNA-miR-221 inhibitor (LNA-i-miR-221) transfection compared to scrambled control (LNA-i-miR-NC). C) OPM-2 cell growth inhibition. MM cells not bearing t(4;14): RPMI-8226 (D), KMS12-BM (E) and INA-6 (F) are not affected by transfection with LNA-i-miR-221 as compared to scrambled LNA sequences. Averaged values ±SD from 3 independent experiments.
Figure 3. Molecular effects induced by LNA-i-miR-221…
Figure 3. Molecular effects induced by LNA-i-miR-221 transfection in MM cells.
miR-221(A) q-RT-PCR 24, 48 and 72 hours after transfection with LNA-i-miR-221 and LNA-i-miR-NC in NCI-H929 cells. The results are shown as miRNA expression levels after normalization with RNU44 and ΔΔCt calculations. Data represent the average of 3 independent experiments ±SD. B) q-RT-PCR of p27Kip1 mRNA expression 24 and 48 hours after transfection with LNA-i-miR-221 or scrambled control in NCI-H929 cells. Data represent the average of 3 independent experiments ±SD after normalization with GAPDH mRNA and ΔΔCt calculations. (*) P

Figure 4. In vivo anti-tumor activity of…

Figure 4. In vivo anti-tumor activity of LNA-i-miR-221 in MM-xenografted SCID/NOD mice.

Effects of LNA-i-miR-221…

Figure 4. In vivo anti-tumor activity of LNA-i-miR-221 in MM-xenografted SCID/NOD mice.
Effects of LNA-i-miR-221 after 2 intraperitoneal injections of 25 mg/kg (A) and 4 intravenous injections of 25 mg/kg (B). MM-xenografted mice were treated with saline solution of LNA-i-miR-221 or LNA-i-miR-NC as control. The average tumor volume of each group ±SD is reported. P values were calculated for miRNA inhibitors versus scrambled oligonucleotides (NC) and significant P values (P<0.05) are indicated by asterisks. C) q-RT-PCR of miR-221 in retrieved tumors treated intravenously with LNA-i-miR-221 or LNA-i-miR-NC. The results are shown as averaged miRNA expression after normalization with RNU44 and ΔΔCt calculations as compared to control (miRNA expression in LNA-i-miR-NC treated animals) ±SD.*P<0.05. D) q-RT-PCR of miR-221 in liver, kidney and heart tissue biopsies of mice after 2 weeks of treatment. The results are shown for each organ as average miRNA expression after normalization with snoRNA-202 and ΔΔCt calculations with respect to miRNA expression in LNA-i-miR-NC-treated animals. The results represents the average value obtained in 3 different mice ±SD. **P<0.01.

Figure 5. LNA-i-miR-221 antiproliferative activity and target…

Figure 5. LNA-i-miR-221 antiproliferative activity and target silencing in retrieved MM xenografted tumors.

A) q-RT-PCR…

Figure 5. LNA-i-miR-221 antiproliferative activity and target silencing in retrieved MM xenografted tumors.
A) q-RT-PCR of p27Kip1 mRNA levels in treated tumors retrieved from mice after intravenous LNA-i-miR-221 treatment. Raw Ct values were normalized to GAPDH housekeeping mRNA and expressed as ΔΔCt values calculated using the comparative cross threshold method (miRNA expression in LNA-i-miR-NC treated animals) ±SD. B) Western blot analysis of p27Kip1 protein in retrieved tumors from mice treated with LNA-i-miR-221 inhibitors or LNA-i-miR-NC. GAPDH was the protein loading control. C) H&E (200-fold magnification), p27Kip1 (200-fold magnification) and Ki-67 (10–fold magnification) immunohistochemistry staining of xenografted tumors retrieved from treated animals. Representative images are shown.
Figure 4. In vivo anti-tumor activity of…
Figure 4. In vivo anti-tumor activity of LNA-i-miR-221 in MM-xenografted SCID/NOD mice.
Effects of LNA-i-miR-221 after 2 intraperitoneal injections of 25 mg/kg (A) and 4 intravenous injections of 25 mg/kg (B). MM-xenografted mice were treated with saline solution of LNA-i-miR-221 or LNA-i-miR-NC as control. The average tumor volume of each group ±SD is reported. P values were calculated for miRNA inhibitors versus scrambled oligonucleotides (NC) and significant P values (P<0.05) are indicated by asterisks. C) q-RT-PCR of miR-221 in retrieved tumors treated intravenously with LNA-i-miR-221 or LNA-i-miR-NC. The results are shown as averaged miRNA expression after normalization with RNU44 and ΔΔCt calculations as compared to control (miRNA expression in LNA-i-miR-NC treated animals) ±SD.*P<0.05. D) q-RT-PCR of miR-221 in liver, kidney and heart tissue biopsies of mice after 2 weeks of treatment. The results are shown for each organ as average miRNA expression after normalization with snoRNA-202 and ΔΔCt calculations with respect to miRNA expression in LNA-i-miR-NC-treated animals. The results represents the average value obtained in 3 different mice ±SD. **P<0.01.
Figure 5. LNA-i-miR-221 antiproliferative activity and target…
Figure 5. LNA-i-miR-221 antiproliferative activity and target silencing in retrieved MM xenografted tumors.
A) q-RT-PCR of p27Kip1 mRNA levels in treated tumors retrieved from mice after intravenous LNA-i-miR-221 treatment. Raw Ct values were normalized to GAPDH housekeeping mRNA and expressed as ΔΔCt values calculated using the comparative cross threshold method (miRNA expression in LNA-i-miR-NC treated animals) ±SD. B) Western blot analysis of p27Kip1 protein in retrieved tumors from mice treated with LNA-i-miR-221 inhibitors or LNA-i-miR-NC. GAPDH was the protein loading control. C) H&E (200-fold magnification), p27Kip1 (200-fold magnification) and Ki-67 (10–fold magnification) immunohistochemistry staining of xenografted tumors retrieved from treated animals. Representative images are shown.

References

    1. Pichiorri F, Suh SS, Ladetto M, Kuehl M, Palumbo T, et al. (2008) MicroRNAs regulate critical genes associated with multiple myeloma pathogenesis. Proc Natl Acad Sci U S A 105: 12885–12890.
    1. Chi J, Ballabio E, Chen XH, Kusec R, Taylor S, et al. (2011) MicroRNA expression in multiple myeloma is associated with genetic subtype, isotype and survival. Biol Direct 6: 23.
    1. Wu P, Agnelli L, Walker BA, Todoerti K, Lionetti M, et al. (2013) Improved risk stratification in myeloma using a microRNA-based classifier. Br J Haematol.
    1. Corthals SL, Sun SM, Kuiper R, de Knegt Y, Broyl A, et al. (2011) MicroRNA signatures characterize multiple myeloma patients. Leukemia 25: 1784–1789.
    1. Lionetti M, Biasiolo M, Agnelli L, Todoerti K, Mosca L, et al. (2009) Identification of microRNA expression patterns and definition of a microRNA/mRNA regulatory network in distinct molecular groups of multiple myeloma. Blood 114: e20–26.
    1. Tagliaferri P, Rossi M, Di Martino MT, Amodio N, Leone E, et al. (2012) Promises and challenges of MicroRNA-based treatment of multiple myeloma. Curr Cancer Drug Targets 12: 838–846.
    1. Thorsen SB, Obad S, Jensen NF, Stenvang J, Kauppinen S (2012) The therapeutic potential of microRNAs in cancer. Cancer J 18: 275–284.
    1. Rossi M, Amodio N, Di Martino MT, Caracciolo D, Tagliaferri P, et al. (2013) From target therapy to miRNA therapeutics of human multiple myeloma: theoretical and technological issues in the evolving scenario. Curr Drug Targets.
    1. Misso G, Zappavigna S, Castellano M, De Rosa G, Di Martino MT, et al. (2013) Emerging pathways as individualized therapeutic target of multiple myeloma. Expert Opin Biol Ther 13 Suppl 1S95–S109.
    1. Amodio N, Di Martino MT, Neri A, Tagliaferri P, Tassone P (2013) Non-coding RNA: a novel opportunity for the personalized treatment of multiple myeloma. Expert Opin Biol Ther 13 Suppl 1S125–137.
    1. Dimopoulos K, Gimsing P, Gronbaek K (2013) Aberrant microRNA expression in multiple myeloma. Eur J Haematol 91: 95–105.
    1. Di Martino MT, Leone E, Amodio N, Foresta U, Lionetti M, 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. Amodio N, Di Martino MT, Foresta U, Leone E, Lionetti 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. Amodio N, Leotta M, Bellizzi D, Di Martino MT, D’Aquila P, 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, Gulla A, Cantafio ME, Lionetti M, Leone E, et al. (2013) In vitro and in vivo anti-tumor activity of miR-221/222 inhibitors in multiple myeloma. Oncotarget 4: 242–255.
    1. Leone E, Morelli E, Di Martino MT, Amodio N, Foresta U, et al. (2013) Targeting miR-21 inhibits in vitro and in vivo multiple myeloma cell growth. Clin Cancer Res 19: 2096–2106.
    1. Quintavalle C, Garofalo M, Zanca C, Romano G, Iaboni M, et al. (2012) miR-221/222 overexpession in human glioblastoma increases invasiveness by targeting the protein phosphate PTPmu. Oncogene 31: 858–868.
    1. Galardi S, Mercatelli N, Farace MG, Ciafre SA (2011) NF-kB and c-Jun induce the expression of the oncogenic miR-221 and miR-222 in prostate carcinoma and glioblastoma cells. Nucleic Acids Res 39: 3892–3902.
    1. Zhang CZ, Zhang JX, Zhang AL, Shi ZD, Han L, et al. (2010) MiR-221 and miR-222 target PUMA to induce cell survival in glioblastoma. Mol Cancer 9: 229.
    1. Pineau P, Volinia S, McJunkin K, Marchio A, Battiston C, et al. (2010) miR-221 overexpression contributes to liver tumorigenesis. Proc Natl Acad Sci U S A 107: 264–269.
    1. Sredni ST, Bonaldo Mde F, Costa FF, Huang CC, Hamm CA, et al. (2010) Upregulation of mir-221 and mir-222 in atypical teratoid/rhabdoid tumors: potential therapeutic targets. Childs Nerv Syst 26: 279–283.
    1. Mercatelli N, Coppola V, Bonci D, Miele F, Costantini A, et al. (2008) The inhibition of the highly expressed miR-221 and miR-222 impairs the growth of prostate carcinoma xenografts in mice. PLoS One 3: e4029.
    1. Felicetti F, Errico MC, Bottero L, Segnalini P, Stoppacciaro A, et al. (2008) The promyelocytic leukemia zinc finger-microRNA-221/−222 pathway controls melanoma progression through multiple oncogenic mechanisms. Cancer Res 68: 2745–2754.
    1. Hwang MS, Yu N, Stinson SY, Yue P, Newman RJ, et al. (2013) miR-221/222 Targets Adiponectin Receptor 1 to Promote the Epithelial-to-Mesenchymal Transition in Breast Cancer. PLoS One 8: e66502.
    1. Jikuzono T, Kawamoto M, Yoshitake H, Kikuchi K, Akasu H, et al. (2013) The miR-221/222 cluster, miR-10b and miR-92a are highly upregulated in metastatic minimally invasive follicular thyroid carcinoma. Int J Oncol 42: 1858–1868.
    1. Garofalo M, Romano G, Di Leva G, Nuovo G, Jeon YJ, et al. (2012) EGFR and MET receptor tyrosine kinase-altered microRNA expression induces tumorigenesis and gefitinib resistance in lung cancers. Nat Med 18: 74–82.
    1. Garofalo M, Di Leva G, Romano G, Nuovo G, Suh SS, et al. (2009) miR-221&222 regulate TRAIL resistance and enhance tumorigenicity through PTEN and TIMP3 downregulation. Cancer Cell 16: 498–509.
    1. Geary RS, Yu RZ, Levin AA (2001) Pharmacokinetics of phosphorothioate antisense oligodeoxynucleotides. Curr Opin Investig Drugs 2: 562–573.
    1. Najafi-Shoushtari SH, Kristo F, Li Y, Shioda T, Cohen DE, et al. (2010) MicroRNA-33 and the SREBP host genes cooperate to control cholesterol homeostasis. Science 328: 1566–1569.
    1. Ng R, Song G, Roll GR, Frandsen NM, Willenbring H (2012) A microRNA-21 surge facilitates rapid cyclin D1 translation and cell cycle progression in mouse liver regeneration. J Clin Invest 122: 1097–1108.
    1. Putta S, Lanting L, Sun G, Lawson G, Kato M, et al. (2012) Inhibiting microRNA-192 ameliorates renal fibrosis in diabetic nephropathy. J Am Soc Nephrol 23: 458–469.
    1. Zarjou A, Yang S, Abraham E, Agarwal A, Liu G (2011) Identification of a microRNA signature in renal fibrosis: role of miR-21. Am J Physiol Renal Physiol 301: F793–801.
    1. Liu G, Friggeri A, Yang Y, Milosevic J, Ding Q, et al. (2010) miR-21 mediates fibrogenic activation of pulmonary fibroblasts and lung fibrosis. J Exp Med 207: 1589–1597.
    1. Boon RA, Seeger T, Heydt S, Fischer A, Hergenreider E, et al. (2011) MicroRNA-29 in aortic dilation: implications for aneurysm formation. Circ Res 109: 1115–1119.
    1. Maegdefessel L, Azuma J, Toh R, Merk DR, Deng A, et al. (2012) Inhibition of microRNA-29b reduces murine abdominal aortic aneurysm development. J Clin Invest 122: 497–506.
    1. Worm J, Stenvang J, Petri A, Frederiksen KS, Obad S, et al. (2009) Silencing of microRNA-155 in mice during acute inflammatory response leads to derepression of c/ebp Beta and down-regulation of G-CSF. Nucleic Acids Res 37: 5784–5792.
    1. Montgomery RL, Hullinger TG, Semus HM, Dickinson BA, Seto AG, et al. (2011) Therapeutic inhibition of miR-208a improves cardiac function and survival during heart failure. Circulation 124: 1537–1547.
    1. Grueter CE, van Rooij E, Johnson BA, DeLeon SM, Sutherland LB, et al. (2012) A cardiac microRNA governs systemic energy homeostasis by regulation of MED13. Cell 149: 671–683.
    1. Boon RA, Iekushi K, Lechner S, Seeger T, Fischer A, et al. (2013) MicroRNA-34a regulates cardiac ageing and function. Nature 495: 107–110.
    1. Janssen HL, Reesink HW, Lawitz EJ, Zeuzem S, Rodriguez-Torres M, et al. (2013) Treatment of HCV infection by targeting microRNA. N Engl J Med 368: 1685–1694.
    1. Lionetti M, Musto P, Di Martino MT, Fabris S, Agnelli L, et al. (2013) Biological and clinical relevance of miRNA expression signatures in primary plasma cell leukemia. Clin Cancer Res 19: 3130–3142.
    1. Burger R, Le Gouill S, Tai YT, Shringarpure R, Tassone P, et al. (2009) Janus kinase inhibitor INCB20 has antiproliferative and apoptotic effects on human myeloma cells in vitro and in vivo. Mol Cancer Ther 8: 26–35.
    1. Neri P, Tassone P, Shammas M, Yasui H, Schipani E, et al. (2007) Biological pathways and in vivo antitumor activity induced by Atiprimod in myeloma. Leukemia 21: 2519–2526.
    1. Tassone P, Neri P, Burger R, Savino R, Shammas M, et al. (2005) Combination therapy with interleukin-6 receptor superantagonist Sant7 and dexamethasone induces antitumor effects in a novel SCID-hu In vivo model of human multiple myeloma. Clin Cancer Res 11: 4251–4258.
    1. Tassone P, Neri P, Carrasco DR, Burger R, Goldmacher VS, et al. (2005) A clinically relevant SCID-hu in vivo model of human multiple myeloma. Blood 106: 713–716.
    1. Livak KJ, Schmittgen TD (2001) Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 25: 402–408.
    1. Neri P, Tagliaferri P, Di Martino MT, Calimeri T, Amodio N, et al. (2008) In vivo anti-myeloma activity and modulation of gene expression profile induced by valproic acid, a histone deacetylase inhibitor. Br J Haematol 143: 520–531.
    1. Lanford RE, Hildebrandt-Eriksen ES, Petri A, Persson R, Lindow M, et al. (2010) Therapeutic silencing of microRNA-122 in primates with chronic hepatitis C virus infection. Science 327: 198–201.

Source: PubMed

3
Tilaa