identification of dormancy-associated article micrornas for ... · characterized pairs of dormant...

18
Identication of Dormancy-Associated MicroRNAs for the Design of Osteosarcoma- Targeted Dendritic Polyglycerol Nanopolyplexes Galia Tiram, Ehud Segal, Adva Krivitsky, Rony Shreberk-Hassidim, Shiran Ferber, Paula Ofek, Taturo Udagawa, Liat Edry, § Noam Shomron, § Maayan Roniger, Batsheva Kerem, Yuval Shaked, Sarit Aviel-Ronen, #,Iris Barshack, #,Marcelo Calderó n, Rainer Haag, and Ronit Satchi-Fainaro* ,Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel Vertex Pharmaceuticals, Cambridge, Massachusetts 02142, United States § Department of Cell & Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel Department of Genetics, The Life Sciences Institute, Edmond J. Safra Campus, The Hebrew University, Jerusalem 91905, Israel Department of Molecular Pharmacology, Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa 32000, Israel # Department of Pathology, Sheba Medical Center, Tel Hashomer 52621, Israel Talpiot Medical Leadership Program, Sheba Medical Center, Tel Hashomer 52621, Israel Institut fü r Chemie und Biochemie, Freie Universitä t Berlin, Berlin 14195, Germany Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel * S Supporting Information ABSTRACT: The presence of dormant, microscopic cancerous lesions poses a major obstacle for the treatment of metastatic and recurrent cancers. While it is well- established that microRNAs play a major role in tumori- genesis, their involvement in tumor dormancy has yet to be fully elucidated. We established and comprehensively characterized pairs of dormant and fast-growing human osteosarcoma models. Using these pairs of mouse tumor models, we identied three novel regulators of osteosarco- ma dormancy: miR-34a, miR-93, and miR-200c. This report shows that loss of these microRNAs occurs during the switch from dormant avascular into fast-growing angiogenic phenotype. We validated their downregulation in patientstumor samples compared to normal bone, making them attractive candidates for osteosarcoma therapy. Successful delivery of miRNAs is a challenge; hence, we synthesized an aminated polyglycerol dendritic nanocarrier, dPG-NH 2 , and designed dPG-NH 2 -microRNA polyplexes to target cancer. Reconstitution of these microRNAs using dPG-NH 2 polyplexes into Saos- 2 and MG-63 cells, which generate fast-growing osteosarcomas, reduced the levels of their target genes, MET proto- oncogene, hypoxia-inducible factor 1α, and moesin, critical to cancer angiogenesis and cancer cellsmigration. We further demonstrate that these microRNAs attenuate the angiogenic capabilities of fast-growing osteosarcomas in vitro and in vivo. Treatment with each of these microRNAs using dPG-NH 2 signicantly prolonged the dormancy period of fast-growing osteosarcomas in vivo. Taken together, these ndings suggest that nanocarrier-mediated delivery of microRNAs involved in osteosarcoma tumorhost interactions can induce a dormant-like state. KEYWORDS: microRNA, polymeric nanomedicine, hyperbranched polymer, dendrimer, polyplex, tumor dormancy, osteosarcoma T umor dormancy, a state in which tumors exist without expanding in mass beyond 13 mm, has important implications for the early detection and treatment of cancer. Dormant tumors are highly prevalent in the general Received: October 1, 2015 Accepted: January 27, 2016 Published: January 27, 2016 Article www.acsnano.org © 2016 American Chemical Society 2028 DOI: 10.1021/acsnano.5b06189 ACS Nano 2016, 10, 20282045

Upload: others

Post on 11-Aug-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Identification of Dormancy-Associated Article MicroRNAs for ... · characterized pairs of dormant and fast-growing human osteosarcoma models. Using these pairs of mouse tumor models,

Identification of Dormancy-AssociatedMicroRNAs for the Design of Osteosarcoma-Targeted Dendritic PolyglycerolNanopolyplexesGalia Tiram,† Ehud Segal,† Adva Krivitsky,† Rony Shreberk-Hassidim,† Shiran Ferber,† Paula Ofek,†

Taturo Udagawa,‡ Liat Edry,§ Noam Shomron,§ Maayan Roniger,∥ Batsheva Kerem,∥ Yuval Shaked,⊥

Sarit Aviel-Ronen,#,∇ Iris Barshack,#,¶ Marcelo Calderon,⊗ Rainer Haag,⊗ and Ronit Satchi-Fainaro*,†

†Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel‡Vertex Pharmaceuticals, Cambridge, Massachusetts 02142, United States§Department of Cell & Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel∥Department of Genetics, The Life Sciences Institute, Edmond J. Safra Campus, The Hebrew University, Jerusalem 91905, Israel⊥Department of Molecular Pharmacology, Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa 32000,Israel#Department of Pathology, Sheba Medical Center, Tel Hashomer 52621, Israel∇Talpiot Medical Leadership Program, Sheba Medical Center, Tel Hashomer 52621, Israel⊗Institut fur Chemie und Biochemie, Freie Universitat Berlin, Berlin 14195, Germany¶Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel

*S Supporting Information

ABSTRACT: The presence of dormant, microscopiccancerous lesions poses a major obstacle for the treatmentof metastatic and recurrent cancers. While it is well-established that microRNAs play a major role in tumori-genesis, their involvement in tumor dormancy has yet to befully elucidated. We established and comprehensivelycharacterized pairs of dormant and fast-growing humanosteosarcoma models. Using these pairs of mouse tumormodels, we identified three novel regulators of osteosarco-ma dormancy: miR-34a, miR-93, and miR-200c. This reportshows that loss of these microRNAs occurs during theswitch from dormant avascular into fast-growing angiogenic phenotype. We validated their downregulation in patients’tumor samples compared to normal bone, making them attractive candidates for osteosarcoma therapy. Successful deliveryof miRNAs is a challenge; hence, we synthesized an aminated polyglycerol dendritic nanocarrier, dPG-NH2, and designeddPG-NH2-microRNA polyplexes to target cancer. Reconstitution of these microRNAs using dPG-NH2 polyplexes into Saos-2 and MG-63 cells, which generate fast-growing osteosarcomas, reduced the levels of their target genes, MET proto-oncogene, hypoxia-inducible factor 1α, and moesin, critical to cancer angiogenesis and cancer cells’ migration. We furtherdemonstrate that these microRNAs attenuate the angiogenic capabilities of fast-growing osteosarcomas in vitro and in vivo.Treatment with each of these microRNAs using dPG-NH2 significantly prolonged the dormancy period of fast-growingosteosarcomas in vivo. Taken together, these findings suggest that nanocarrier-mediated delivery of microRNAs involved inosteosarcoma tumor−host interactions can induce a dormant-like state.

KEYWORDS: microRNA, polymeric nanomedicine, hyperbranched polymer, dendrimer, polyplex, tumor dormancy, osteosarcoma

Tumor dormancy, a state in which tumors exist withoutexpanding in mass beyond 1−3 mm, has importantimplications for the early detection and treatment of

cancer. Dormant tumors are highly prevalent in the general

Received: October 1, 2015Accepted: January 27, 2016Published: January 27, 2016

Artic

lewww.acsnano.org

© 2016 American Chemical Society 2028 DOI: 10.1021/acsnano.5b06189ACS Nano 2016, 10, 2028−2045

Page 2: Identification of Dormancy-Associated Article MicroRNAs for ... · characterized pairs of dormant and fast-growing human osteosarcoma models. Using these pairs of mouse tumor models,

population. In 1993, a study by Black and Welch demonstratedthat the prevalence of dormant tumors in autopsies ofotherwise healthy individuals ranged from 40 to 100%,depending on the cancer type.1 However, dormant tumorscan be present not only as one of the early stages of tumordevelopment, but also as micrometastases and as recurrentdisease following treatment.2 Although great efforts have beenmade to shed light on the tumor dormancy phenomenon, itsunderlying molecular and cellular mechanisms remain un-clear.2−5 Several mechanisms have been proposed for theescape of tumors from dormancy including immunoescape, anexit from cell cycle arrest and the induction of angiogenesis.6

Failure of a tumor to undergo the “angiogenic switch” andrecruit new vasculature results in a nonangiogenic andnonprogressing dormant tumor.7

Osteosarcoma is the most common primary bone cancer andthe sixth leading cancer in children under the age of 15. Thecurrent standard of care for osteosarcoma patients is surgery,neoadjuvant and adjuvant chemotherapy, which results in a 5-year survival rate of 60−80%. However, many of these patientswill suffer from recurrent disease, either due to minimal residualdisease or the presence of dormant micrometastatic lesions atthe time of diagnosis.8 Understanding the early stages by whichosteosarcomas develop from dormant occult into largevascularized tumors could facilitate the development of newdiagnostic and therapeutic tools for this malignancy.

Among the emerging players in regulation of physiologicaland pathological processes are microRNAs (miRNAs), smallnoncoding RNA molecules which regulate gene expression atthe post-transcriptional level.9 Despite the great advancementsin molecularly targeted therapies in the last two decades,resistance to these medications often arises due to theactivation of compensatory mechanisms that maintain thefunction of alternative targets.10,11 miRNAs have the potentialto overcome these compensatory mechanisms, as a singlemiRNA can regulate entire networks of genes. Numerousstudies have pointed out miRNAs as potential therapeuticagents and targets for cancer. Therefore, we hypothesized thatspecific miRNAs may have a role in the switch from dormancyto progressive disease. We further postulated that unravellingnovel dormancy associated miRNAs will potentially lead to thedevelopment of novel diagnostic and therapeutic tools formalignancies. Several studies have demonstrated dysregulationof miRNAs in osteosarcoma tumorigenesis, metastasis andresistance to chemotherapy.12 Among these miRNAs areknown regulators of other malignancies, including oncomiR-21, miR-34a, and miR-16.13−15 Two recent studies demon-strated the involvement of specific miRNAs in tumordormancy.16,17 Nonetheless, additional research investigatingthe molecular mechanisms governing osteosarcoma dormancyis necessary, in order to translate these insights into therapeuticapplications. Furthermore, the potential therapeutic propertiesof nonself oligonucleotides, e.g., miRNAs, suffer from

Figure 1. Human osteosarcoma dormancy model. (A) Dormancy periods of Saos-2-E and Saos-2-D tumors (n = 5) following subcutaneousinoculation into SCID mice. (B) Representative images of flipped skin of tumor-bearing mice 21 days postinoculation. (C)Immunohistochemical staining of Saos-2-D and Saos-2-E tumors 21 days postinoculation (n = 5). Sections were stained with H&E orstained for microvessels (CD31), proliferating cells (Ki67), and apoptotic cells (Caspase 3). Arrows point to positive staining. Scale barsrepresent 100 μm. (D) Quantification of microvessel density and number of vessels with lumen in size-matched tumors. (E) Contrast-enhanced ultrasound (US) imaging (left) and 3D relative blood volume (right) of matrigel plugs containing Saos-2 cells. (F) Hemoglobinconcentration within matrigel plugs. Data represent mean ± SEM.

ACS Nano Article

DOI: 10.1021/acsnano.5b06189ACS Nano 2016, 10, 2028−2045

2029

Page 3: Identification of Dormancy-Associated Article MicroRNAs for ... · characterized pairs of dormant and fast-growing human osteosarcoma models. Using these pairs of mouse tumor models,

limitations such as degradability in plasma, immunogenicity,lack of membrane-crossing abilities of the oligonucleotides, andinability to escape from the endosome in order to reach theircytoplasmic targets.To overcome these challenges, we have developed dendritic

polyglycerolamine (dPG-NH2), a novel polyglycerol-basednanocarrier to deliver oligonucleotides to tumors in vivo.18,19

This cationic hyperbranched polymer facilitates an efficientdelivery of oligonucleotides by improving their stability,intracellular trafficking, silencing efficacy and accumulation atthe tumor site due to the enhanced permeability and retention(EPR) effect. Furthermore, this novel nanocarrier demon-strated low cytotoxicity, an efficient delivery of active siRNAinto cells and a successful silencing of a luciferase gene,ectopically overexpressed in a human glioblastoma cell line.20

Here, we exploited dPG-NH2-miRNA polyplexes to deliver theselected miRNAs for osteosarcoma therapy. This studyidentifies, validates, and evaluates the therapeutic potential ofthree distinct miRNAs, miR-34a, miR-93, and miR-200c, when

polyplexed to dPG-NH2 nanocarrier in order to regulateosteosarcoma dormancy and progression.

RESULTS

Osteosarcoma Cells Generating Dormant TumorsDisplay Impaired Angiogenic Potential in Vivo. UsingSaos-2 human osteosarcoma cells line, defined as non-tumorigenic by the ATCC, we have developed a model ofosteosarcoma dormancy. When inoculated subcutaneously (sc)into mice, Saos-2 cells generate small masses that remaindormant (Saos-2-D for “dormant”) for a prolonged period oftime until they spontaneously escape by adopting an angiogenicphenotype and grow in mass. From these “escaped” tumors, weisolated a new tumor cell line, which upon reinoculation intomice, rapidly recruits blood vessels and forms establishedvascularized tumors (Saos-2-E for “escape”) (Figure 1A). Toexplore the mechanisms underlying the switch from dormancy,we employed this pair of cell lines and an additional pair ofnontumorigenic MG-63 human osteosarcoma cells and its

Figure 2. Saos-2 cells retain their in vivo phenotypes following orthotopic and intravenous tumor cells inoculation. (A) Dormancy periods ofSaos-2-E and Saos-2-D tumors (n = 5) following orthotopic inoculation into the tibia of SCID mice. (B and C) Micro-CT 3D images (B) andH&E staining (C) of tibias and lungs of mice bearing intratibial Saos-2 tumors 100 days following inoculation. T: Tumor. Scale bars represent100 μm. (D) Kaplan−Meier analysis for survival of mice following intravenous inoculation of Saos-2 cells (D, n = 4; E, n = 5) into the lateraltail vein. (E and F) Ex vivo imaging of GFP-labeled Saos-2 lung colonies (E) and H&E staining of lungs (F) 50 days following intravenousinoculation of tumor cells. Scale bars represent 100 μm.

ACS Nano Article

DOI: 10.1021/acsnano.5b06189ACS Nano 2016, 10, 2028−2045

2030

Page 4: Identification of Dormancy-Associated Article MicroRNAs for ... · characterized pairs of dormant and fast-growing human osteosarcoma models. Using these pairs of mouse tumor models,

counterpart MG-63-E transfected with Ras, which generatesfast-growing tumors.Three weeks following tumor cell inoculation, when tumor

volumes were 1−3 mm3, Saos-2-E tumors recruited new bloodvessels, while Saos-2-D tumors showed no evidence forangiogenesis, as shown by flipping the skin of tumor-bearingmice (Figure 1B). CD31 staining showed that microvesselsdensity and the number of vessels with lumen (i.e., functionalvessels) of 1−3 mm3 Saos-2-E tumors were significantly highercompared with those of size-matched Saso-2-D tumors (Figure1C,D). Despite some CD31-positive staining in Saos-2-Dtumors, the lack of functional vessels prevented the oxygen andnutritional supply necessary for tumor growth and therefore,these tumors remained harmless to the host and asymptomatic.In contrast, vessels with lumens formed in Saos-2-E tumorswere functional and, therefore, supported rapid tumor growth.Moreover, Saos-2-D and Saos-2-E tumors both containedproliferating cells, but almost no apoptotic cells (Figure 1C).

This finding rules out the possibility of “cellular dormancy” asthe underlying cause for the in vivo dormant phenotype.21

Support to the increased in vivo angiogenic potential of Saos-2-E cells and to the differences in blood vessels’ functionalitybetween the pair of cells was found in the matrigel plugangiogenesis assay.22 Three weeks following inoculation of thematrigel/cells mixture, plugs were assessed for neovasculariza-tion. The ability of Saos-2-E cells to recruit functional vesselsinto the plug was significantly increased compared to Saos-2-Dcells, as demonstrated by the high present of contrast agent andhemoglobin concentration within plugs containing Saos-2-Ecells (Figure 1E,F).

Saos-2 Dormancy Model Imitates the Clinical Settingof Osteosarcoma. To rule-out the possibility that the patternof differential tumor growth in this model is a product ofartificial subcutaneous inoculation, Saos-2 cells were inoculatedorthotopically into the tibia and systemically through the tailvein for metastases establishment. Saos-2-E tumors escaped 60days following orthotopic inoculation, whereas Saos-2-D

Figure 3. Saos-2 cells generating dormant and fast-growing osteosarcoma have similar growth, invasion, migration, and DNA replicationkinetics. (A) Growth rate. (B) Invasion through a matrigel-coated chamber or through an endothelial monolayer. Representative images ofinvading cells are shown. Scale bar represents 100 μm. (C) Migration rate of control Saos-2 cells (10% FBS, Normoxia), cells grown in serum-depleted medium and cells subjected to hypoxia (1% O2). Representative images of migrating cells are shown. Scale bar represents 100 μm.(D) Changes in HIF1α and VEGF levels following 24 h hypoxia, quantified by qRT-PCR. (E) Average fork rate (kb/min) of Saos-2-D cellsand Saos-2-E cells. (F) Fork rate (kb/min) distribution of Saos-2-D and Saos-2-E cells (n = 140). (G) Origin density. Average fork distance(kb) of out- and in-going forks in Saos-2-D (n = 95) and Saos-2-E cells (n = 88). Proliferation and migration data is represented in mean ±SD. Replication analysis data is represented in mean ± SEM.

ACS Nano Article

DOI: 10.1021/acsnano.5b06189ACS Nano 2016, 10, 2028−2045

2031

Page 5: Identification of Dormancy-Associated Article MicroRNAs for ... · characterized pairs of dormant and fast-growing human osteosarcoma models. Using these pairs of mouse tumor models,

tumors remained dormant for 140 days (Figure 2A). Thesegrowth patterns were similar to those resulting fromsubcutaneous inoculation. However, while no differences weredemonstrated in growth patterns of Saos-2-E tumors, theescape of Saos-2-D tumors following orthotopic inoculationwas significantly faster (140 days) than following subcutaneousinoculation (240 days). This supports our hypothesis and thegeneral notion that orthotopic tumors, while inoculated in theappropriate microenvironment, gain increased support fromtheir stroma compared to subcutaneously inoculated tu-

mors.23,24 It is possible that this increased stromal supportaffected Saos-2-D tumors to a greater extent, since they requiremore time to “educate” and “reprogram” the microenvironmentand are less self-sufficient compared to Saos-2-E tumors. Inaddition, while the subcutaneous microenvironment is relativelypoorly vascularized, the tibia has a rich blood supply withsizable feeding vessels.25 Therefore, when inoculated into thetibia, Saos-2-D cells are able to recruit blood vessels and toform large angiogenic tumors more rapidly than in thesubcutaneous tissue. Micro-CT imaging performed 100 days

Figure 4. miR-34a, miR-93 and miR-200c downregulation is associated with escape from dormancy and tumor progression of humanosteosarcoma. (A) Heat map of miRNAs overexpressed in Saos-2-D cells, compared with Saos-2-E cells. Highly expressed miRNAs are labeledin red, while poorly expressed miRNAs are labeled in gray, based on the Ct value of each miRNA obtained by qRT-PCR. (B) Relativeexpression of the selected miRNAs in Saos-2 size-matched tumors (1−3 mm3) 3 weeks following inoculation, quantified by qRT-PCR. (C)miR-34a, miR-93, and miR-200c relative expression in 15 primary osteosarcoma (P) and 9 lung metastases (M) FFPE specimens. Data isshown as log2 of fold change of osteosarcoma tumors relative to normal bone FFPE specimens, quantified by qRT-PCR. Group averages areshown in the framed graphs on the right. (D) Representative images and quantification of miR-200c expression by in situ hybridization (ISH).Scale bars represent 20 μm. (E) miR-200c relative expression in Saos-2-D cells and Saos-2-E primary and lung metastases (Saso-2-E M) cells,compared to hFOB human osteoblasts. Data is shown as log2 of fold change of relative hFOB cells, quantified by qRT-PCR. Data representsmean ± SEM.

ACS Nano Article

DOI: 10.1021/acsnano.5b06189ACS Nano 2016, 10, 2028−2045

2032

Page 6: Identification of Dormancy-Associated Article MicroRNAs for ... · characterized pairs of dormant and fast-growing human osteosarcoma models. Using these pairs of mouse tumor models,

following inoculation showed areas of calcification inside andoutside the tibial shaft in tibias inoculated with Saos-2-E cells, aphenomenon typical to osteoid-forming osteosarcoma (Figure2B). Moreover, at this time point, mice bearing Saos-2-Etumors developed calcified pulmonary metastases, the mostcommon manifestation of metastatic osteosarcoma (Figure 2B).Similar clinical manifestations were observed following escapeof Saos-2-D tumors (Figure S1A). Histological analysis 100days postinoculation revealed that tibias inoculated with Saos-2-D cells remained intact and showed presence of dormantmicroscopic lesions that did not metastasize to the lungs.Conversely, Saos-2-E tumors invaded and destroyed the corticalbone and spread into the lungs (Figure 2C).Similar growth kinetics of dormant and fast-growing tumors

was maintained in the experimental lung metastasis model. Fiftydays following systemic intravenous injection of Saos-2-E cells,rapid weight loss and overt metastatic colonization wereobserved. In contrast, mice injected with Saos-2-D cellsexperienced no weight loss and had no apparent lung colonies.Only 200 days following inoculation, dormant lung coloniesescaped and became symptomatic (Figure 2D,E, Figure S1B).H&E staining showed that while lungs of mice inoculated withSaos-2-D cells showed small tumor foci, multiple tumorcolonies were observed in lungs of mice inoculated withSaos-2-E cells (Figure 2F).Osteosarcoma Cells Generating Dormant or Fast-

Growing Tumors Have Similar Growth, Invasion,Migration, and DNA Replication Kinetics. We nextevaluated the in vitro tumorigenic potential of Saos-2-D andSaos-2-E cells. We found no significant differences in theirproliferation rates (Figure 3A). The ability to invade throughmatrigel or endothelial monolayer and migrate toward serumwas also similar in both cell lines (Figure 3B,C). However,following serum-starvation or exposure to hypoxic conditions(1% O2), migration of Saos-2-E cells toward serum wassignificantly enhanced compared with that of Saos-2-D cells(Figure 3C). This suggests that Saos-2-E cells are more capableof overcoming stress conditions, like hypoxia and nutrientdeprivation, which arise during tumor development. To furtherevaluate the response of Saos-2 cells to hypoxia, changes inhypoxia inducible factor α (HIF1α) and its downstream targetvascular endothelial growth factor (VEGF) following hypoxiawere analyzed by qRT-PCR. While in Saos-2-D cells HIF1αlevels were reduced and VEGF levels were not changedfollowing hypoxia, a significant increase in their levels wasobserved in Saos-2-E cells (Figure 3D). Therefore, Saos-2-Ecells have an improved capacity to respond to hypoxicconditions in the tumor microenvironment, a trait that mightexplain the above-mentioned increased angiogenic potential ofSaos-2-E tumors at a microscopic size.In recent years, a number of studies have shown that in early

stages of cancer development, cells are proliferating but fail tosupport normal DNA replication. Hence, the cells areundergoing replication stress leading to genomic instabilityand tumorigenicity.26,27 To test whether the difference in thetumorigenic potential between Saos-2-D and Saos-2-E cellsmight result from differences in their replication dynamics, weused the DNA combing technique which enables to analyze thereplication of single DNA molecules.28 In two independentexperiments, there was no significant change either in thereplication rate of the cells (Figure 3E,F), or in the distancebetween adjacent origins (Figure 3G). Therefore, the differ-

ences observed in vivo cannot be attributed to differences inDNA replication dynamics.

Dormancy-Associated miRNA Are Downregulated inHuman Osteosarcoma Specimens. To explore the basicmechanisms that govern the switch from a dormant to a fast-growing phenotype, Saos-2-D and Saos-2-E cells were analyzedfor their miRNA profile (Figure 4A, Figure S2 and Table S1).Our focus was on miRNAs that were downregulated in fast-growing versus dormant-tumor-forming cells (Figure 4A). Outof those miRNAs, miR-200c, miR-34a, and miR-93 (represent-ing a mild, intermediate, or substantial differential expression,respectively) were selected for further evaluation. Expressionlevels of these miRNAs were further evaluated in size-matched(1−3 mm3) tumors, showing that miR-34a, miR-93, and miR-200c are upregulated in Saos-2-D tumors compared to Saos-2-Etumors also in the in vivo setting (Figure 4B).Next, we set to validate the relevance of dormancy-associated

miRNAs in osteosarcoma clinical specimens. Due to the clinicalinaccessibility of dormant tumors, miRNA levels in primary andmetastatic osteosarcoma specimens were compared with thoseof normal bones. qRT-PCR analysis for miRNA levelsdemonstrated that although expression levels of these threemiRNAs vary between patients (Figure 4C, single patientsdisplayed), there is an overall clear trend of miRNAdownregulation in primary and metastatic osteosarcomaspecimens (Figure 4C, Averages). To note, all samples in ourstudy were of conventional osteosarcoma, defined by TheCollege of American Pathologists as high grade tumors.29

Therefore, the observed variation in miRNA levels most likelydoes not depend upon tumor grade. To evaluate whether theexpression signature correlates with tumor stage, which reflectstumor progression state, we compared the average miRNAlevels of primary tumors (defined as stage II−III) andmetastatic tumors (defined as stage IV). While no significantdifferences were found in miR-34a and miR-93 expression inprimary versus metastatic samples, miR-200c was significantlyhigher in the metastatic specimens, suggesting its role in themetastatic spread of osteosarcoma (Figure 4C, Averages).Interestingly, miR-200c, with the mildest differential expression,showed the most remarkable downregulation in tumor samplescompared to normal bone.Having identified miR-200c as the miRNA which down-

regulation was the most significant, its expression level wasfurther evaluated using anti-miR-200c in situ hybridization(ISH) probe. In accordance with the qRT-PCR results, normalbone samples from clinical specimens expressed higher levels ofmiR-200c compared with primary and metastatic osteosarcomaspecimens (Figure 4D). To test whether a similar miRNAexpression pattern is found in Saos-2 metastases versus primarytumors, we isolated four new cell lines from spontaneous lungmetastases generated from Saos-2-E tumors following intratibialinoculation (see Figure 2B). When comparing miR-200c levelsin Saos-2 cells to their level in hFOB human osteoblasts, i.e.,normal bone cells, a correlation to miR-200c levels in humanspecimens was found. miR-200c was downregulated comparedto normal human osteoblasts in all types of Saos-2 cells (Figure4E), but its level was higher in metastatic Saos-2-E (Saos-2-E-M) cells compared to Saos-2-E cells, similar to the trend seen inhuman specimens.

miR-93 and miR-200c Revert Cells Generating Fast-Growing Tumors into a Dormant NonangiogenicPhenotype. Having shown that cells generating dormantand fast-growing tumors differ mainly in their ability to respond

ACS Nano Article

DOI: 10.1021/acsnano.5b06189ACS Nano 2016, 10, 2028−2045

2033

Page 7: Identification of Dormancy-Associated Article MicroRNAs for ... · characterized pairs of dormant and fast-growing human osteosarcoma models. Using these pairs of mouse tumor models,

to the tumor microenvironment in vivo, we set to evaluate therole of the suspected dormancy-associated miRNAs inosteosarcoma tumor growth and angiogenesis in vivo. miR-93and miR-200c, representing either a high or a low differentialexpression, respectively, were constitutively overexpressed byretroviral infection in Saos-2-E cells, and phenotypic changeswere monitored ex vivo and in vivo.Overexpression of miR-93 and miR-200c prolonged the

dormancy period of Saos-2-E tumors in immunodeficient mice.Saos-2-E and Saos-2-E control miR-infected cells establishedlarge angiogenic tumors within 70 days, whereas Saos-2-E miR-200c, Saos-2-E miR-93, and Saos-2-D tumors remaineddormant for 120, 140, and 220 days, respectively (Figure5A). These changes in the dormancy period correlate with themicroarray results, as miR-200c, with the low differentialexpression, “escaped” faster from dormancy than miR-93 withthe higher differential expression. Furthermore, the mediansurvival of mice bearing Saos-2-E tumors (120 days) wassignificantly extended following overexpression of miR-200c ormiR-93 (∼210 days) (Figure 5B). During these dormancyperiods, cells were detectable by noninvasive intravital CRIMaestro imaging system (Figure 5C). To note, reconstitution

of miRNAs downregulated in Saos-2-D cells (i.e., miR-137,miR-147, and miR-205) did not affect the dormancy periods ofthese cells, defined as “Time to switch” (Figure S2). Thissuggests that the switch from dormancy in our model ispredominantly governed by loss of specific miRNAs.To evaluate the angiogenic activity of Saos-2-E tumors

following miRNA overexpression, we used an ex vivo analysis ofcirculating endothelial progenitor cells (CEPs). CEPs, recruitedfrom the bone marrow during tumor neovascularization,30 canbe utilized as surrogate markers for angiogenic activity.31 Sinceviable CEPs contribute mainly to the early steps of tumorvascularization,32 we evaluated their level 14 days post-inoculation. While there was no difference in viable CEPslevels between nontumor bearing mice and mice bearing Saos-2-D tumors, their level was significantly higher in mice bearingSaos-2-E tumors, suggesting that fast-growing tumors areassociated with increased systemic angiogenic activity (Figure5D). In addition, introduction of miR-93 and miR-200c toSaos-2-E cells reverted the high levels of viable CEPs found inblood of mice bearing Saos-2-E tumors. This suggests that atleast in the context of systemic angiogenesis, fast-growingtumors overexpressing miR-93 and miR-200c became similar to

Figure 5. miR-93 and miR-200c reduce the angiogenic potential and prolong the tumor dormancy period of fast-growing osteosarcoma in vivo.(A) Tumor growth. (B) Kaplan−Meier analysis for survival. P-values were determined using log rank test, compared to control. (C)Noninvasive imaging of GFP-labeled Saos-2 tumors 95 days postinoculation. (D) Levels of circulatory endothelial progenitor (CEP) cells 14days postinoculation. (E) H&E staining and immunohistochemical staining for CD31 and quantification of microvessel density of Saos-2tumors 21 days postinoculation (n = 5). Scale bars represent 50 μm. Data represents mean ± SEM.

ACS Nano Article

DOI: 10.1021/acsnano.5b06189ACS Nano 2016, 10, 2028−2045

2034

Page 8: Identification of Dormancy-Associated Article MicroRNAs for ... · characterized pairs of dormant and fast-growing human osteosarcoma models. Using these pairs of mouse tumor models,

dormant tumors (Figure 5D). Immunostaining for microvesselsin microscopic size-matched tumors showed that Saos-2-Etumors overexpressing miR-93 and miR-200c were almostavascular compared with Saos-2-E control miR tumors (Figure5E).Synthesis and Physicochemical Characterization of

dPG-NH2-miRNA Polyplexes. Retroviral overexpressing ofselected miRNAs into Saos-2 cells generating fast-growingtumors showed that dormancy-associated miRNAs are goodcandidate for osteosarcoma therapy. To further explore theirtherapeutic potential, we decided to exploit the miRNAreplacement approach, to reconstitute miRNAs under-ex-pressed in fast-growing osteosarcomas using miRNA mimics.For this purpose, polyglycerol-amine (dPG-NH2) (Figure 6A)was synthesized as previously described.20 dPG-NH2, synthe-

sized for this study, bears 175 amines/mol polymer, thusenabling electrostatic complexation with the negatively chargedmiRNA (Figure 6B).The ability of dPG-NH2 to complex miRNAs was evaluated

by incubating increasing amounts of polymer with a constantamount of miRNA. The efficacy of polyplex formation wasassessed by electrophoresis mobility shift assay (EMSA),demonstrating complexation starting from an N/P ratio of 9(Figure 6C). This polymer/miRNA ratio (equivalent to 2:1molar ratio) was found to induce maximal miRNA activity andwas therefore chosen for all consecutive experiments. Themorphology of the polyplexes was evaluated by SEM,demonstrating a diameter of 100 nm (Figure 6D,E). dPG-NH2-miRNA polyplexes were further analyzed for theirhydrodynamic diameter and zeta potential using DLS. Mixture

Figure 6. Physicochemical and biological characterization of a dendritic polyglycerol nanocarrier for miRNA delivery. (A) Chemical structureof dPG-NH2. The depicted structure represents an idealized fragment of a 14.5 kDa dPG-NH2 molecule. (B) Theoretical structure of dPG-NH2-miRNA polyplex. (C) EMSA of dPG-NH2-miRNA polyplexes. (D) SEM image of dPG-NH2-miR-93 polyplexes (N/P 9) as obtained byQuanta 200 FEG Environmental SEM. (E) Table summarizing the physicochemical characterization of dPG-NH2-miR-93 polyplexes at amolar ratio of 2:1 (N/P 9). Zeta potential, hydrodynamic diameter and polydispersity index (PDI) were obtained by zetasizer ZS, and theaverage diameter was obtained by Quanta 200 FEG Environmental SEM. (F) Red blood cells lysis assay of dPG-NH2-miR-93 polyplexes (2:1molar ratio). Results are presented as percent of hemoglobin released following 1 h incubation with the different treatments. SDS was used asa positive control and dextran as a negative control. Data represent mean ± SD.

ACS Nano Article

DOI: 10.1021/acsnano.5b06189ACS Nano 2016, 10, 2028−2045

2035

Page 9: Identification of Dormancy-Associated Article MicroRNAs for ... · characterized pairs of dormant and fast-growing human osteosarcoma models. Using these pairs of mouse tumor models,

of miRNA with dPG-NH2 at pH 7.4 yielded polyplexes withpositive charges (17.0 ± 8.68 mV), as expected.20 Thehydrodynamic diameter of the polyplexes was 80.32 ± 23.79nm with a polydispersity index of 0.092 (Figure 6E). This lowpolydispersity suggests that the polyplexes formed are highlystable. Moreover, the particle’s hydrodynamic diameter,measured by SEM and DLS, reflects an assembly of severaldPG-NH2-miRNAs, as previously reported by Fischer andcolleagues.19

To evaluate the physiological biocompatibility of dPG-NH2-miRNA polyplexes, an ex vivo hemolysis assay was performed.

Briefly, rat red blood cells (RBC) solution (2 wt %/wt) wasincubated with serial dilutions of dPG-NH2-miR-93 (2:1 molarratio) and the absorbance of hemoglobin released from theRBCs was measured. The results show that dPG-NH2-miRNApolyplexes were not hemolytic at concentrations up to 5 mg/mL. The positive control of sodium dodecyl sulfate (SDS)showed dose-dependent hemolysis and the negative controldextran showed no hemolysis (Figure 6F).

Dormancy-Associated miRNAs Downregulate Angio-genesis and Migration-Related Pathways. Following aTargetScan and miRDB search for potential targets, we

Figure 7. Dormancy-associated miRNAs target genes essential to cancer angiogenesis and migration. (A) Venn diagram representing the setsof miRNA target genes as predicted by miRDB and TargetScan alone and their overlap. (B) Relative expression of miR-34a, miR-93, and miR-200c target genes in dormant versus fast-growing tumor-generating cells, quantified by qRT-PCR. (C) Relative expression of cMET, HIF1α,and MSN in Saos-2-E and MG-63-E cells following transfection with dPG-NH2-miRNA polyplexes, quantified by qRT-PCR. (D) Sprouting ofendothelial cells from the aortic wall in the presence of conditioned media from Saos-2 or MG-63 cells reintroduced with miRNA mimics.Scale bar represents 100 μm. (E) Relative quantity of angiogenesis-related proteins secreted by Saos-2 cells transfected miRNA mimics. Datawas obtained using an angiogenesis protein array for conditioned media, and is represented as mean pixel density, compared to conditionedmedia of cells introduced with control miR. (F) Changes in HIF1α and VEGF levels following 24 h hypoxia, quantified by qRT-PCR. Datarepresents mean ± SD.

ACS Nano Article

DOI: 10.1021/acsnano.5b06189ACS Nano 2016, 10, 2028−2045

2036

Page 10: Identification of Dormancy-Associated Article MicroRNAs for ... · characterized pairs of dormant and fast-growing human osteosarcoma models. Using these pairs of mouse tumor models,

obtained an extended list, out of which we found targets thathave been previously implicated as regulators of tumorigenesis(Figure 7A). qRT-PCR showed that mRNA levels of threecancer-related target genes are significantly upregulated in Saos-2-E compared to Saos-2-D cells: MET proto-oncogene(cMET), targeted by miR-34a; hypoxia-inducible factor 1α(HIF1α), targeted by miR-93; and moesin (MSN), targeted bymiR-200c (Figure 7B). This upregulation, although seeminglymodest (1.5−2.5 fold change), can have paramount biologicaleffect, as previously shown.33,34 Both cMET and HIF1α wereshown to induce pro-angiogenic signaling pathways incancers.35,36 MSN, a protein localized to filopodia, was reportedto have a role in tumor cells migration and metastatic abilities.37

This data may account for the aforementioned differences inthe angiogenic potential and the stress-induced migratorycapabilities between Saos-2-D and Saos-2-E cells. To confirmthat these genes are targets of our dormancy-associatedmiRNAs in osteosarcoma, we additionally exploited ourabove-mentioned MG-63 model. Target genes mRNA levels

were determined following transfection of Saos-2-E and MG-63-E with dPG-NH2/miRNA mimics polyplexes (200 nM dPG-NH2; 100 nM miRNA). A marked reduction of cMET wasobserved in both cell lines following reconstitution of miR-34a,miR-93, and miR-200c. HIF1α levels were downregulated byreconstitution of miR-93 and miR-200c. miR-200c also reducedMSN levels in both cell lines (Figure 7C).The effect of these miRNAs on the angiogenic phenotype of

osteosarcoma cells was further evaluated using the aortic ringassay. Sprouting of endothelial cells in the presence ofconditioned media from Saos-2-E and MG-63-E cells wassignificantly reduced following transfection with miR-34a ormiR-200c to the same extent as in the presence of conditionedmedia from cells generating dormant tumors, Saos-2-D or MG-63. Transfecting with miR-93 in Saos-2-E cells resulted in asimilar inhibitory effect on endothelial cell sprouting, but didnot affect MG-63-E-induced sprouting (Figure 5D).Using an angiogenesis antibody array, we further evaluated

the effect of miR-34a, miR-93, and miR-200c on the serum

Figure 8. miR-34a, miR-93, and miR-200c inhibit osteosarcoma tumor growth when administered with dPG-NH2 dendritic nanocarrier. (A)Schematic illustration of experimental design. (B) Relative miRNA expression in Saos-2-E tumors following treatment with dPG-NH2-miRNAmimic polypexes, compared to treatment with dPG-NH2-control miR. (C) Antitumor effect of dPG-NH2 complexed with miR-34a, miR-93, ormiR-200c (treatments marked with black arrows). P-value of treatments compared to control at day 64: 0.028 (n = 5). (D) Kaplan−Meieranalysis for survival. P-values were determined using log rank test, compared to control. (E) Body weight change, expressed as percent changefrom the day of treatment initiation. Data represents mean ± SEM.

ACS Nano Article

DOI: 10.1021/acsnano.5b06189ACS Nano 2016, 10, 2028−2045

2037

Page 11: Identification of Dormancy-Associated Article MicroRNAs for ... · characterized pairs of dormant and fast-growing human osteosarcoma models. Using these pairs of mouse tumor models,

levels of a number of angiogenesis-related proteins in Saos-2-Ecells. In general, an antiangiogenic effect was received, with atendency of little to no effect on the secretion of pro-angiogenicproteins and a marked effect on the secretion of antiangiogenicproteins. miR-34a induced the secretion of the antiangiogenicproteins endostatin, transforming growth factor beta 1 (TGF-β1), TIMP metallopeptidase inhibitor 4 (TIMP-4) andthrombospondin-1 (TSP-1). miR-93 reduced the secretion oftissue factor and induced the secretion of endostatin, whilemiR-200c reduced tissue factor secretion and induced thesecretion of endostatin, TGF-β1, TIMP-1, TIMP-4, and TSP-1(Figure 7E).Next, we evaluated the effect of miR-34a, miR-93, and miR-

200c on the in vitro phenotype of Saos-2-E cells. Growth andmigration rates of Saos-2-E cells were not affected byreconstitution of control miR or our miRNAs (FigureS3A,B). No effect on HUVEC migration toward conditionedmedia of cells reconstituted with these miRNAs was observed(Figure S3C). However, following starvation, the ability ofSaos-2-E cells to migrate and to invade through an endothelialcell monolayer was significantly reduced by the dormancy-associated miRNAs (Figure S3D,E). These suggests that thesemiRNAs reduce the ability of Saos-2-E cells to overcome stressconditions.To test this hypothesis, we evaluated the influence of our

miRNAs on the response to an additional stress condition, ahypoxic microenvironment. Our miRNAs interrupted theability of Saos-2-E cells to respond to hypoxia, either byreducing HIF1α levels (by miR-34a and miR-93), or byreducing VEGF levels (by miR-93 and miR-200c) (Figure 7F).HIF1α and VEGF downregulation by miR-93 polyplex duringhypoxia corresponds with the reduction in HIF1α mRNA levelsby miR-93 polyplex, and suggests an important role of thismiRNA in osteosarcoma response to hypoxia. Moreover, thefact that miR-93 and miR-200c polyplexes did not affect VEGFsecretion in normal conditions, as received by the angiogenesisarray (Figure 7E), but significantly reduced VEGF levelsfollowing hypoxia emphasizes our notion that these dormancy-associated miRNAs have a role within the tumor microenviron-ment.Nanocarrier-Directed Delivery of miR-34a, miR-93, or

miR-200c Mimics Using dPG-NH2 Inhibits Saos-2-ETumor Growth. We next evaluated the therapeutic potentialof dPG-NH2-miRNA polylexes in vivo. Tumors wereestablished by subcutaneous inoculation of Saos-2-E cells intoSCID mice. Tumor progression was monitored by calipermeasurement (width × length2 × 0.52) and treatment wasinitiated when tumors reached ∼40 mm3 (Figure 8A). miRNAaccumulation within tumor tissues following treatment with thepolyplexes was validated by qRT-PCR (Figure 8B). dPG-NH2-miR-34a, dPG-NH2-miR-93, and dPG-NH2-miR-200c poly-plexes were all able to convert Saos-2-E tumors into aphenotype that resembles dormant tumors. This dormantstate persisted for a period of ∼40 days, followed by tumorescape, when the effect of the withdrawn treatment stoppedhaving an influence on tumor growth (Figure 8C). As a resultof this dormancy period prolongation, mice median survivalwas substantially extended by 55 days following treatment withmiR-93 and by 74 days following treatment with miR-34a ormiR-200c (Figure 8D). Treatment with the polyplexes did notaffect mice weight to a great extent. The maximum averageweight loss was under 4%, and mice regained their weight

following treatment withdrawal in all treatment groups (Figure8E).

DISCUSSIONSince the discovery of the RNA interference (RNAi)machinery, numerous RNAi-based strategies for therapeuticgene silencing have been developed. RNAi delivery systemswere designed to enable accumulation of oligonucleotides atthe target tissue, facilitating their internalization into the cellsreaching their cytoplasmic target. However, despite numerousattempts, most RNAi systems that reached clinical trials aretargeted to the liver. Moreover, the great promise of RNAitherapeutics has been halted by limitations such as poorpharmacokinetic properties, immunogenicity, inability to crosscellular membranes and properly escape from the endosomes,and high toxicity.38−41 Therefore, greater efforts must be madeto overcome these limitations in order to fully exploit thetherapeutic potential of oligonucleotides.miRNA-based therapies represent a relatively new field, as

most previous efforts were focused on solutions for siRNAdelivery. With the realization that silencing a single pathway incancer results in the development of compensatory mecha-nisms, miRNAs have become attractive candidates as theyregulate entire networks of genes. Therefore, advances inclinical translation of miRNA-based nanomedicines lag behindthose of siRNA-based ones. Currently, there is only oneanticancer miRNA formulation in clinical trial. In fact, the firstcancer-targeted miRNA to enter clinical trials is a liposome-based miR-34a mimic (MRX34) used for patients withhepatocellular carcinoma.We hereby show the rational design of RNAi nanomedicines

starting from the establishment of novel tumor mouse modelsof osteosarcoma, through the discovery and validation of novelkey miRNA players in osteosarcoma progression, up to thecreation of new nanosized miRNA-mimics polyplexes. Usingmodels of osteosarcoma dormancy and clinical specimens, wedemonstrate that (i) tumor growth differences between cellsgenerating dormant and fast-growing tumors are not attributedto differences in replications kinetics; (ii) miR-34a, miR-93, andmiR-200c have a role in the switch of osteosarcoma fromdormancy to progressive disease; (iii) miR-200c is involved inosteosarcoma progression; (iv) miR-34a, miR-93 and miR-200clevels are decreased in metastatic osteosarcoma, both in clinicalsamples and in metastases-derived cells compared to normaltissues and cells; and (v) these dormancy-associated miRNAshave therapeutic potential when polyplexed with a polymericnanocarrier, as opposed to previous published studies thatdemonstrated their in vivo biological effect following ectopicoverexpression.We successfully delivered miR-93, miR-200c, and miR-34a to

human osteosarcoma cells both in vitro and in vivo using dPG-NH2. dPG-NH2 formed a stable complex with these preselectedmiRNAs, of circa 80 nm size and zeta potential of 17.0 ± 8.68mV, at N/P 9, that showed the optimal activity. Our relativelywide distribution in zeta potential might result from thestochastic nature of surface activation of PG dendrimers withamine moieties and the uncontrolled process of self-assembly ofthe miRNA and the positively charged dendrimers. However, itis not uncommon for such polyplexes or their free nanocarriersto have such a wide zeta potential distribution.42,43 Despite thenarrow polydispersity of the polyplex, received by DLSmeasurements, the SEM image showed heterogeneous particledistribution. This might result from the sample preparation

ACS Nano Article

DOI: 10.1021/acsnano.5b06189ACS Nano 2016, 10, 2028−2045

2038

Page 12: Identification of Dormancy-Associated Article MicroRNAs for ... · characterized pairs of dormant and fast-growing human osteosarcoma models. Using these pairs of mouse tumor models,

technique for SEM, in which samples are air-dried. Suchsamples suffer from strong interactions with the substrate andcrystallization artifacts that can lead to altered shape. The DLSmeasurement, on the other hand, analyzes the sample in asolution. This analysis reflects the assembly of particles inconditions that resemble their physiological therapeuticapplications. This method is highly sensitive to heterogeneityand yet have pointed out narrow polydispersity of ourpolyplexes. In addition, it was previously demonstrated thatsamples that showed narrow polydispersity analyzed by DLSexhibited irregular amorphous shape and high polydispersitywhen imaged in dry conditions.44,45 dPG-NH2-miRNApolyplexes (2 mg/kg miRNA with 4 mg/kg dPG-NH2) showedhigh in vivo therapeutic efficacy with low toxicity following eightcourses of treatment. Such treatment dose and regimen are notuncommon and were previously described for miRNA-baseddelivery systems.46−49 Moreover, in the clinic, treatmentregimens of chemotherapeutics and biological drugs for bonemalignancies often include daily administration for a period of5−7 days (e.g., Cyclophosphamide, Ifosfamide, Etoposide)50,51

or longer (e.g., Imatinib, Sunitinib)52,53 up to several months.This suggests that the clinical use of our therapeutic strategy isfeasible. Moreover, evaluation of the effect of increasingconcentrations of our polyplex on RBC revealed that nohemolysis occurred at concentrations up to 5 mg/mL, while therelevant equivalent in vivo dose is 100-fold lower (Figure 6F).In the past two decades, great efforts were made to elucidate

the tumor dormancy phenomenon.2−5,54 Due to the inacces-sibility to dormant lesions in the clinic, most efforts were putinto the establishment and characterization of appropriatedormancy models, which would imitate the clinical setting oftumor development.3,4,54 Here, we postulate that a stablegenetic reprogramming has occurred during the switch ofosteosarcoma to the fast-growing phenotype. This reprogram-ing did not manifest in changes in growth, migration, invasion,and replication kinetics, but rather in pronounced differences intumor growth patterns and angiogenic potentials. This is inagreement with the “angiogenic dormancy” hypothesissuggesting that dormant tumors remain metabolically activeand do not expand due to impaired angiogenesis.3,4,7 Severalstudies demonstrated that the switch from dormancy ispredominantly controlled by the ability to promote angio-genesis.5,54 It is clear that the endothelial involvement is not theonly one, but other stromal compartments such as thoseconsisting of immune cells and cancer-associated fibroblastscontribute to the “stromal switch” toward a symptomaticprogressive disease. Another postulated explanation for thesedifferences, beside the tumor−host interactions, is the enrichedcancer stem cells (CSC) population, otherwise termed “tumorinitiating cell (TIC)”, found among cells generating fast-growing tumors (Figure S4), as CSCs are considered to bemore tumorigenic.55

To date, there is little evidence for the direct involvement ofmiRNAs in tumor dormancy. miR-190, miR-580, and miR-588were recently reported as regulators of tumor dormancy.16

These miRNAs were found to induce a transcriptional switchtoward antiangiogenesis by targeting angiogenesis-relatedpathways. However, data regarding the phenotypic impact ofthese miRNAs on angiogenesis and tumorigenesis is limited.Moreover, thus far, none of the dormancy-associated signaturesreported (genes or miRNAs) were translated into a therapeuticapplication as done here.

In this study, we suggest that miR-34a, miR-93, and miR-200c, upregulated in dormant tumors and healthy bones,prevent tumor progression by reducing the mRNA levels ofgenes critical to tumor angiogenesis and cancer progression ingeneral. These three miRNAs are known tumorigenesisregulators: miR-34a and miR-200c were found to have tumorsuppressive properties,56,57 whereas miR-93 was found to haveeither a promoting or suppressive properties in malignan-cies.58,59 miR-34a and miR-93 have been previously implicatedas regulators of osteosarcoma tumorigenesis.58,60,61 This studyshows a direct link between miR-200c and osteosarcomaprogression and between all three miRNAs and the switch fromtumor dormancy.miR-34a is a known “tumor suppressor” miRNA.56 Several

studies have shown downregulation of miR-34a in osteosarco-ma.60,61 One of the predicted targets of miR-34a, cMET, wasfound to have a key role in osteosarcoma.62 We report here thatcMET, an important contributor to angiogenesis and tumori-genesis, is a putative target of miR-34a in osteosarcoma andindeed was downregulated following introduction of miR-34a.miR-93 was reported to have a differential role in

osteosarcomagenesis.58 Although this study showed miR-93overexpression in several osteosarcoma cell lines compared toosteoblasts, levels of miR-93 in MG-63 and Saos-2 were withinnormal values. This data supports our results, as both MG-63and Saos-2 cell lines are defined as nontumorigenic cells by theATCC. Moreover, they reported no difference in miR-93 levelsbetween primary osteosarcoma tumors and paired normaltissues. Nevertheless, a larger clinical sample is required to fullyunderstand the role of miR-93 in osteosarcoma. So far, noevidence for an antiangiogenic effect of miR-93 was found incancer. We suggest here that the antiangiogenic properties ofmiR-93 in osteosarcoma are derived from inhibition of HIF1αexpression. This factor upregulates expression of proteins thatpromote angiogenesis and other survival factors in severalmalignancies including osteosarcoma.63

Members of the miR-200 family have been extensivelyreported to act as “tumor suppressors” in carcinomas, primarilyby targeting epithelial to mesenchymal (EMT)-related genes.64

miR-200b was found to be upregulated in osteosarcoma cellsfollowing treatment with the anticancer agent diallyl disulfide;65

however, there is no direct evidence for a role of miR-200c inosteosarcoma progression. We show that miR-200c is down-regulated in primary and metastatic osteosarcoma clinicalsamples. Interestingly, both metastatic clinical specimens andSaos-2 lung metastases express higher miR-200c levelscompared to primary tumors. This suggests that loss of miR-200c enables metastasis, and that following metastaticcolonization, miR-200c levels are restored. Similar findingswere previously demonstrated in breast cancer clinicalsamples.66 In terms of therapeutic potential, restoring miR-200c levels might be more beneficial for the treatment ofprimary disease, as it may prevent metastatic spread. A clearantiangiogenic effect of miR-200c was demonstrated in ourstudy, which can be attributed to downregulation of VEGFduring hypoxia. Furthermore, we show here that one of thetargets of miR-200c in osteosarcoma is MSN, a regulator ofmigration and metastases. This data is in line with the above-mentioned differential expression of miR-200c in primary andmetastatic osteosarcoma. Interestingly, another member of theVEGF family, VEGF-C, was reported to promote cancermetastases via upregulation of MSN.67 This suggests that there

ACS Nano Article

DOI: 10.1021/acsnano.5b06189ACS Nano 2016, 10, 2028−2045

2039

Page 13: Identification of Dormancy-Associated Article MicroRNAs for ... · characterized pairs of dormant and fast-growing human osteosarcoma models. Using these pairs of mouse tumor models,

may be a link between the signaling cascades of VEGF andMSN in osteosarcoma.It is important to state that the miRNAs evaluated in this

manuscript represent only a part of the dormancy signature, asthe additional differentially expressed miRNAs are potentialdormancy regulators. The specific role of these miRNAs intumor dormancy will be evaluated in future studies. Moreover,since a single miRNA can regulate entire networks of genes, it isclear that not only the aforementioned miRNA targets areinvolved in osteosarcoma progression. Therefore, the functionof additional targets of these miRNAs in the switch ofosterosarcoma from dormancy will need to be further exploredmechanistically. This will be feasible using appropriatenanocarriers that efficiently and safely deliver preselectedmiRNAs to cancerous tissues.

CONCLUSIONSThe anticancer therapeutic benefit of miRNA-based therapieshas been reported previously in numerous studies.68 Exploitingour well-characterized dendritic nanocarriers,20 we showed thetherapeutic efficacy of dormancy-associated miRNAs. Uncover-ing novel dormancy regulators can be harnessed to treat tumorsfollowing their escape from dormancy and has the potential tobe used as a prophylactic measure for specific risk groups (i.e.,patients bearing a molecular signature and those in risk forrecurrence originating from a dormant metastasis). Anadditional potential benefit of dormancy-promoting therapeu-tics is prolongation of the therapeutic window, giving anenhanced opportunity to kill tumor cells using chemo-therapeutics as dormant tumors consists of highly proliferatingcells.In summary, our report combines experimental and clinical

studies to provide insights on the molecular mechanismsinvolved in osteosarcoma dormancy and tumor progression. Byimplementing this approach on other malignancies, newpotential candidates for therapeutic, diagnostic and prognosticapplications for cancer can be revealed and targeted utilizingPG-based nanocarriers.

METHODSCell Culture. Saos-2 and MG-63 human osteosarcoma cell lines

and human embryonic kidney 293T cells (HEK 293T) were obtainedfrom the American Type Culture Collection (ATCC, Manassas, VA,USA). Green fluorescent protein (GFP)-labeled Saos-2 cell line wasestablished as previously described.4 MG-63-Ras cells were establishedby transfection with activated Ras as previously described,4 in order togenerate an in vivo fast-growing angiogenic cell line (MG-63-E). Cellswere cultured in Dulbecco’s modified Eagle’s medium (DMEM)supplemented with 10% fetal bovine serum (FBS), 100 μg/mLpenicillin, 100 U/mL streptomycin, 12.5 U/mL nystatin, 2 mM L-glutamin (Biological Industries, Kibbutz Beit Haemek, Israel). Humanumbilical vein endothelial cells (HUVEC) were isolated as previouslydescribed69 and cultured in M199 medium (Biological Industries,Kibbutz Beit Haemek, Israel), supplemented with 20% FBS, 100 mg/mL penicillin, 100 U/mL streptomycin, 12.5 U/mL nystatin, 2 mM L-glutamin, 100 mg/mL heparin, and 50 mg/mL of endothelial cellgrowth supplement (Biomedical Technologies, Inc., MA, USA). Cellswere grown at 37 °C in 5% CO2.hFOB 1.19 human osteoblasts were obtained from the ATCC and

cultured in 1:1 mixture of phenol-free Dulbecco’s modified Eagle’smedium/Ham’s F-12 medium (DMEM/F12) supplemented with 10%FBS, 2 mM L-glutamin and 0.3 mg/mL G418. Cells were grown at 34°C in 5% CO2.Animal Studies. All animals were housed in Tel Aviv University’s

animal facility and the experiments were approved by our institutional

animal care and use committee (IACUC) and conducted inaccordance with NIH guidelines.

For subcutaneous inoculation, 6−8 weeks old SCID male mice wereinoculated into the right flank with 1 × 106 Saos-2 cells (n = 5).Tumor progression was monitored by caliper measurement (width ×length2 × 0.52) and by CRI Maestro noninvasive intravital imagingsystem. Two weeks post inoculation, retro-orbital blood was collectedfor analysis of circulatory endothelial progenitor (CEP) cells. Micewere euthanized when tumor size reached 1.5 cm3 or when micebecame moribund.

For orthotopic intratibial tumor cell inoculation, 25 × 104 Saos-2cells were injected into the right proximal tibia of 4 weeks-old maleSCID mice. Briefly, mice were anesthetized using ketamine (100 mg/kg) and xylazine (12 mg/kg) and treated with a depilatory cream(Veet) to expose the anterior surface of the right tibia and the kneejoint. The knee of the mice was flexed to a 90° position and 50 μL ofthe cell suspension was injected into the right tibia using 27 G needle.The needle was inserted into the proximal tuberosity of the tibia usinga rotational (drilling) motion of the syringe. Tumor growth andmetastatic spread were monitored weekly using TomoScope SynergymicroCT scanner (CT imaging, Germany). Mice were euthanizedwhen tumor size reached 1 cm3 or when mice became moribund andimmediately perfused intracardially with PBS followed by 4%formaldehyde.

For the experimental lung metastasis model, 5 × 105 Saos-2 cellswere injected into the tail vein of 6−8 weeks old SCID male mice.Mice were monitored for weight change 3 times a week andeuthanized when a rapid weight loss was observed (15−20% within afew days). Immediately following euthanasia, mice were perfusedintracardially as mentioned above. Lungs were then imaged ex vivo formetastatic colonization using CRI Maestro imaging system.

Intravital Noninvasive Imaging of GFP-Labeled Saos-2Tumors. CRI Maestro noninvasive fluorescence imaging system wasused to follow tumor progression of mice bearing subcutaneous GFP-labeled tumors. Mice were anesthetized using ketamine (100 mg/kg)and xylazine (12 mg/kg), treated with a depilatory cream (Veet) andplaced inside the imaging system. Multispectral image-cubes wereacquired through 450−720 nm spectral range in 10 nm steps usingexcitation (465 nm long-pass) and emission (515 nm long-pass) filterset. Mice autofluorescence and undesired background signals wereeliminated by spectral analysis and linear unmixing algorithm.

Micro-CT Scanning and Analysis. TomoScope SynergymicroCT scanner (CT imaging, Germany) was used to monitorSaos-2 tumor progression following intratibial inoculation. Mice wereanesthetized using ketamine (100 mg/kg) and xylazine (12 mg/kg)and placed inside the imaging system. Data was acquired using 360°individual projection collected every 1° to complete one rotationaround the animal, with X-ray tube voltage of 40 kV. Cross-sectionalimages (DICOM format) were generated from the projected CTimages using TomoScope image reconstruction software (CT imaging,Germany). Tumor volume was calculated from DICOM images usingthe manufacturer’s software.

In Vivo Matrigel Plug Angiogenesis Assay. To analyze theangiogenic potential of Saos-2 lines in vivo, we used the matrigel plugmodel system.70 A total of 1 × 106 cells were mixed with growthfactor-reduced liquefied matrigel (BD Bioscience, NJ, USA) andinjected sc (final volume, 0.5 mL) into the flanks of 6-week-old maleSCID mice. Three weeks post matrigel inoculation, vascularizationwithin the plugs was evaluated by microbubble contrast-enhancedultrasound imaging. Mice were then euthanized and plugs wererecovered and photographed. Hemoglobin content within the plug wasmeasured using Drabkin’s reagent (Sigma-Aldrich, MO, USA).

Microbubble Contrast-Enhanced Ultrasound Imaging. Ultra-sound was performed using a Vevo2100 from VisualSonics, Inc.(Toronto, Ontario, Canada) using the 55 MHz 708 probe. Mice wereanesthetized and hair on the tumor area was removed using adepilatory cream. Nontargeted microbubbles (VisualSonics, Inc.,Toronto, Ontario, Canada) were mixed with saline and injected intothe tail vein of mice. 3D contrast enhanced cine loop of the tumor wasacquired using the 3D acquisition motor immediately after injection.

ACS Nano Article

DOI: 10.1021/acsnano.5b06189ACS Nano 2016, 10, 2028−2045

2040

Page 14: Identification of Dormancy-Associated Article MicroRNAs for ... · characterized pairs of dormant and fast-growing human osteosarcoma models. Using these pairs of mouse tumor models,

Following microbubbles destruction, a second 3D contrast cine loopwas taken. The difference in video intensity from subtraction of thepre- and postdestruction image frames was automatically displayed bythe software as a colored (green) overlay on the grayscale images.Images were analyzed offline for 3D relative blood volume (PA) insidethe tumor using the Vevo2100 imaging software.Immunohistochemistry. FFPE samples of tumor nodules were

sectioned at 5 μm thick. One section per sample was deparaffinized,rehydrated, and stained by hematoxylin and eosin (H&E). Additionalsections were stained by immunohistochemistry for microvessels usingCD31 antibody (Dianova), proliferating cells using Ki67 (Abcam) andapoptotic cells using cleaved caspase 3 antibody (Epitomics). Briefly,slides were pretreated with 10 mM citrate, pH 6.0, for 20 at 95 °C. Allfurther steps were performed at room temperature (RT) in a hydratedchamber. Slides were covered with 3% H2O2 in methanol for 10 min toquench endogenous peroxidase activity, followed by incubation with2% goat serum for 30 min to block nonspecific binding sites. Primaryrat anti-mouse CD31 (1:10; Dianova, Hamburg, Germany) antibodywas applied in 1% BSA at RT for 2 h. Rabbit anti-human Ki67 (1:100;Abcam, MA, USA) and rabbit monoclonal anti-caspase 3 (1:100;Epitomics, CA, USA) antibodies were applied in 1% BSA at RT for 1h. A broad spectrum biotinylated secondary antibody was added for 1h for CD31 or 30 min for Ki67 and caspase 3. Slides were thenincubation with streptavidin−horseradish peroxidase conjugate for 30min (Histostain, Life Technologies, CA, USA). Following furtherwashing, immunoperoxidase staining was developed using ImmPACTDAB diluent kit (Vector Laboratories, CA, USA) per themanufacturer’s instructions and counterstained with hematoxylin.Microvessel density (MVD) was calculated as previously described.71

Briefly, tumors were scanned for areas of high vessel density (i.e., “hotspots”) at low power. Then, individual microvessels were counted at ahigher power (×200 field). Vessels with lumen were defined as vesselspositively stained for CD31, with an open lumen that, in most cases,contained red blood cells and were counted at high power (×200field).Growth Curve. Saos-2 cells were plated into 24-well plates in

triplicates (1 × 104 cells/well). Following incubation (37 °C; 5%CO2), cells were counted daily by Coulter Counter (BeckmanCoulter) over a 4-day period.Migration Assays. Cell migration assays were performed using 8

μm Transwell inserts (Costar Corp., Corning, NY, USA). Cells (1 ×105) were added to the upper chamber of the Transwell. Following 2 hincubation, cells were allowed to migrate for an additional 4 h to thelower chamber of Transwell inserts containing DMEM supplementedwith 10% FBS or conditioned media. Cells were then fixed with ice-cold methanol and stained using hema 3 stain system (Thermo FisherScientific, MA, USA). The stained migrated cells were imaged usingNikon TE2000E inverted microscope integrated with Nikon DS5cooled CCD camera by 10× objective, brightfield illumination. Totalarea of migrated cells from the captured images per membrane wasanalyzed using NIH ImageJ software.Transendothelial Migration Assay. HUVEC (25 × 104 cells)

were grown to confluency in the upper chamber of 24-well Transwellinserts precoated with 10 μg/mL fibronectin. Following 24 hincubation, HUVEC monolayer was washed and 5 × 104 GFP-labeledSaos-2 cells were added to the upper chamber. After 1 h of incubation,DMEM supplemented with 10% FBS was added to lower chamber.Cells were allowed to migrate to the lower compartment for anadditional 6 h and then fixed using 3.7% formaldehyde. Thefluorescence of the migrated cells was imaged using Nikon TE2000Einverted microscope integrated with Nikon DS5 cooled CCD cameraby 10× objective. Fluorescence signal was measured using NIH ImageJsoftware.Molecular Combing. The method was performed as described in

Bester et al.27 Briefly, unsynchronized cells were sequentially pulselabeled by two thymidine analogues: iododeoxyuridine (IdU) andchlorodeoxyuridine (CldU), 50 μM for 30 min each. At the end of thelabeling period, the DNA was extracted, combed and visualized byfluorescent antibodies. The length of the replication signals and thedistances between origins were measured in micrometers and

converted to kilobases according to a constant and sequence-independent stretching factor (1 μm = 2 kb), as previously reported.28

For the replication rate analysis, a minimum of 140 signals wereanalyzed for each sample, containing both the IdU and CldU signals.The distance between origins was extracted by analyzing the distancebetween out- and in-going forks. The analysis was performed on aminimum of 88 forks and higher. All combing experiments wereperformed twice and yielded similar results. Representative data fromone set of the experiments are shown and expressed as mean ± SEvalues.

MicroRNA Arrays. Saos-2 cells generating dormant and fast-growing tumors were analyzed for their miRNA profile as previouslydescribed.72 miRNA relative levels were calculated based on thecomparative threshold cycle (Ct) method. Candidate miRNAs wereselected according to a differential expression of over 15-fold betweencells that generate dormant versus fast-growing tumors. Suspectedtarget genes associated with regulation of angiogenesis and onco-genesis were predicted using TargetScan and miRDB databases.

Quantitative Real-Time PCR. Total RNA was isolated fromcultured cells using EZ-RNA II total RNA isolation kit (Biologicalindustries, Kibbutz Beit Haemek, Israel), according to themanufacturer’s protocol. For total RNA isolation from FFPE tissues,RecoverAll Total Nucleic Acid Isolation Kit (Ambion, TX, USA) wasused, according to the manufacturer’s protocol. One microgram ofRNA was reverse transcribed using miScript II RT kit (Qiagen, CA,USA). Expression levels of miR-34a, miR-93, and miR-200c and theirtarget genes (NOTCH1, BCL2, MET, STAT3, ADAM9, HIF1α,ZEB1, PLCG1, MSN) were analyzed using SYBR green based real-time PCR. miRNAs expression levels were quantified with miScriptPrimer assays (Qiagen, CA, USA) and normalized to RNU6. miRNAtarget genes levels were quantified using custom qPCR primers(Syntezza Bioscience Ltd., Jerusalem, Israel) and normalized toGAPDH.

Formalin-Fixed Paraffin-Embedded (FFPE) Human Osteo-sarcoma Specimens. FFPE samples were obtained with the approvalof the Institutional Review Board (IRB) and in compliance with alllegal and ethical considerations for human subject research. A total of34 FFPE samples were collected from Sheba Medical Center tissuearchive: 15 samples of primary osteosarcoma, 9 samples ofosteosarcoma lung metastases and 10 control samples from patientswho underwent acute, traumatic injuries to the long bones. Allhistological slides were reviewed by the study pathologists. FFPEsamples were analyzed for miRNA levels by qRT-PCR and for miRNAlocalization by in situ hybridization.

miRNA in Situ Hybridization (ISH) on FFPE Specimens.miRNA ISH was performed using digoxigenin-labeled miRCURYLNA ISH probes (Exiqon, Vedbaek, Denmark), according to themanufacturer’s protocol with slight modifications. Briefly, 6 μmsections were collected from each FFPE sample. Sections weredeparaffinized, rehydrated and then permeabilized with proteinase K(10 μg/mL, Roche Diagnostics, IN, USA) for 10 min at 37 °C.Sections were then applied with hybridization mix and hybridizedovernight in a humidified chamber. Following hybridization, sectionswere washed with sodium chloride sodium citrate (SSC) buffer(Ventana Medical Systems, AZ, USA), blocked for 1 h in blockingsolution (Roche), and then incubated for 2 h in anti-DIG-alkalinephosphatase (AP) antibody (Roche Diagnostics, IN, USA) at roomtemperature. Following further washing, sections were incubated withAP substrate until desired sensitivity was achieved and counterstainedwith fast nuclear red.

miRNA Stable Overexpression. Stable miRNA overexpressionwas performed using pCDH-CMV-hygromycin retroviral vectorscontaining the miRNA genomic region or scrambled DNA (SystemsBiosciences, CA, USA). HEK 293T cells were transfected with thespecific vector using pPACKH1 HIV LentiVector Packaging kit(Systems Biosciences). Forty-eight hours following transfection,retroviral particles containing supernatant were collected. Saos-2cells were then infected with the viral particles and selected for stableexpression using hygromycin.

ACS Nano Article

DOI: 10.1021/acsnano.5b06189ACS Nano 2016, 10, 2028−2045

2041

Page 15: Identification of Dormancy-Associated Article MicroRNAs for ... · characterized pairs of dormant and fast-growing human osteosarcoma models. Using these pairs of mouse tumor models,

Measurement of Circulating Endothelial Precursors (CEPs)by Flow Cytometry. Blood was obtained from anesthetized mice byretro-orbital sinus bleeding. Briefly, a capillary pipet containinganticoagulant (0.1 M EDTA) was inserted in the lateral canthus andblood was collected from the retro-orbital sinus. After collection, thepipet was removed and bleeding stopped when the eye returned to anormal position. EDTA anticoagulated blood samples were used toobtain a complete blood count. Samples were kept cold (4 °C) at alltimes and subsequently counted. Blood samples (10 μL) were mixedwith TURK solution (90 μL in DDW supplemented with 1% aceticacid). White blood cells were counted by a Z1 Coulter ParticleCounter (Beckman Coulter) for assay normalization. CEPs and CECswere quantitated using flow cytometry, as described previously.73

Briefly, monoclonal antibodies were used to detect all populations withthe following antigenic phenotypes: for CEPs, CD13+/VEGFR2+/CD117+/CD45−/dim; for detecting CECs, CD13+/VEGFR2+/CD45−/dim. Nuclear staining was used in some experiments toexclude platelets or cellular debris. 7-Aminoactinomycin D (7AAD)was used to distinguish apoptotic and dead cells from viable cells. Afterred cell lysis, cell suspensions were analyzed. After acquisition of atleast 200 000 cells per sample, analyses were considered informativewhen an adequate number of events (i.e., >50, typically 50−150) werecollected in the CEP or CEC enumeration gate in untreated controlanimals. Percentages of stained cells were determined and comparedwith appropriate negative controls. Positive staining was defined asbeing greater than nonspecific background staining. Flow cytometrystudies were performed on Cyan ADP flow cytometer (BeckmanCoulter) and analyzed with Summit (Beckman Coulter) software. Allmonoclonal antibodies were purchased from BD Biosciences orBioLegend and used for flow cytometry analysis in accordance with themanufacturer’s protocols.Dendritic Polyglycerolamine (dPG-NH2) Synthesis. Dendritic

polyglycerol with average molecular weight of 14.5 kDa (PDI = 1.8)was prepared following literature procedures.74,75 Synthesis of dPG-NH2 analogues was executed according to a three-step protocol aspreviously reported.76 To convert 90% of all hydroxyl groups on dPGinto amines, the hydroxyl groups were first activated usingmethanesulfonyl chloride. Mesyl groups were substituted by azidesin a successive step. Finally, reduction of the azides bytriphenylphosphine via Staudinger reduction resulted in formation ofthe desired amines. For optimal purification, extensive dialysis wascarried out after each reaction step. Quantification of functionalizationdegree was accomplished by 1H NMR spectroscopy. 1H NMR (300MHz, CD3OD): δ = 3.31−2.40 functionalized PG groups), 4.01−3.21(PG). 13C NMR (75 MHz, CD3OD): δ = 83.0−65.5 (PG), 55.5−43.6functionalized PG groups).Electrophoresis Mobility Shift Assay (EMSA). The optimal

ratio for the polyplex formation was studied by electrophoreticmobility shift assay (EMSA) as described previously.20 In brief, 100pmol of miR-93 was incubated with dPG-NH2 at 0.5, 2, 4.5, 9, and 18N/P ratio (equivalent to 0.12:1, 0.5:1, 1:1, 2:1, and 4:1 molar ratios ofcarrier to miRNA) for 20 min at RT. Mobility of free and nanocarrier-complexed miRNA was then analyzed by agarose gel electrophoresis.N/P ratios were calculated according to standard formulas reported inthe literature.77,78

Scanning Electron Microscope (SEM). Polymer solution at 0.1mg/mL concentration was mixed with miRNA solution at theindicated molar ratio and incubated at room temperature for 20min. Samples were dropped on a Si wafer and air-dried for 30 min.The dry samples were coated with 4 nm layer of Cr. SEM images weretaken by Quanta 200 FEG Environmental SEM (FEI, OR, USA) athigh vacuum and 5.0 kV.Dynamic Light Scattering (DLS) and Zeta Potential

Determination. The mean hydrodynamic diameter of the dPG-NH2-miR-93 polyplex and the zeta-potential measurements wereperformed using a ZetaSizer Nano ZS instrument with an integrated 4mW He−Ne laser (λ = 633 nm; Malvern Instruments Ltd., Malvern,Worcestershire, U.K.). dPG-NH2-miR-93 polyplexes were prepared bydissolving 0.25 or 0.05 mg of dendrimer and the indicated molar ratioof miR-93 in 1 mL of DDW for the hydrodynamic radius

measurement or the zeta potential measurement, respectively. Sampleswere incubated for 20−30 min at room temperature, then PBS wasadded from ×10 stock to a final buffer concentration of 15 mM, pH =7.4, to the zeta potential sample. All measurements were performed at25 °C using polystyrol/polystyrene (10 × 4 × 45 mm) cell for DLSanalysis and folded capillary cell (DTS 1070) for zeta potentialmeasurements. Results are representative of 3 repeats.

Hemolysis Assay. Rat red blood cells (RBC) solution (2 wt%/wt) was incubated with serial dilutions of dPG-NH2-miR-93polyplexes (2:1 molar ratio) for 1 h at 37 °C. The highest polyplexconcentration (5000 μg/mL) is 100-fold higher than the one used forthe in vivo experiments (50 μg/mL), adjusted to dilution in mouseblood volume. Sodium dodecyl sulfate (SDS) was used as a positivecontrol and dextran (Mw ∼ 70 kDa) as a negative control. Followingcentrifugation, the supernatants were transferred to a new plate andabsorbance was measured at 550 nm using a SpectraMax M5e platereader (Molecular Devices, CA, USA). The results were expressed aspercentage of hemoglobin released by 1 wt %/vol solution of TritonX100 (100% lysis).

miRNA Transfection. miRNAs (100 nM) (Table S2) wereallowed to form a polyplex with PG-NH2 (200 nM; 2:1 molar ratio) inserum free medium for 20 min at room temperature. Saos-2 or MG-63cells were added with PG-NH2-miRNA polyplexes and incubated for48 h at 37 °C in 5% CO2. Cells were analyzed for miRNA levels andtarget genes mRNA levels using qRT-PCR.

Conditioned Media Preparation. The angiogenic properties ofosteosarcoma cell lines were evaluated by the effect of theirconditioned media on the sprouting of endothelial cells from theaortic wall. To generate conditioned media, Saos-2 cells (7.5 × 105) orMG-63 cells (1.2 × 106) were plated on 10 cm2 tissue culture plates.Twenty-four hours later, cells were transfected with 100 nM miRNAcomplexed with 200 nM PG-NH2. Following 48 h incubation at 37 °Cin 5% CO2, conditioned media were harvested and filtered through0.45 μm syringe filter to remove cells and debris.

Aortic Ring Sprouting Assay. Aorta was resected from a BALB/cmouse, sliced to 1 mm pieces and placed in 48 wells plate precoatedwith matrigel (BD Bioscience, NJ, USA) basement membrane (250μL/well; 10 mg/mL). Additional matrigel basement membrane (100μL/well; 10 mg/mL) was added and allowed to polymerize at 37 °Cfor 30 min. Conditioned media from Saos-2 or MG-63 cells (300 μL)was then added. Sprouting of endothelial cells from the resected aortaslices was imaged following 7 days incubation in 37 °C using NikonTE2000E inverted microscope integrated with Nikon DS5 cooledCCD camera by 15× objective, brightfield illumination.

Quantification of Serum Levels of Angiogenesis-RelatedFactors. Saos-2 cells were evaluated for serum levels of angiogenesis-related factors using a human angiogenesis protein array (R&Dsystems, MN, USA), according to the manufacturer’s instructions.Briefly, Saos-2-E cells were transfected with 100 nM miRNA or controlRNA and allowed to grow for 72 h. Conditioned media was thencollected and concentrated using 3K NMWL Centricon centrifugalfilters (Millipore, MA, USA) to a final volume of 500 μL. Angiogenesisarray membranes were added with conditioned media and furtherprocessed according to the kit procedure. Data is represented as meanpixel density, compared to conditioned media of cells introduced withcontrol miR.

Antitumor Activity of dPG-NH2-miRNA-Mimic Polyplexes.dPG-NH2 dendritic nanocarrier, synthesized and characterized aspreviously described,19,20 was used to deliver miR-34a mimic, miR-93mimic or miR-200c mimic (Thermo Fisher scientific, MA, USA) totumor-bearing mice. miRNA mimic sequences were modified forenhanced stability as previously described79 (Table S2). Briefly, micebearing 40 mm3 Saos-2-E tumors were injected intratumorally eighttimes, every 3 days, with miRNA-mimic/negative control miRNAcomplexed with dPG-NH2 (4 mg/kg dPG-NH2 with 2 mg/kgmiRNA) or with saline (n = 5 mice/group). Animal were monitoredtwice a week for general health, body weight and tumor volume.

Sphere-Forming Assay. Six-well plates were coated with poly(2-hydroxyethyl methacrylate) (30 mg/mL in ethanol) and left overnightat room temperature until the ethanol evaporated. Saos-2 cells were

ACS Nano Article

DOI: 10.1021/acsnano.5b06189ACS Nano 2016, 10, 2028−2045

2042

Page 16: Identification of Dormancy-Associated Article MicroRNAs for ... · characterized pairs of dormant and fast-growing human osteosarcoma models. Using these pairs of mouse tumor models,

then plated (50 000 cells/well) in knockout DMEM (LifeTechnologies, CA, USA) supplemented with 20% FBS, 10%nonessential amino acids, 100 ng/mL human epidermal growth factor(Peprotech, NJ, USA), 100 ng/mL human basic fibroblast growthfactor (Peprotech, NJ, USA) and 10 ng/mL stem cells factor(Peprotech,, NJ, USA). Cells were cultured in 5% CO2 at 37 °C fora week.Statistical Methods. Data are expressed as mean ± standard

deviation (SD) for in vitro assays or ±stanard error of the mean (SEM)for in vivo assays. Statistical analysis for two sets of data was performedusing an unpaired t test. For more than two data sets, one-way analysisof variance (ANOVA) was performed followed by a post-hoc test.Statistical significance of differences in overall survival was determinedusing log-rank test. Significance was defined as P < 0.05.

ASSOCIATED CONTENT

*S Supporting InformationThe Supporting Information is available free of charge on theACS Publications website at DOI: 10.1021/acsnano.5b06189.

Additional experimental data as Figures S1−S4 andTables S1 and S2 (PDF)

AUTHOR INFORMATION

Corresponding Author*E-mail: [email protected].

NotesThe authors declare no competing financial interest.

ACKNOWLEDGMENTS

The Satchi-Fainaro laboratory’s research leading to these resultshas received partial funding from the European ResearchCouncil under the European Union’s Seventh FrameworkProgramme (FP/2007-2013)/ERC Consolidator Grant Agree-ment n. [617445], the Israel Science Foundation (Grant No.1309/10, 918/14), Swiss Bridge Award, Israel Cancer ResearchFoundation (ICRF). We thank Cathleen Schlesener for thepreparation of polyglycerolamine and Dr. Camila Avivi for herprofessional assistance with the histology analysis.

REFERENCES(1) Black, W. C.; Welch, H. G. Advances in Diagnostic Imaging andOverestimations of Disease Prevalence and the Benefits of Therapy. N.Engl. J. Med. 1993, 328, 1237−1243.(2) Udagawa, T. Tumor Dormancy of Primary and SecondaryCancers. APMIS 2008, 116, 615−628.(3) Achilles, E. G.; Fernandez, A.; Allred, E. N.; Kisker, O.; Udagawa,T.; Beecken, W. D.; Flynn, E.; Folkman, J. Heterogeneity ofAngiogenic Activity in a Human Liposarcoma: A Proposed Mechanismfor ″No Take″ of Human Tumors in Mice. Journal of the NationalCancer Institute 2001, 93, 1075−1081.(4) Udagawa, T.; Fernandez, A.; Achilles, E. G.; Folkman, J.;D’Amato, R. J. Persistence of Microscopic Human Cancers in Mice:Alterations in the Angiogenic Balance Accompanies Loss of TumorDormancy. FASEB J. 2002, 16, 1361−1370.(5) Satchi-Fainaro, R.; Ferber, S.; Segal, E.; Ma, L.; Dixit, N.; Ijaz, A.;Hlatky, L.; Abdollahi, A.; Almog, N. Prospective Identification ofGlioblastoma Cells Generating Dormant Tumors. PLoS One 2012, 7,e44395.(6) Almog, N. Molecular Mechanisms Underlying Tumor Dormancy.Cancer Lett. 2010, 294, 139−146.(7) Gimbrone, M. A., Jr.; Leapman, S. B.; Cotran, R. S.; Folkman, J.Tumor Dormancy in Vivo by Prevention of Neovascularization. J. Exp.Med. 1972, 136, 261−276.

(8) American Cancer Society. What Are the Survival Rates forOsteosarcoma? http://www.cancer.org/cancer/osteosarcoma/detailedguide/osteosarcoma-survival-rates (accessed December 2015).(9) Bartel, D. P. Micrornas: Genomics, Biogenesis, Mechanism, andFunction. Cell 2004, 116, 281−297.(10) Logue, J. S.; Morrison, D. K. Complexity in the SignalingNetwork: Insights from the Use of Targeted Inhibitors in CancerTherapy. Genes Dev. 2012, 26, 641−650.(11) Holohan, C.; Van Schaeybroeck, S.; Longley, D. B.; Johnston, P.G. Cancer Drug Resistance: An Evolving Paradigm. Nat. Rev. Cancer2013, 13, 714−726.(12) Zhou, G.; Shi, X.; Zhang, J.; Wu, S.; Zhao, J. Micrornas inOsteosarcoma: From Biological Players to Clinical Contributors, aReview. J. Int. Med. Res. 2013, 41, 1−12.(13) Ziyan, W.; Shuhua, Y.; Xiufang, W.; Xiaoyun, L. Microrna-21 IsInvolved in Osteosarcoma Cell Invasion and Migration. Med. Oncol.2011, 28, 1469−1474.(14) Yan, K.; Gao, J.; Yang, T.; Ma, Q.; Qiu, X.; Fan, Q.; Ma, B.Microrna-34a Inhibits the Proliferation and Metastasis of Osteosarco-ma Cells Both in Vitro and in Vivo. PLoS One 2012, 7, e33778.(15) Jones, K. B.; Salah, Z.; Del Mare, S.; Galasso, M.; Gaudio, E.;Nuovo, G. J.; Lovat, F.; LeBlanc, K.; Palatini, J.; Randall, R. L.; Volinia,S.; Stein, G. S.; Croce, C. M.; Lian, J. B.; Aqeilan, R. I. MirnaSignatures Associate with Pathogenesis and Progression of Osteo-sarcoma. Cancer Res. 2012, 72, 1865−1877.(16) Almog, N.; Ma, L.; Schwager, C.; Brinkmann, B. G.; Beheshti,A.; Vajkoczy, P.; Folkman, J.; Hlatky, L.; Abdollahi, A. ConsensusMicro Rnas Governing the Switch of Dormant Tumors to the Fast-Growing Angiogenic Phenotype. PLoS One 2012, 7, e44001.(17) Almog, N.; Briggs, C.; Beheshti, A.; Ma, L.; Wilkie, K. P.;Rietman, E.; Hlatky, L. Transcriptional Changes Induced by theTumor Dormancy-Associated Microrna-190. Transcription 2013, 4,177.(18) Wan, K.; Ebert, B.; Voigt, J.; Wang, Q.; Dai, Y.; Haag, R.;Kemmner, W. In Vivo Tumor Imaging Using a Novel Rnai-BasedDetection Mechanism. Nanomedicine 2012, 8, 393−398.(19) Fischer, W.; Calderon, M.; Schulz, A.; Andreou, I.; Weber, M.;Haag, R. Dendritic Polyglycerols with Oligoamine Shells Show LowToxicity and High Sirna Transfection Efficiency in Vitro. BioconjugateChem. 2010, 21, 1744−1752.(20) Ofek, P.; Fischer, W.; Calderon, M.; Haag, R.; Satchi-Fainaro, R.In Vivo Delivery of Small Interfering Rna to Tumors and TheirVasculature by Novel Dendritic Nanocarriers. FASEB J. 2010, 24,3122−3134.(21) Naumov, G. N.; MacDonald, I. C.; Chambers, A. F.; Groom, A.C. Solitary Cancer Cells as a Possible Source of Tumour Dormancy?Semin. Cancer Biol. 2001, 11, 271−276.(22) Ferber, S.; Tiram, G.; Satchi-Fainaro, R. MonitoringFunctionality and Morphology of Vasculature Recruited by FactorsSecreted by Fast-Growing Tumor-Generating Cells. J. Visualized Exp.2014, e51525.(23) Fidler, I. J. Orthotopic Implantation of Human ColonCarcinomas into Nude Mice Provides a Valuable Model for theBiology and Therapy of Metastasis. Cancer Metastasis Rev. 1991, 10,229−243.(24) Fukumura, D.; Yuan, F.; Monsky, W. L.; Chen, Y.; Jain, R. K.Effect of Host Microenvironment on the Microcirculation of HumanColon Adenocarcinoma. Am. J. Pathol. 1997, 151, 679−688.(25) Tortora, G. J.; Derrickson, B. Principles of Anatomy andPhysiology, 13th ed.; Wiley: Hoboken, NJ, 2012.(26) Bartkova, J.; Rezaei, N.; Liontos, M.; Karakaidos, P.; Kletsas, D.;Issaeva, N.; Vassiliou, L. V.; Kolettas, E.; Niforou, K.; Zoumpourlis, V.C.; Takaoka, M.; Nakagawa, H.; Tort, F.; Fugger, K.; Johansson, F.;Sehested, M.; Andersen, C. L.; Dyrskjot, L.; Orntoft, T.; Lukas, J.; et al.Oncogene-Induced Senescence Is Part of the Tumorigenesis BarrierImposed by DNA Damage Checkpoints. Nature 2006, 444, 633−637.(27) Bester, A. C.; Roniger, M.; Oren, Y. S.; Im, M. M.; Sarni, D.;Chaoat, M.; Bensimon, A.; Zamir, G.; Shewach, D. S.; Kerem, B.

ACS Nano Article

DOI: 10.1021/acsnano.5b06189ACS Nano 2016, 10, 2028−2045

2043

Page 17: Identification of Dormancy-Associated Article MicroRNAs for ... · characterized pairs of dormant and fast-growing human osteosarcoma models. Using these pairs of mouse tumor models,

Nucleotide Deficiency Promotes Genomic Instability in Early Stages ofCancer Development. Cell 2011, 145, 435−446.(28) Herrick, J.; Bensimon, A. Single Molecule Analysis of DNAReplication. Biochimie 1999, 81, 859−871.(29) Rubin, B. P.; Antonescu, C. R.; Gannon, F. H.; Hunt, J. L.;Inwards, C. Y.; Klein, M. J.; Kneisl, J. S.; Montag, A. G.; Peabody, T.D.; Reith, J. D.; Rosenberg, A. E.; Krausz, T.; Members of the CancerCommittee. Protocol for the Examination of Specimens from Patientswith Tumors of Bone. Arch. Pathol. Lab. Med. 2010, 134, e1−7.(30) Lyden, D.; Hattori, K.; Dias, S.; Costa, C.; Blaikie, P.; Butros, L.;Chadburn, A.; Heissig, B.; Marks, W.; Witte, L.; Wu, Y.; Hicklin, D.;Zhu, Z.; Hackett, N. R.; Crystal, R. G.; Moore, M. A.; Hajjar, K. A.;Manova, K.; Benezra, R.; Rafii, S. Impaired Recruitment of Bone-Marrow-Derived Endothelial and Hematopoietic Precursor CellsBlocks Tumor Angiogenesis and Growth. Nat. Med. 2001, 7, 1194−1201.(31) Shaked, Y.; Ciarrocchi, A.; Franco, M.; Lee, C. R.; Man, S.;Cheung, A. M.; Hicklin, D. J.; Chaplin, D.; Foster, F. S.; Benezra, R.;Kerbel, R. S. Therapy-Induced Acute Recruitment of CirculatingEndothelial Progenitor Cells to Tumors. Science 2006, 313, 1785−1787.(32) Ciarrocchi, A.; Jankovic, V.; Shaked, Y.; Nolan, D. J.; Mittal, V.;Kerbel, R. S.; Nimer, S. D.; Benezra, R. Id1 Restrains P21 Expressionto Control Endothelial Progenitor Cell Formation. PLoS One 2007, 2,e1338.(33) Miquerol, L.; Langille, B. L.; Nagy, A. Embryonic DevelopmentIs Disrupted by Modest Increases in Vascular Endothelial GrowthFactor Gene Expression. Development 2000, 127, 3941−3946.(34) Cervi, D.; Shaked, Y.; Haeri, M.; Usenko, T.; Lee, C. R.; Haigh,J. J.; Nagy, A.; Kerbel, R. S.; Yefenof, E.; Ben-David, Y. EnhancedNatural-Killer Cell and Erythropoietic Activities in Vegf-a-Over-expressing Mice Delay F-Mulv-Induced Erythroleukemia. Blood2007, 109, 2139−2146.(35) Carmeliet, P.; Dor, Y.; Herbert, J. M.; Fukumura, D.;Brusselmans, K.; Dewerchin, M.; Neeman, M.; Bono, F.;Abramovitch, R.; Maxwell, P.; Koch, C. J.; Ratcliffe, P.; Moons, L.;Jain, R. K.; Collen, D.; Keshert, E. Role of Hif-1alpha in Hypoxia-Mediated Apoptosis, Cell Proliferation and Tumour Angiogenesis.Nature 1998, 394, 485−490.(36) You, W. K.; McDonald, D. M. The Hepatocyte Growth Factor/C-Met Signaling Pathway as a Therapeutic Target to InhibitAngiogenesis. BMB Rep 2008, 41, 833−839.(37) Li, X.; Roslan, S.; Johnstone, C. N.; Wright, J. A.; Bracken, C. P.;Anderson, M.; Bert, A. G.; Selth, L. A.; Anderson, R. L.; Goodall, G. J.;Gregory, P. A.; Khew-Goodall, Y. Mir-200 Can Repress Breast CancerMetastasis through Zeb1-Independent but Moesin-Dependent Path-ways. Oncogene 2014, 33, 4077.(38) Scomparin, A.; Polyak, D.; Krivitsky, A.; Satchi-Fainaro, R.Achieving Successful Delivery of Oligonucleotides - from Physico-Chemical Characterization to in Vivo Evaluation. Biotechnol. Adv. 2015,33, 1294.(39) Merdan, T.; Kunath, K.; Petersen, H.; Bakowsky, U.; Voigt, K.H.; Kopecek, J.; Kissel, T. Pegylation of Poly(Ethylene Imine) AffectsStability of Complexes with Plasmid DNA under in Vivo Conditions ina Dose-Dependent Manner after Intravenous Injection into Mice.Bioconjugate Chem. 2005, 16, 785−792.(40) Nishiamia, N.; Kataoka, K. Pic Micelles for Gene Delivery. InPolymer Therapeutics Ii: Polymers as Drugs, Conjugates and GeneDelivery Systems; Satchi-Fainaro, R.; Duncan, R., Eds. Springer-Verlag:Heidelberg, Germany, 2006; Vol. 193, pp 67−101.(41) Zuckerman, J. E.; Gritli, I.; Tolcher, A.; Heidel, J. D.; Lim, D.;Morgan, R.; Chmielowski, B.; Ribas, A.; Davis, M. E.; Yen, Y.Correlating Animal and Human Phase Ia/Ib Clinical Data with Calaa-01, a Targeted, Polymer-Based Nanoparticle Containing Sirna. Proc.Natl. Acad. Sci. U. S. A. 2014, 111, 11449−11454.(42) Hwang, H. S.; Kang, H. C.; Bae, Y. H. Bioreducible Polymers asa Determining Factor for Polyplex Decomplexation Rate andTransfection. Biomacromolecules 2013, 14, 548−556.

(43) Vaidyanathan, S.; Anderson, K. B.; Merzel, R. L.; Jacobovitz, B.;Kaushik, M. P.; Kelly, C. N.; van Dongen, M. A.; Dougherty, C. A.;Orr, B. G.; Banaszak Holl, M. M. Quantitative Measurement ofCationic Polymer Vector and Polymer-Pdna Polyplex Intercalationinto the Cell Plasma Membrane. ACS Nano 2015, 9, 6097−6109.(44) Cohen, Z. R.; Ramishetti, S.; Peshes-Yaloz, N.; Goldsmith, M.;Wohl, A.; Zibly, Z.; Peer, D. Localized Rnai Therapeutics ofChemoresistant Grade Iv Glioma Using Hyaluronan-Grafted Lipid-Based Nanoparticles. ACS Nano 2015, 9, 1581−1591.(45) Evans, B. C.; Hocking, K. M.; Kilchrist, K. V.; Wise, E. S.;Brophy, C. M.; Duvall, C. L. Endosomolytic Nano-Polyplex PlatformTechnology for Cytosolic Peptide Delivery to Inhibit PathologicalVasoconstriction. ACS Nano 2015, 9, 5893−5907.(46) Sun, Z.; Song, X.; Li, X.; Su, T.; Qi, S.; Qiao, R.; Wang, F.;Huan, Y.; Yang, W.; Wang, J.; Nie, Y.; Wu, K.; Gao, M.; Cao, F. In VivoMultimodality Imaging of Mirna-16 Iron Nanoparticle Reversing DrugResistance to Chemotherapy in a Mouse Gastric Cancer Model.Nanoscale 2014, 6, 14343−14353.(47) Shu, D.; Li, H.; Shu, Y.; Xiong, G.; Carson, W. E., 3rd; Haque,F.; Xu, R.; Guo, P. Systemic Delivery of Anti-Mirna for Suppression ofTriple Negative Breast Cancer Utilizing Rna Nanotechnology. ACSNano 2015, 9, 9731−9740.(48) Babae, N.; Bourajjaj, M.; Liu, Y.; Van Beijnum, J. R.; Cerisoli, F.;Scaria, P. V.; Verheul, M.; Van Berkel, M. P.; Pieters, E. H.; VanHaastert, R. J.; Yousefi, A.; Mastrobattista, E.; Storm, G.; Berezikov, E.;Cuppen, E.; Woodle, M.; Schaapveld, R. Q.; Prevost, G. P.; Griffioen,A. W.; Van Noort, P. I.; et al. Systemic Mirna-7 Delivery InhibitsTumor Angiogenesis and Growth in Murine Xenograft Glioblastoma.Oncotarget 2014, 5, 6687−6700.(49) Wang, S.; Cao, M.; Deng, X.; Xiao, X.; Yin, Z.; Hu, Q.; Zhou, Z.;Zhang, F.; Zhang, R.; Wu, Y.; Sheng, W.; Zeng, Y. DegradableHyaluronic Acid/Protamine Sulfate Interpolyelectrolyte Complexes asMirna-Delivery Nanocapsules for Triple-Negative Breast CancerTherapy. Adv. Healthcare Mater. 2015, 4, 281−290.(50) Bernstein-Molho, R.; Kollender, Y.; Issakov, J.; Bickels, J.; Dadia,S.; Flusser, G.; Meller, I.; Sagi-Eisenberg, R.; Merimsky, O. ClinicalActivity of Mtor Inhibition in Combination with Cyclophosphamide inthe Treatment of Recurrent Unresectable Chondrosarcomas. CancerChemother. Pharmacol. 2012, 70, 855−860.(51) Van Winkle, P.; Angiolillo, A.; Krailo, M.; Cheung, Y. K.;Anderson, B.; Davenport, V.; Reaman, G.; Cairo, M. S. Ifosfamide,Carboplatin, and Etoposide (Ice) Reinduction Chemotherapy in aLarge Cohort of Children and Adolescents with Recurrent/RefractorySarcoma: The Children’s Cancer Group (Ccg) Experience. Pediatr.Blood Cancer 2005, 44, 338−347.(52) Stacchiotti, S.; Longhi, A.; Ferraresi, V.; Grignani, G.;Comandone, A.; Stupp, R.; Bertuzzi, A.; Tamborini, E.; Pilotti, S.;Messina, A.; Spreafico, C.; Gronchi, A.; Amore, P.; Vinaccia, V.; Casali,P. G. Phase Ii Study of Imatinib in Advanced Chordoma. J. Clin. Oncol.2012, 30, 914−920.(53) George, S.; Merriam, P.; Maki, R. G.; Van den Abbeele, A. D.;Yap, J. T.; Akhurst, T.; Harmon, D. C.; Bhuchar, G.; O’Mara, M. M.;D’Adamo, D. R.; Morgan, J.; Schwartz, G. K.; Wagner, A. J.; Butrynski,J. E.; Demetri, G. D.; Keohan, M. L. Multicenter Phase Ii Trial ofSunitinib in the Treatment of Nongastrointestinal Stromal TumorSarcomas. J. Clin. Oncol. 2009, 27, 3154−3160.(54) Almog, N.; Ma, L.; Raychowdhury, R.; Schwager, C.; Erber, R.;Short, S.; Hlatky, L.; Vajkoczy, P.; Huber, P. E.; Folkman, J.; Abdollahi,A. Transcriptional Switch of Dormant Tumors to Fast-GrowingAngiogenic Phenotype. Cancer Res. 2009, 69, 836−844.(55) Reya, T.; Morrison, S. J.; Clarke, M. F.; Weissman, I. L. StemCells, Cancer, and Cancer Stem Cells. Nature 2001, 414, 105−111.(56) Misso, G.; Di Martino, M. T.; De Rosa, G.; Farooqi, A. A.;Lombardi, A.; Campani, V.; Zarone, M. R.; Gulla, A.; Tagliaferri, P.;Tassone, P.; Caraglia, M. Mir-34: A New Weapon against Cancer?Mol. Ther.–Nucleic Acids 2014, 3, e194.(57) Feng, X.; Wang, Z.; Fillmore, R.; Xi, Y. Mir-200, a New StarMirna in Human Cancer. Cancer Lett. 2014, 344, 166−173.

ACS Nano Article

DOI: 10.1021/acsnano.5b06189ACS Nano 2016, 10, 2028−2045

2044

Page 18: Identification of Dormancy-Associated Article MicroRNAs for ... · characterized pairs of dormant and fast-growing human osteosarcoma models. Using these pairs of mouse tumor models,

(58) Montanini, L.; Lasagna, L.; Barili, V.; Jonstrup, S. P.; Murgia, A.;Pazzaglia, L.; Conti, A.; Novello, C.; Kjems, J.; Perris, R.; Benassi, M. S.Microrna Cloning and Sequencing in Osteosarcoma Cell Lines:Differential Role of Mir-93. Cell. Oncol. 2012, 35, 29−41.(59) Fang, L.; Deng, Z.; Shatseva, T.; Yang, J.; Peng, C.; Du, W. W.;Yee, A. J.; Ang, L. C.; He, C.; Shan, S. W.; Yang, B. B. Microrna Mir-93Promotes Tumor Growth and Angiogenesis by Targeting Integrin-Beta8. Oncogene 2011, 30, 806−821.(60) He, C.; Xiong, J.; Xu, X.; Lu, W.; Liu, L.; Xiao, D.; Wang, D.Functional Elucidation of Mir-34 in Osteosarcoma Cells and PrimaryTumor Samples. Biochem. Biophys. Res. Commun. 2009, 388, 35−40.(61) Ouyang, L.; Liu, P.; Yang, S.; Ye, S.; Xu, W.; Liu, X. A Three-Plasma Mirna Signature Serves as Novel Biomarkers for Osteosarco-ma. Med. Oncol. 2013, 30, 340.(62) Dani, N.; Olivero, M.; Mareschi, K.; van Duist, M. M.; Miretti,S.; Cuvertino, S.; Patane, S.; Calogero, R.; Ferracini, R.; Scotlandi, K.;Fagioli, F.; Di Renzo, M. F. The Met Oncogene Transforms HumanPrimary Bone-Derived Cells into Osteosarcomas by TargetingCommitted Osteo-Progenitors. J. Bone Miner. Res. 2012, 27, 1322−1334.(63) El Naggar, A.; Clarkson, P.; Zhang, F.; Mathers, J.; Tognon, C.;Sorensen, P. H. Expression and Stability of Hypoxia Inducible Factor1alpha in Osteosarcoma. Pediatr Blood Cancer 2012, 59, 1215−1222.(64) Gregory, P. A.; Bert, A. G.; Paterson, E. L.; Barry, S. C.; Tsykin,A.; Farshid, G.; Vadas, M. A.; Khew-Goodall, Y.; Goodall, G. J. TheMir-200 Family and Mir-205 Regulate Epithelial to MesenchymalTransition by Targeting Zeb1 and Sip1. Nat. Cell Biol. 2008, 10, 593−601.(65) Li, Y.; Zhang, J.; Zhang, L.; Si, M.; Yin, H.; Li, J. DiallylTrisulfide Inhibits Proliferation, Invasion and Angiogenesis ofOsteosarcoma Cells by Switching on Suppressor Micrornas andInactivating of Notch-1 Signaling. Carcinogenesis 2013, 34, 1601−1610.(66) Korpal, M.; Ell, B. J.; Buffa, F. M.; Ibrahim, T.; Blanco, M. A.;Celia-Terrassa, T.; Mercatali, L.; Khan, Z.; Goodarzi, H.; Hua, Y.; Wei,Y.; Hu, G.; Garcia, B. A.; Ragoussis, J.; Amadori, D.; Harris, A. L.;Kang, Y. Direct Targeting of Sec23a by Mir-200s Influences CancerCell Secretome and Promotes Metastatic Colonization. Nat. Med.2011, 17, 1101−1108.(67) He, M.; Cheng, Y.; Li, W.; Liu, Q.; Liu, J.; Huang, J.; Fu, X.Vascular Endothelial Growth Factor C Promotes Cervical CancerMetastasis Via up-Regulation and Activation of Rhoa/Rock-2/MoesinCascade. BMC Cancer 2010, 10, 170.(68) Ling, H.; Fabbri, M.; Calin, G. A. Micrornas and Other Non-Coding Rnas as Targets for Anticancer Drug Development. Nat. Rev.Drug Discovery 2013, 12, 847−865.(69) Jaffe, E. A.; Nachman, R. L.; Becker, C. G.; Minick, C. R.Culture of Human Endothelial Cells Derived from Umbilical Veins.Identification by Morphologic and Immunologic Criteria. J. Clin. Invest.1973, 52, 2745−2756.(70) Ferber, S.; Tiram, G.; Satchi-Fainaro, R. MonitoringFunctionality and Morphology of Vasculature Recruited by FactorsSecreted by Fast-Growing Tumor-Generating Cells. J. Visualized Exp.2014, e51525.(71) Weidner, N.; Semple, J. P.; Welch, W. R.; Folkman, J. TumorAngiogenesis and Metastasis–Correlation in Invasive Breast Carcino-ma. N. Engl. J. Med. 1991, 324, 1−8.(72) Mor, E.; Cabilly, Y.; Goldshmit, Y.; Zalts, H.; Modai, S.; Edry,L.; Elroy-Stein, O.; Shomron, N. Species-Specific Microrna RolesElucidated Following Astrocyte Activation. Nucleic Acids Res. 2011, 39,3710−3723.(73) Shaked, Y.; Bertolini, F.; Man, S.; Rogers, M. S.; Cervi, D.;Foutz, T.; Rawn, K.; Voskas, D.; Dumont, D. J.; Ben-David, Y.; Lawler,J.; Henkin, J.; Huber, J.; Hicklin, D. J.; D’Amato, R. J.; Kerbel, R. S.Genetic Heterogeneity of the Vasculogenic Phenotype ParallelsAngiogenesis; Implications for Cellular Surrogate Marker Analysis ofAntiangiogenesis. Cancer Cell 2005, 7, 101−111.(74) Sunder, A.; Kramer, M.; Hanselmann, R.; Mulhaupt, R.; Frey, H.Molecular Nanocapsules Based on Amphiphilic HyperbranchedPolyglycerols. Angew. Chem., Int. Ed. 1999, 38, 3552−3555.

(75) Sunder, A.; Heinemann, J.; Frey, H. Controlling the Growth ofPolymer Trees: Concepts and Perspectives for HyperbranchedPolymers. Chem. - Eur. J. 2000, 6, 2499−2506.(76) Kramer, M.; Stumbe, J. F.; Grimm, G.; Kaufmann, B.; Kruger,U.; Weber, M.; Haag, R. Dendritic Polyamines: Simple Access to NewMaterials with Defined Treelike Structures for Application in NonviralGene Delivery. ChemBioChem 2004, 5, 1081−1087.(77) Cain, B. F.; Baguley, B. C.; Denny, W. A. Potenial AntitumorAgents. 28. Deoxyribonucleic Acid Polyintercalating Agents. J. Med.Chem. 1978, 21, 658−668.(78) Guillot-Nieckowski, M.; Joester, D.; Stohr, M.; Losson, M.;Adrian, M.; Wagner, B.; Kansy, M.; Heinzelmann, H.; Pugin, R.;Diederich, F.; Gallani, J. L. Self-Assembly, DNA Complexation, and PhResponse of Amphiphilic Dendrimers for Gene Transfection.Langmuir 2007, 23, 737−746.(79) Hoerter, J. A.; Walter, N. G. Chemical Modification Resolvesthe Asymmetry of Sirna Strand Degradation in Human Blood Serum.RNA 2007, 13, 1887−1893.

ACS Nano Article

DOI: 10.1021/acsnano.5b06189ACS Nano 2016, 10, 2028−2045

2045