In Vitro and Vivo Activity Against Multiple Myeloma Cells Of a Novel Locked Nucleic Acid (LNA)-Mir-221 Inhibitor

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3166-3166
Author(s):  
Maria Teresa Di Martino ◽  
Annamaria Gullà ◽  
Maria Eugenia Gallo Cantafio ◽  
Emanuela Altomare ◽  
Nicola Amodio ◽  
...  

Abstract miR-221/222 are two highly homologous microRNAs (miRNAs), encoded in tandem on the chromosome X, whose up-regulation has been found in several malignancies and are thought to promote cell proliferation via down-regulation of p27 and/or p57, two negative regulators of G1 to S phase cell cycle progression. We demonstrated up-regulation of both miRNAs in malignant plasma cells (PCs) from multiple myeloma (MM) patients belonging to distinct TC (translocation/Cyclin) groups, including TC2 and TC4. A rising body of evidence suggests that silencing miRNAs with oncogenic potential could represent a novel approach for human cancer therapy. We previously demonstrated that silencing miR-221/222 exerts significant anti-MM activity and triggers canonical targets in vitro and in vivo. Here, in the aim to progress to clinical translation of our proof-of-principle findings, we investigated the anti-tumor activity and the appropriateness for systemic delivery of a novel and originally designed LNA-miR-221, a 13-mer antisense miR-221 inhibitor, which took advantage of locked nucleic acid (LNA) technology and phosphorothioate backbone chemistry for increasing affinity for miR-221 and nuclease resistance. We found that enforced ectopic expression of LNA-miR-221 in t(4;14) MM cells significantly inhibited growth and survival of MM cells in vitro. In treated cells, we detected knock down of miR-221/222 together and increased levels of both p27Kip1 mRNA and protein. Specific activity of this LNA-miR-221 inhibitor was confirmed by the use of a 3’UTR reporter (luciferase renilla/firefly) constructs containing, miR-221 target site. This construct was co-transfected either with miR-221/222 mimics or LNA-miR-221 inhibitor into MM cells. As predicted, a reduced luciferase activity was detected in miR-221/222 mimics co-transfected cells with each 3’UTR reporter plasmid, while increase luciferase activity was measured in MM cells co-transfected LNA-miR-221 inhibitors indicating an efficient and stable binding to the miRNA target sequence. Importantly, we evaluated the systemic delivery of the LNA-miR-221 inhibitor with saline solution vehicle alone by intraperitoneal or intravenous injection route against MM xenografts in SCID/NOD mice. Significant anti-tumor activity was achieved after 2 weeks of treatment at similar extent by both injection routes. Retrieved tumors from treated animals showed efficient inhibition of miR-221/222, as demonstrated by increased levels of p27Kip1 protein in vivo. H&E staining and immunohistochemical analysis showed wide necrosis areas, reduced Ki67 and a significant increase of p27Kip1 cytoplasmic expression in retrieved tumors from LNA-miR-221 inhibitor-treated mice. No changes in mice behavior or organ toxicity were observed in treated mice. Taken together these findings support the rationale for development of this novel and highly efficient LNA-miR-221 inhibitor as a promising anti-MM drug in subsequent primate toxicology studies. Supported by the Italian Association for Cancer Research (AIRC), PI: PT. “Special Program Molecular Clinical Oncology - 5 per mille” n. 9980, 2010/15. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3025-3025 ◽  
Author(s):  
Maria Eugenia Gallo Cantafio ◽  
Annamaria Gulla ◽  
Nicola Amodio ◽  
Emanuela Leone ◽  
Eugenio Morelli ◽  
...  

Abstract miR-221/222 are highly homologous microRNAs (miRNAs) whose upregulation has been found in several malignancies, including multiple myeloma (MM). Both miRNAs are thought to promote cell proliferation via down-regulation of p27 and/or p57, two negative regulators of G1 to S phase cell cycle progression. We proved that silencing of both miRNAs results in significant anti-MM activity and in downregulation of canonical targets both in vitro and in vivo. In the aim to progress to clinical translation, we designed an original 13mer synthetic inhibitor, specific for systemic delivery, named LNA-i-miR-221, which took advantage from both locked nucleic acid (LNA) technology and phosphorothioate backbone, for increasing the seed sequence binding and nuclease resistance in vivo, respectively. Since no data are presently available, we evaluated the specificity of LNA-i-miR-221 to inhibit endogenous miR-221 and the pharmacokinetic properties (i.e. tissue distribution) in mice and Cynomolgus monkeys. We demonstrated that LNA-i-miR-221 inhibit growth and survival of MM cells bearing high miR-221 levels. Moreover, LNA-i-miR-221-mediated silencing of miR-221 triggered upregulation of p27Kip1 mRNA and protein, evaluated by q-RT-PCR and western blotting, respectively. In vivo, i.p.treatment with 25 mg/kg of LNA-i-miR-221 reduced tumor growth in SCID/NOD mice bearing MM xenografts. Tumors and vital organs (including liver, kidney, bone marrow and heart) were harvested from treated animals and evaluated for pharmacokinetics purposes using in situ hybridization (ISH) assay. After a single i.p. dose of 25 mg/kg, we detected the presence of LNA-i-miR-221 from 2 up to 7 days in tumors and mouse tissues. Interestingly, no toxicity was observed even after long-lasting presence of the inhibitors in vital organs. We also evaluated the LNA-i-miR-221 half-life in mouse plasma using HPLC-MS/MS, which confirmed the rapid uptake of LNA-i-miR-221 in tissues. To evaluate the maximum tolerated doses, in vivo dose escalation treatments was performed using doses from 10 to 100 mg/kg delivered at days 1,4,8,15,22. All treatments were well tolerated. No changes in mice behavior or organ toxicity were observed in treated mice. Finally, a pilot toxicity study was performed in Cynomolgus monkeys. PK results demonstrated that LNA-i-miR-221 has a short serum half-life with a rapid tissue uptake and minimum urinary escretion. In conclusion, our results suggest that LNA-i-miR-221 is a promising anti-MM agent associated with a safety profile in mice and in monkeys, supporting the rationale for development of this novel miR-221 inhibitor in early clinical trials. Disclosures No relevant conflicts of interest to declare.


PLoS ONE ◽  
2014 ◽  
Vol 9 (2) ◽  
pp. e89659 ◽  
Author(s):  
Maria Teresa Di Martino ◽  
Annamaria Gullà ◽  
Maria Eugenia Gallo Cantafio ◽  
Emanuela Altomare ◽  
Nicola Amodio ◽  
...  

2021 ◽  
Vol 12 (5) ◽  
Author(s):  
Xuxing Shen ◽  
Chao Wu ◽  
Meng Lei ◽  
Qing Yan ◽  
Haoyang Zhang ◽  
...  

AbstractCarfilzomib, a second-generation proteasome inhibitor, has significantly improved the survival rate of multiple myeloma (MM) patients, but its clinical application is still restricted by drug resistance and cardiotoxicity. Here, we identified a novel proteasome inhibitor, D395, and assessed its efficacy in treating MM as well as its cardiotoxicity at the preclinical level. The activities of purified and intracellular proteasomes were measured to determine the effect of D395 on the proteasome. CCK-8 and flow cytometry experiments were designed to evaluate the effects of D395 on cell growth and apoptosis. The effects of D395 and carfilzomib on serum enzyme activity, echocardiography features, cardiomyocyte morphology, and hERG channels were also compared. In our study, D395 was highly cytotoxic to MM cell lines and primary MM cells but not normal cells, and it was well tolerated in vivo. Similar to carfilzomib, D395 inhibited osteoclast differentiation in a dose-dependent manner. In particular, D395 exhibited lower cardiotoxicity than carfilzomib in all experiments. In conclusion, D395 is a novel irreversible proteasome inhibitor that has remarkable anti-MM activity and mild cardiotoxicity in vitro and in vivo.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 614-614
Author(s):  
Phil Kearney ◽  
Majken Westergaard ◽  
Henrik F. Hansen ◽  
Ellen M. Staarup ◽  
Troels Koch ◽  
...  

Abstract Much discussion has centred around the utility and benefits of siRNA in both target validation and as a therapeutic option. This has been driven by significant publications including that of Soutcheck et al (Nature432, 173–177 2004), which demonstrated liver targeting as well as in vivo efficacy when siRNA against ApoB was tethered to a cholesterol moiety. Santaris Pharma has developed a third generation nucleic acid chemistry referred to as locked nucleic acid (LNA) which delivers unmatched affinity and stabiliy benefits, largely overcoming the drawbacks associated with traditional antisense molecules. We therefore sought to compare this chemistry with targets which siRNA has been successfully used in in vivo/in vitro settings. The same motif used in the Soutcheck study was targeted with a LNA molecule, and the free siRNA activity was compared to the cholesterol linked and free LNA molecules in their ability ot down regulate ApoB expression. LNA (SPC3197) inhibited ApoB expression by 90% while at an equimolar concentration siRNA was ineffective in the liver and jejunum. Cholesterol linked siRNA was only effective in the jejunum (c50% reduction in mRNA) Fig1. Only the LNA mediated inhibition of ApoB expression was paralleled by a drop in serum cholesterol in the host animal. In a second model siRNA molecules targeting Hif-1a mRNA (Yu et al Lab Invest84, 553–561 2004) were compared to our lead LNA molecule targeting Hif-1a, SPC2968. Interestingly in in vitro analyses these 2 molecules were equally effective. However in a murine model the increased half life of the LNA molecules translated to a potent inhibition of Hif-1a as measured by QPCR. This effect was noted in jejunum and liver, and persisted for at least 4 days. Hif-1a inhibition mediated by siRNA was not seen in any tissue analysed (Fig 2). Finally a 3rd molecule targeting Bcl-2 has entered clinical Phase 1 trials, and data will be presented documenting its improved affinity and stabitily in relation to competitor molecules such as Genasense. Figure Figure


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1718-1718 ◽  
Author(s):  
Toshihiko Ishii ◽  
Asher Alban Chanan-Khan ◽  
Jazur Jafferjee ◽  
Noreen Ersing ◽  
Takeshi Takahashi ◽  
...  

Abstract BIW-8962 is a humanized anti-ganglioside GM2 (GM2) monoclonal antibody, produced by Poteligent technology to enhance ADCC activity. GM2 is expressed on many cancer cells including multiple myeloma (MM), small cell lung cancer and glioma cells. In this study, we evaluated the anti-myeloma activity of BIW-8962 in preclinical myeloma models both in vitro and in vivo. Expression of GM2 was analyzed in 15 human MM cell lines by FCM. Eleven out of 15 MM cell lines had positive surface expression of GM2. GM2 as a potential target was then verified in primary MM samples obtained from patients. Eleven out of 15 samples were positive for GM2. We then used two GM2 positive MM cell lines (U266B1 and KMS-11) and evaluated ADCC and CDC activity of BIW-8962 in vitro. BIW-8962 exhibited a potent ADCC and less potent CDC activity. In vivo anti-tumor activity of BIW-8962 was then examined using the standard subcutaneous xenograft model; KMS-11 was inoculated in the flank of SCID mice. BIW-8962 (intravenously administered biweekly for 3 weeks) exhibited a potent anti-tumor activity from as low a dose level as 0.1 mg/kg. Furthermore, in a more clinically relevant model, in which OPM-2/GFP (GM2 positive MM cell line) cells were intravenously inoculated into SCID mice with preferentially tumor growth within the bone marrow microenvironment, BIW-8962 (intravenously administered biweekly for 4 weeks, 10 mg/kg) suppressed OPM-2/GFP cell growth and serum M protein elevation, demonstrating in vivo anti-myeloma effect of BIW-8962. Our preclinical investigations rationalize clinical evaluation of BIW-8962 in patients with MM. Currently BIW-8962 is being investigated in a Phase 1 study in patients with multiple myeloma.


BMC Cancer ◽  
2016 ◽  
Vol 16 (1) ◽  
Author(s):  
Anna Torres ◽  
Joanna Kozak ◽  
Agnieszka Korolczuk ◽  
Paulina Wdowiak ◽  
Ewa Domańska-Glonek ◽  
...  

2005 ◽  
Vol 94 (1-3) ◽  
pp. 111-121 ◽  
Author(s):  
Sébastien Maillard ◽  
Thibault Ameller ◽  
Juliette Gauduchon ◽  
Angélique Gougelet ◽  
Fabrice Gouilleux ◽  
...  

ChemBioChem ◽  
2005 ◽  
Vol 6 (6) ◽  
pp. 1104-1109 ◽  
Author(s):  
Kees Fluiter ◽  
Miriam Frieden ◽  
Jeroen Vreijling ◽  
Christoph Rosenbohm ◽  
Marit B. De Wissel ◽  
...  

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2416-2416 ◽  
Author(s):  
Parisa Asvadi ◽  
Darren R. Jones ◽  
Rosanne D. Dunn ◽  
Andre B.H. Choo ◽  
Matthew J. Raison ◽  
...  

Abstract Despite high dose chemotherapy and autologous stem cell transplant, multiple myeloma (MM) remains an incurable malignancy, with a median 5 year survival of less than 20%. With the exception of idiotype, few antigen targets have been identified that would facilitate specific immunotherapy of MM. We have previously described a murine monoclonal antibody that recognizes a conformation-dependent epitope on free human kappa light chains and a cell surface antigen, KMA, expressed on kappa MM plasma (MMκ) cells (Raison, RL and Boux, HA Mol Immunol 1985 22:1393). Here we show that the murine antibody, designated mKap, bound specifically to a range of MMκ cell lines and inhibited the in vitro growth of these cells. Flow cytometric analysis (Annexin-V and PI staining) of MMκcell lines incubated with mKap demonstrated a dose dependent induction of apoptosis. Furthermore, the presence of activated caspases in mKap treated cells was detected using the CaspACE™ FITC-VAD-FMK reagent. The induction, by mKap, of apoptosis in MMκ cells occured in the absence of cross-linking second antibody or effector cells. In vivo, anti-tumor activity by mKap was demonstrated in a SCID mouse tumor xenograft model. Tumor growth was measured by quantitation of secreted myeloma Ig over a period of 6 weeks. From week 4 onwards, significantly lower serum concentrations of myeloma Ig were detected in animal groups receiving 3.0, 1.5, 0.3 and 0.15 mg total mKap compared to the untreated control (P<0.005 at week 4; P<0.0001 at week 5). The tumor-restricted specificity of mKap, coupled with its ability to inhibit MMκ cell growth in vitro and in vivo, suggests the potential use of either a chimeric or humanised version of this antibody, alone or in combination with chemotherapy, for the treatment of kappa MM.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2523-2523
Author(s):  
Jeffrey W. Strovel ◽  
Tammy Lawrence ◽  
Pachai Natarajan ◽  
Stephen Glanowski ◽  
Irina Lonskaya ◽  
...  

Abstract Multiple Myeloma (MM) is a fatal form of hematologic cancer characterized by the clonal expansion of plasma cells in the bone marrow. Recent finding by several research laboratories have shown that Wnt/β-Catenin signaling pathway is activated in MM leading to the translocation of β-Catenin to the nucleus where it binds to T cell factor (TCF) and drives transcription of genes involved in the progression of cancer (Derksen et. Al., PNAS 2004). The mechanism by which Wnt/β-Catenin signaling is activated in MM is not well understood, however, β-Catenin is expressed in the majority of MM cell lines and inhibition of the pathway with a dominant negative form of TCF4 or the small molecule PKF115-584 have shown anti-proliferative effects in in vitro and in vivo cell line and xenograft models making inhibition of Wnt/β-Catenin signaling a promising modality for the treatment of MM (Sukhdeo et al., PNAS 2007). To this end, Avalon Pharmaceuticals has developed a series of small molecule compounds, lead candidate series-363 (LC-363), with potent inhibitory effects on Wnt/β-Catenin signaling and anti-proliferative effects in colon cancer cell lines and in vivo models (EORTC, 2007). LC-363 compounds down regulate expression of the noted β-Catenin/TCF transcriptional targets c-jun, fra-1, and PPARδ as well as increase expression of the Dickkopf family of secreted proteins 1 and 3 which inhibit Wnt signaling. In this study, we report on the in vitro characteristics of LC-363 in MM cell line models. We show that LC-363 compounds have broad and potent growth inhibitory and cytotoxic effects on MM cell lines. Additionally, these effects are mediated through a G1 phase cell cycle block, apoptosis, and decrease in cytoplasmic levels of β-Catenin. Importantly, we show the relationship between expression level of β-Catenin protein and sensitivity to LC-363 compounds in terms of growth inhibition and apoptosis. Lastly, we describe the gene expression signatures induced by LC-363 in MM cell models across multiple time and dose studies to detail their dynamic effects on expression of genes within the Wnt/β-Catenin signaling pathway. In conclusion, we believe that inhibition of Wnt/β-Catenin signaling is a viable therapeutic alternative for treatment of MM patients with activated Wnt/β-Catenin signaling and to that end; LC-363 compounds are promising candidates currently in pre-clinical development for treatment of cancers with activated Wnt/β-Catenin signaling including MM.


Sign in / Sign up

Export Citation Format

Share Document