Nanomedicine in osteosarcoma therapy: Micelleplexes for delivery of nucleic acids and drugs toward osteosarcoma-targeted therapies
Journal Pre-proofs Review article Nanomedicine in osteosarcoma therapy: micelleplexes for delivery of nucleic acids and drugs for osteosarcoma-targete...
Journal Pre-proofs Review article Nanomedicine in osteosarcoma therapy: micelleplexes for delivery of nucleic acids and drugs for osteosarcoma-targeted therapy Miguel Pereira-Silva, Carmen Alvarez-Lorenzo, Angel Concheiro, Ana Cláudia Santos, Francisco Veiga, Ana Figueiras PII: DOI: Reference:
European Journal of Pharmaceutics and Biopharmaceutics
Received Date: Revised Date: Accepted Date:
16 July 2019 9 September 2019 31 October 2019
Please cite this article as: M. Pereira-Silva, C. Alvarez-Lorenzo, A. Concheiro, A. Cláudia Santos, F. Veiga, A. Figueiras, Nanomedicine in osteosarcoma therapy: micelleplexes for delivery of nucleic acids and drugs for osteosarcoma-targeted therapy, European Journal of Pharmaceutics and Biopharmaceutics (2020), doi: https:// doi.org/10.1016/j.ejpb.2019.10.013
This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Nanomedicine in osteosarcoma therapy: micelleplexes for delivery of nucleic acids and drugs for osteosarcoma-targeted therapy
Miguel Pereira-Silva1, Carmen Alvarez-Lorenzo3, Angel Concheiro3, Ana Cláudia Santos1,2 Francisco Veiga1,2, Ana Figueiras1,2*
1Department
of Pharmaceutical Technology, Faculty of Pharmacy, University of
Coimbra, Portugal;
2REQUIMTE/LAQV,
Group of Pharmaceutical Technology, Faculty of Pharmacy,
University of Coimbra, Coimbra, Portugal;
3Departamento
de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma
(GI-1645), Facultad de Farmacia and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Spain.
*Corresponding author: Dr. Ana Figueiras Phone: (+351) 239 488 431 Fax. (+351) 239 488 503 E-mail address: [email protected] Postal address: Faculty of Pharmacy, University of Coimbra, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal
Abstract Osteosarcoma(OS) represents the main cancer affecting bone tissue, and one of the most frequent in children. In this review we discuss the major pathological hallmarks of this pathology, its current therapeutics, new active biomolecules, as well as the nanotechnology outbreak applied to the development of innovative strategies for selective OS targeting. Small RNA molecules play a role as key-regulator molecules capable of orchestrate different responses in what concern cancer proliferation, migration and invasiveness. Frequently associated with lung metastasis, new strategies are urgent to upgrade the therapeutic outcomes and the life-expectancy prospects. Hence, the prominent rise of micelleplexes as multifaceted and efficient structures for small nucleic acid delivery and selective drug targeting is revisited here with special emphasis in ligandmediated active targeting. Future landmarks towards the development of novel nanostrategies for both OS diagnosis and OS therapy improvements are also discussed. Keywords Cancer targeting; osteosarcoma; doxorubicin; micelleplexes; gene delivery; active targeting Funding This work was supported by the grant FCT PTDC/CTM-BIO/1518/2014 from the Portuguese Foundation for Science and Technology (FCT) and the European Community Fund (FEDER) through the COMPETE2020 program.This work received financial support from National Funds (FCT/MEC, Fundação para a Ciência e Tecnologia/Ministério da Educação e Ciência) through project UID/QUI/50006/2013, co-financed by European Union (FEDER under the Partnership Agreement PT2020). It was also supported by the grant FCT PTDC/BTM-MAT/30255/2017 (POCI-010145-FEDER-030255) from the Portuguese Foundation for Science and Technology (FCT) and the European Community Fund (FEDER) through the COMPETE2020 program. This research was also funded by MINECO (SAF2017-83118-R), Agencia Estatal de Investigación (AEI) Spain, Xunta de Galicia (Grupo de Referencia Competitiva ED431C 2016/008), and FEDER (Spain).
1. Introduction Cancer comprises one of the biggest challenges in nowadays’ Medicine, with an expected increase in new cases in the near future and strong impact at all levels of socio-sanitary care[1, 2]. Cancer burden in 2018 was estimated in its incidence and mortality, corresponding to 18.1 million new cancer cases and 9.6 million deaths related to this disease[2]. Osteosarcoma(OS) represents the commonest primary malignance of the bone tissue, affecting children and adolescents at much higher rate than other segments of population. This malignant bone tumor is characterized by the production of immature osteoid extracellular matrix and has a mesenchymal origin[3]. Whilst continuous efforts regarding therapies to improve and ameliorate clinical outcomes, including chemotherapy and limb salvage surgery have leveraged the 5-year survival rate up to >65% in localized resectable tumors, in the case of advanced and metastasized osteosarcoma the rate markedly decreases to ca. 20%[4]. Moreover, akin to other types of cancer, there is a remarked tendency for invasion and early metastasis, primarily to the lungs, worsening the therapeutic outcomes and life expectancy among patients diagnosed with this rare cancer. Several pathways and molecules of different origins may contribute to the instalment and progression of this disease. Bearing in mind that the medical therapy for this disease has met little changes throughout the years since the 1970’s, new therapies are urgently needed. Innovative therapies able to improve OS therapy efficiency and capable of lower the toxicity and adverse effects related to the conventional chemotherapy bestow categorical relevance, as potential advances are under development regarding cancer therapy in OS[5]. Novel drug delivery systems have met intensive developments, and nanotechnology applied to biomedicine has emerged as a multi-advantageous approach, since it can provide remarkable improvements in drug bioavailability
(enhanced solubility and stability), biodistribution and controlled release by not only prolonging drug systemic circulation, but also enabling the site specific delivery of the drug and its accumulation in tumor sites, thus achieving a selective targeting profile [6]. Polymer-based nanosystems are interesting platforms in different targeted therapies and the combination of both therapeutic and diagnostic molecules is being received with special enthusiasm as synergistic strategies for tackling tumor cells. RNA interference (RNAi) technology has provided interesting results in using small interfering molecules playing roles in cancer regulation processes. Henceforth, different nanocarriers have been explored as anti-cancer active platforms for OS eradication, and within the panorama of polymeric nanoparticles, micelleplexes arrived as interesting nanocarriers for gene delivery. Micelleplexes result from the interaction of genetic material with amphiphilic copolymers bearing one or more cationic blocks. The hydrophobic block confers an inner hydrophobic core ideal for hydrophobic drugs encapsulation, while the corona-forming hydrophilic block is usually made of polycations including polyethyleneimine (PEI), poly(2dimethylaminoethyl methacrylate) PDMAEMA and polyaminoacids, such as poly-Llysine (PLL) [7]. The high density of positive charges on the cationic block allows for efficient nucleic acid condensation. Charge neutralization strongly contributes to the selfassembly of the block copolymer leading to micelle-like nucleic acid-polymer nanocomplexes. The cationic properties ensure not only the establishment of electrostatic interactions with the negatively-charged phosphate groups of nucleic acids, hence mediating efficient nucleic acid complexation, but also interactions with the negativelycharged membranes, thus contributing to cellular uptake and endosomal escape. The buffer capacities of polycations allows for endosomal disruption and further cytosolic release of the nucleic acid payload[8, 9]. Besides, as multifunctional tunable
nanosystems, micelleplexes may be endowed with stimuli-responsive properties for nucleic acid release [10] and can be surface decorated with ligands for active targeting strategies and/or cell penetrating peptides for increased cell membrane penetration, maximizing nucleic acid delivery into cells[9]. 2. Osteosarcoma’s pathology and therapeutic regimen 2.1. Osteosarcoma’s pathological hallmarks The complexity of the pathological pathways associated to the initiation, proliferation and invasion of OS are undoubtedly still a challenge as for many other cancers. The interconnection among different underplayers and the wide number and scope of molecules, pathways and systems involved have been described to a certain extent in diverse literature. Wnt pathway is perhaps one of the most extensively studied in this specific type of cancer and, despite the need for further investigations, it appears to play an important role in what cancer cells growth is concerned. The canonical Wnt signalling is the most broadly studied and through its activation via a Wnt ligand, a signal cascade is triggered resulting in activation of specific mediators, followed by the dissociation of a proteic complex, thus resulting on the intracellular accumulation of the protein BCatenin, that further translocates to the nucleus stimulating the transcription of several genes, such as the c-myc and cyclin D1, enhancing cellular responses and potentially contributing to cell growth and proliferation[11]. It should be noted that cyclins are true key-players in the cell growth process and cell cycle regulation[12]. As a group of signal transduction pathways, there are other signalling mechanisms, also called the noncanonical Wnt signalling pathways, without B-catenin involvement, also important for several cellular mechanisms such as cytoskeleton and calcium levels regulation, but less studied. Notch signalling has also revealed an important pathway in which there is an
overexpression of Notch genes and ligands in OS cell lines with greater metastization capabilities, thus suggesting an important association between the tumour ability to metastasize and the important regulatory function of the notch pathway signalling [13]. Other significant pathways well-described in literature are the Ezrin pathway[14, 15] and the Ephrin-receptors mediated molecular signalling pathway[16, 17]. In fact, the probable role of ezrin, a member of the ERM (ezrin/radixin/moesin) group of proteins functioning as a membrane-cytoskeleton linker[18], in metastasis formation and cancer progression has been studied. Ezrin can activate NF-kappaB pathway leading to a EGF-induced epithelial-mesenchymal transition (EMT), instigating the tumor progression through a NF-kappaB-ezrin route[19]. Aberrant signalling is found in OS and receptor tyrosine kinases (RTKs) overexpression and implication in tumor progression is well documented, such as the Axl, EphB2 (ephrin B2 receptor), FGFR2 (fibroblast growth factor receptor 2), and Ret [20]. A growing body of evidence suggests that the extracellular matrix metalloproteinases (MMP), including MMP-1, MMP-2 and MMP-9, play a role in proliferation, invasion and metastasis of OS cells[21-23]. Mutations in the retinoblastoma (Rb) gene Rb-1 and p53 gene have long been identified as genetic alterations associated with the development of OS[24]. Interactions between the OS cells and the lung are of utmost importance for lung colonization and the establishment of metastasis, reinforcing the importance of the lung metastatic niche[5]. Despite the need for further investigations, some mediators have been identified, for instance IL-6 and CXCL8, the two possibly implied in tumor tropism[25]. An illustration of the principal pathways occurring in OS can be found in Figure 1. Besides, several potential prognosis biomarkers have been studied regarding their implications in the development of OS, metastasis formation and predictive of poor survival chances, such as apoptosis antigen 1 (Fas)-negative OS cells, which can evade normal elimination mechanisms and potentiate metastization [26, 27]
and nucleic acids, such as microRNA 183 downregulation implied in cancer migration and invasiveness, via Ezrin pathway[28]. Besides, vascular endothelial growth factor (VEGF) is a widely-touted prognostic marker, a key regulator of angiogenesis with significant clinical importance regarding OS, its progression and prognosis. Indeed, its overexpression in osteosarcoma patients appears to be an indicative of poor prognosis[29]. Therefore, a connection between the potential biomarker’s expression and either the increasing histopathological marks (increase of tumor size and/or metastatic processes) and clinical marks (advanced tumor grading and clinical staging and decreased overall survival) are not only relevant aspects regarding OS prognosis and evolution context, but also to the development of new targeted therapies based on such dysregulated pathological mechanisms, paving the way to new avenues for OS treatment, as discussed later in this paper.
< Figure 1 > Figure 1. Osteosarcoma view through cell proliferation, invasion and metastasis mechanisms including lung colonization and specific interaction with the tumoral niche.
2.2. Osteosarcoma therapy: from conventional to innovative therapies The entailed pathological features of OS appearance, progression and tissue-invasion abilities have undoubtedly painted OS as a highly malignant, prone-to metastasis, rapidlyspreading cancer, also related to relatively poor prognosis and clinical outcome. The overall exposure cycle from which the patient is affected by the disease, including risk factors, diagnosis, staging and preconized treatments have been widely outlined (Figure 2).
< Figure 2> Figure 2: Osteosarcoma: key facts and general perspective.
Despite the therapeutic arsenal, there is a significant percentage of unsuccessful cases, thus calling for new and more efficient strategies. The search for new molecular targets for therapeutic purposes has met significant disappointment and remains a significant challenge[30]. According to the American Cancer Society, the three main treatments applicable are surgery, chemotherapy and radiotherapy. Albeit surgical resection and combinational chemotherapy are incorporated in standard-care therapies, radiotherapy can be prescribed especially in the case of non-resectable tumors, in which tumor removal surgery cannot be done effectively after chemotherapy was given. Radiotherapy helps not only by contributing to the control of the expansion of cancerous cells, thus adding to the therapeutic success, but also to ameliorate and relieve the well-known symptoms associated with the cancer therapy itself[31]. Adjuvant chemotherapy has shown to improve the prognosis of OS patients and is usually administered via drug combos, such as dual administration of doxorubicin and cisplatin[4]. Significant progresses have been documented recently, regarding adjuvant chemotherapy in OS therapy, as other drug combinations have shown therapeutic advantageousness[32]. Whilst the general protocol of therapeutics in OS differ upon the type and evolution of the disease, the sequence of treatment is usually initiated by a period of before-surgery chemotherapy, which can be helpful in the case of non-resectable tumors, followed by the tumor-removal surgery and an additional adjuvant chemotherapy period. Nevertheless, the outcome for patients with advanced stages OS remains dismal, hence other strategies have been developed for paving the way to new treatment avenues[33]. Efforts to carry out new and innovative drug development strategies that leverage new research, and clinical trials have been
undertaken. Hence, different approaches with potential clinical relevance regarding OS therapy are under research focus. Immunotherapy has received considerable attention as an increasingly attractive strategy for OS treatment[5]. Nevertheless, clinical benefit has not yet shown to be sufficient, as therapeutic success is hampered by the rarity of this cancer, its heterogeneity and still insufficient knowledge regarding tumor specific-antigens as new targets[34]. Besides, the complexity of the immune system and immune-cancer cell biointeractions and resistance mechanisms developed by cancer cells have remained a challenge to overall improvements in immunotherapy response[35]. Special emphasis has been given to checkpoint inhibitors-based therapeutic approaches as well as to combined therapies to tackle the immunosuppressive microenvironment of OS[35, 36]. Cellular immunotherapy recurring to gene-engineered tumor-specific T-cells, such as T cell receptor (TCR)engineered T cells and chimeric antigen receptor T (CAR-T) cells [37] can upgrade functionality and precision of T cells[38] and, together with dendritic cell (DC) therapies [39], have shown increasing interest among the current innovative therapies under development. Another strategies including antibody-based ones such as monoclonal antibodies targeting insulin-like growth factor-1 receptor (IGF-1R)[40], insulin-like growth factor-2 receptor (IGF-2R)[41], anti-ganglioside GD2 antibody[42]
and
bispecific antibodies targeting IGF-1R and EGFR[43]have also been explored. Additionally, nucleic acids including ribonucleic acid interference (RNAi) molecules, such as micro RNAs (miRNAs) and small interfering RNAs (siRNAs) have been put into the bargain, as different studies have shown their potential roles mediating OS evolution[44]. However, their potential therapeutic action regarding degradation of messenger RNA (mRNA) through RNAi mechanism with the intent to regulate certain cellular pathways is hampered due to their physicochemical characteristics, such as large
molecular weight, poor lipophilicity, anionic properties due to negatively-charged phosphate groups, which put together along serum instability and increased susceptibility toward enzymatic degradation limits their bioavailability, cellular uptake, endosomal escape and the ultimate transfection efficiency. Therefore, advanced strategies for loading, transportation and delivery of nucleic acid-based therapies to cancer cells are demanded[45]. In this context, several strategies including modifications in nucleic acid chemistry (e.g. for stability enhancement)[46] and optimization of viral[47] and non-viral vectors[48] have been explored. Non-viral vectors draw several advantages such as decreased potential to trigger immune responses, easier manipulation and tunable properties, as opposed to viral ones[49]. Regarding OS targeting, several non-viral strategies are discussed below, with special emphasis on micelleplexes as suitable nanocarriers for nucleic acid delivery to cancer cells.
3. A big leap to nanomedicine: osteosarcoma-targeted therapies Nanomedicine has emerged as one of the standouts of biomedical research technology favouring diagnosis, therapy and management of several diseases. Drugs and other molecules can be successfully loaded into nanocarriers for targeted therapy[50]. For this purpose, several nanosystems – either viral and non-viral - have been developed in order to tackle cancer diseases and to improve the therapeutic outcomes. Nanotechnologybased cancer therapy strategies assume more and more a remarkable relevance in today’s health researches, much abiding to the necessity to enhance targeting efficiency, reduce both systemic adverse effects and drug dosage, among others. Henceforth, different nonviral strategies have been investigated in the context of cancer therapy, integrating different pharmacological and mechanistic approaches, and OS is not an exception. Both passive - taking advantage of enhanced permeability and retention (EPR) effect - and
active targeting strategies - by surface functionalization with targeting ligands and/or payload release via stimuli-responsiveness properties - for OS have been broadly developed, as depicted in Table 1 and 2, respectively.
< Table 1 >
< Table 2 >
3.1. Osteosarcoma-targeted therapies by means of passive targeting Regarding passive targeting, lipid-based nanoparticles, such as liposomes, have been broadly developed to delivery drugs to OS cells. Particularly, doxorubicin was encapsulated in liposomes with natural phospholipids composition, via pH gradient method attaining a final encapsulation efficiency of ca. 84 %[71]. This liposomal doxorubicin (L-Doxorubicin) consists in a spherical vesicle with a hydrophilic core in which doxorubicin was loaded and a lipidic hydrophobic shell, coated with polyethylene glycol (PEG), which improved the stability, increased circulation half-time, diminished uptake by immune cells thus reducing systemic phagocytosis and, therefore, prolonging systemic circulation. Overall, the final liposomes showed an enhanced internalisation of doxorubicin by the OS cell lines and an increase in the cancer cells death, successfully increasing the therapeutic efficacy. PEGylation also facilitates conjugation with other molecules and provides solubility-improving capabilities[6]. Other polymers, such as
bioavailability and consequently the accessibility and accumulation of the nanoparticles to the cancer cells[55]. Several studies can be found in literature reporting the application of nanocarriers to the delivery of different compounds to OS cells, including not only passive but also active targeting purposes[76, 77]. Particularly, a RGD-decorated nanosystem was developed for selective targeting of doxorubicin to the OS cells, showing an improved cellular uptake when compared with the non-targeted nanoparticle. RGD is a
peptide
composed
by
the
amino
acid
sequence
Arg-Gly-Asp
(RGD),
arginylglycylaspartic acid capable of interacting with abundantly expressed surface integrins[78]. Thus, this peptidic ligand was used to specifically target the cancer cells, displaying good features concerning its biocompatibility, safety and targeting abilities. Hence, the nanosystem composed by a biodegradable polymeric micelle loaded with doxorubicin, further decorated with ligands exhibiting high affinity for surface integrins of the cancerous cells (RGD-DOX-PM) showed selective targeting therapy of OS cells, typifying a micellar targeted delivery system with reported selective killing of the malignant cells[63]. Stimuli-responsive conjugated micelles loaded with doxorubicin were designed with acid-sensibility properties due to an acid cleavable hydrazone bond[60], which enabled the selective release of the drug under the well-known tumor microenvironment acidic conditions widely referred in literature[79, 80]. Several physiopathological hallmarks, such as hypoxia, angiogenesis, and a shift from the oxygen-dependent energy production pathways lead to the production of acidic intermediaries, therefore playing a key role in the generation of an acidic extracellular milieu (decrease in pH) not only around the cancer cells but also in the vicinities of noncancerous cells[81]. D-aspartic acid octapeptide has a dual function as micellar component; it serves both as the hydrophilic moiety of the micellar structure and as a ligand for the ligand-directed targeted nanodrug delivery system. pH-sensitive hydrazone
bonds have been broadly used as a strategy in pH-responsive drug delivery cancer nanomedicines[82]. These pH-triggered drug release properties have been extensively applied to a broad set of nanosystems for cancer therapy, including cancer immunotherapy polymeric nanoparticles[83], PEGylated liposomes for chemotherapeutic drug delivery[84], and nanoparticles featuring acid-cleavable maleic acid amides, known as TACMAA[85]. Photodynamic therapy using nanoparticles has been also applied to cancer therapy. There are reports on upconverting nanoparticles containing a photosensitiser agent for breast cancer therapy[86], hollow gold-mitoxantrone PEGylated nanoparticles with the dual function of chemotherapy agent delivery and photodynamic therapy[87], or Fe3O4 and gold-based nanoparticles for theranostics, combining bioimaging and photodynamic therapy[88]. A recent study showed the application of photodynamic therapy as a strategy for OS therapy, using zinc phthalocyanine as the photosensitizer molecule with both photo-physical and photo-chemical attributes. The need for an efficient nanosystem for the loading of the photosensitiser molecule was underlined, owing to the low solubility and crystallization tendencies of the molecule, which could pose a threat to its functional activity[74]. With this purpose in mind, the molecule was incorporated in a PEG-PMAN (amphiphilic block copolymer) polymeric nanoparticle named PEG-PMAN/ZnPc (PPZ), improving the generation of ROS in OS cell lines, as well as cell cycle arrest followed by apoptosis, once light was irradiated and the structure hence activated. This dramatically enhanced the cytotoxicity properties of the nanosystem both in cell cultures and in-vivo experiments[56]. Hyaluronic acid coated cationic liposomes were developed for specific CD-44-targeting, delivering doxorubicin into the OS cells using an interesting intracellular redox-sensible glutathione-triggered drug delivery system via bio-reducible disulfide linker, connecting cholesterol with detachable polyethyleneglycol units
(PEG2000). Nanomedicines featuring redox-sensitive responsive linkers are well documented in literature, such as the case of breast cancer[89] and lung cancer[90, 91]. Viral vectors, such as adenovirus, have been used for OS targeting. This time, a siRNA targeting ezrin was carried in a adenovirus, transported to the human OS MG-63 cells, with interesting results: certain gene levels were augmented, namely Bax, p21, p53, Cyclin D1,Caspase-3, CDK4, whereas others - Bcl-2, metalloproteinases 2 (MMP-2) and 9 (MMP-9) had their expression levels reduced[92]. Cell viability, growth, invasion and migration processes all suffered inhibition after this adenovirus-siRNA ezrin-targeted delivered the inhibitory ribonucleotides, attesting the role of viral vectors for siRNA therapeutic strategies. 3.2. Active targeting to Osteosarcoma cells: specific ligands The development of successful therapeutic strategies for cancer has been notably improved with nanocarriers decorated with surface-bonded molecules of different nature, such as ligands, antibodies and aptamers, for directly guiding to the specific target-cells [93-95]. Most nanocarriers rely solely on the EPR (enhanced permeability and retention) effect, a widely-known passive targeting way in which the specific tumoral milieu can favour the accumulation of nanodrug delivery systems, through increased angiogenesis, diminished lymphatic drainage and a leakage-favouring disorganized architecture of the tumoral tissue [96]. Nevertheless, active targeting emerges as an useful way for enhancing cellular uptake although it has its own challenges since the design of active targeted nanocarriers require sufficient knowledge on the specific tumor cells to be addressed and the nano-bio interactions that can be established with the targeted and non-targeted entities [97]. Overexpression of certain surface cell receptors on cancer cells has revealed to be a promising tool for the development of active targeted drug delivery nanosystems. Folic acid receptors and transferrin receptors are two of the most studied receptors in what
concerns active ligand-targeting nanodelivery systems as a cancer therapy strategy, targeted by the respective ligands: folic acid and transferrin, respectively[98]. Regarding OS, folate entrance into the cancerous cell has been evaluated through the two main mechanisms: folate receptor and reduced folate carrier. Only, folate receptor alpha was overexpressed in OS cancer cell lines but also it was shown that the general folate uptake pathways could play major roles on altering cancer cells’ sensitivity to chemotherapeutic drugs, the development of chemoresistance and the ultimate response to anticancer therapeutics[99-102]. Other relevant targets in OS are ephrin receptors[103], VEGF[104], uPA[105, 106] and ezrin [15, 107]. Recently, ligand-directed toxin for three types of cancer, including OS, was designed. This recombinant protein called eBAT, epidermal growth factor bispecific angiotoxin, showed active targeting towards the epidermal growth factor receptor (EGFR) and the urokinase plasminogen activator receptor (uPAR). In addition to its ability to increase the tumor binding affinity, eBAT also stimulates a cytotoxic response in EGFR and uPAR overexpressing cancer cells. Comparing to the monospecific toxins, the bispecific eBAT showed increased cytotoxicity for the same concentration values, i.e. lower IC50 [108]. An OS-targeted drug delivery system was also recently developed with ligand CD80 and VEGF antibody attached to the magnetic iron oxide nanoparticles. This way, this conjugate system would be able to, not only target OS cells widely-expressed VEGF antigen, which plays major roles in tumor angiogenesis processes, but also the surface cell receptor Cytotoxic T lymphocyte-associated antigen-4 (CTLA4) expressed also by OS cancer cells, in a synergistic attempt for potentiating OS cell death[61]. The VEGFCD80 conjugated nanosystems showed, particularly at 1 µg/mL, the greatest OS cell death efficacy and, after 72 h and three nanoparticle treatments, the CD80+VEGF conjugated nanoparticles showed lower live cell count when compared to the ones only
conjugated with VEGF or CD80. The possible role of the interaction between CD80 ligand and the CTLA-4 receptor, widely known to induce cell death and one of the major standard points in immunotherapy, was also mentioned as a contributor to the results[109]. Moreover, the substantive VEGF receptor expression on OS cells associated with an overexpression of certain genes related to its molecular pathway makes this conjugated nanosystem an interesting strategy for selective targeting and drug delivery to VEGF receptor overexpressing OS cells[110-112]. CD133 aptamers were conjugated with PEGylated poly(lactic-co-glycolic acid) nanoparticles loaded with salinomycin with 50% encapsulation efficiency, for OS cancer stem cells targeting[62]. Cluster of differentiation 133 (CD133) is regarded as a cancer stem cells marker, not only in the case of OS[113] but also in other tumors[114, 115]. The salinomycin-conjugated polymeric nanoparticles (Ap-SAL-NP) were capable of reducing in vivo both tumor volume and tumor weight, when compared to the other formulations (free salinomycin, salinomycin nanoparticles, aptamers-conjugated nanoparticles). At day 60, and comparing to the initial tumoral volume (day 0) the mice treated with free salinomicyn showed a 17.4 times higher tumoral volume, whereas the ones treated with the salinomycin nanoparticles presented 14.2 times higher tumoral volume, contrasting with the aptamer-conjugated salinomycin nanoparticles, this time displaying a 7.1 fold increase in tumoral volume. Selective killing of CD133+ cancer stem cells of OS was obtained as a result, thus showing the future potential of this nanosystem in specific targeting and in cancer therapeutics. In a subsequent study, both the cancer stem cells and the cancer cells were targeted with polymeric-lipid nanoparticles loaded with salinomycin, this time with a dual ligand strategy (EGFR aptamers and the previously used CD133 aptamers), showing interesting results once the cytotoxicity (in two different OS cell lines) of the nanoparticles containing both ligands was superior when compared
to the non-targeted (absence of ligand) or single-targeted (only one ligand) cytotoxic effect, this way representing a better and refined strategy not for only OS cells targeting but also – and synergistically – cancer stem cells targeting[66]. The αvβ3 and αvβ5 integrins have been targeted by Arg-Gly-Asp (RGD) peptide via polymeric micelles composed of amphiphilic block copolymer poly(ethylene glycol)block - poly (trimethylene carbonate) loaded with doxorubicin. RGD-DOX-PM has shown substantial potential as a OS targeting strategy, taking in mind the in vitro results of effective cancer cell killing by the integrin-mediated interactions between the peptide and the cancer cells, as previously described[63].The targeted RGD-DOX polymeric micelles showed a 5.4 fold lower IC50 when compared to the non-targeted control DOXPM in MG-63 cells; this way exhibiting a much higher antitumor activity due to active targeting. Doxorubicin-loaded liposomes were developed in a recent study, this time conjugated with an Ephrin Alpha 2 receptor - targeting peptide called YSA. [51] Ephrins are an extensive family of tyrosine kinase receptors, with functions in cancer proliferation and metastasis well documented, emerging as interesting potential targets for cancer therapy strategies[116, 117]. A couple of studies focusing on ephrin receptors targeting peptides showed the potentiality of ephrin receptors-targeted nanodrug delivery systems [118, 119]. Hence, liposomes were composed by dipalmitoylphosphatidylcholine (DPPC),
cholesterol,
and
polyethyleneglycol-conjugated
distearylphosphatidylethanolamine (DSPE-mPEG), to which the YSA-peptide was conjugated to. Results suggested an increase in specificity and toxicity for this targeted delivery system, when compared to the doxorubicin-free and non-targeted L-doxorubicin, once cytotoxicity to the SAOS cells of the YSA-L-doxorubicin nanosystem was 1.91 fold higher compared to the doxorubicin-free and 1.50 fold higher compared to the non-
targeted counterpart [51]. Nevertheless, OS therapy, as for plenty of other cancers, has major issues regarding chemoresistance [120]. Multidrug resistance (MDR) is a major threat to cancer therapy, as cancer cells become resistant to chemotherapy agents, predominantly by two mechanisms: increased drug efflux transporters on cell membranes, which decreases the intracellular concentration of the drug and by increasing anti-apoptotic signalling pathways [45, 121]. Hence, the codelivery of drugs and nucleic acids, the latter either improving intracellular concentration of chemotherapy agent or activating apoptotic pathways leading to cancer cells apoptosis, can effectively overcome resistance of cancer cells and improve therapy results. Moreover, recent studies reported drug resistance in OS treatment related to long noncoding RNAs (lncRNAs)[122], LIM kinase 1 overexpression[123], circular RNAs (cRNAs)[124], to list a few. Following the ever-increasing importance of this serious issue, a recent study was conducted for dual-targeting of OS, building on previous work, with EphA2 ligand-surface conjugated functionalized cationic liposomes, and intracellular targeting of JNK-interacting protein 1, by siRNA JIP1(Figure 3. A) [67]. Decreased gene expression of JIP1 (46%) and JIP1 mRNA (42%), the latter the target of the JIP1 siRNA, was achieved, and co-delivery of chemotherapeutic drug doxorubicin and gene-silencing JIP1 siRNA showed interesting results in OS cells killing (apoptotic effect), the greatest for the YSA-L-siRNA-DOX active targeted micelleplexes (Figure 3. B). This YSA-L-siRNA-DOX nanosystem has proved to be an interesting strategy for tackling tumor cells and for increasing chemosensitivity within the multidrug resistance (MDR) panorama.
< Figure 3> Figure 3. A) Schematic illustration of the YSA peptide-targeted liposomal DOX-siRNA nanosystem. Nanovesicles were prepared with cholesterol, DPPC, DOTAP, and DSPE–mPEG via pH gradient method.
YSA peptide was conjugated to the liposome as surface modification to target OS cells specifically, using NHS linker strategy. DOX was loaded onto the liposome, whereas siRNA was loaded on the surface of the nanovesicles. Severeal characterization tests and biological assays were carried out to assess the capabilities of the prepared codelivery system. Abbreviations: DOTAP, N-[1-(2,3-dioleoyloxy)propyl]-N,N,Ntrimethylammonium
methyl-sulfate;
DOX,
doxorubicin;
DPPC,
1,2-dipalmitoyl-sn-glycero-3-
phosphocholine; DSPE–mPEG, distearoyl-phosphatidylethanolamine–methyl-poly(ethylene glycol); OS, osteosarcoma. B) OS cell apoptosis by Annexin. Cells were treated with YSA-L, YSA-L-siRNA, DOX, YSA-L-DOX, L-siRNA-DOX, and YSA-L-siRNA-DOX for 48 h. Mean±SD are determined. The symbol * represents a statistically significant difference (P, 0.05) when compared with control. Abbreviations: DOX, doxorubicin; OS, osteosarcoma. Reproduced with permission from [135], copyright by Dovepress.
Polymer-coated folate-targeted gold nanorods (GNRs) were developed for both photothermal and chemotherapy of OS. The two different gold nanorods developed for anticancer drug nutlin-3 delivery were coated with different cytocompatible polymer derivatives: an inulin-based graft copolymer, conjugated with folic acid molecules -INULA-PEG-FA
–
and
α,β-poly(N-2-hydroxyethyl)-DL-aspartamide
(PHEA)
also
conjugated to folic acid - PHEA-EDA-FA. Hyperthermic effect was evaluated in vitro for the two nanosystems and both the nanoparticulate systems demonstrated interesting capabilities for hyperthermic therapy tumor ablation, after NIR laser exposure[64]. Selective tumor accumulation mediated by folic acid targeting was shown on folate receptors-overexpressing U2OS cancer cell lines. Testing in 3D OS models was also done both for chemotherapeutic and hyperthermic therapies, and the results showed that this two active-targeting gold nanorods are interesting strategies for, not only OS chemotherapy but also for photothermal therapy, the latter also extensively referred in literature[125, 126], combining drug molecules and thermic effects for synergistic tumor targeting and its eradication[64]. The utility of gold nanorods as mediators in
photothermic therapy has long been described; their absorptive capacities, stability, biocompatibility and facile biofunctionalization make gold nanorods to be a promising cancer-applied photothermal therapeutic nanomaterial. Other recent study regarding photothermal therapy in the context of OS was developed this time by coating the gold nanorods with polyacrylic acid (PAA), and photothermic therapy through hyperthermic effect was studied in MG63 human OS cell[57]. Apoptosismediated killing of MG63 OS cells, mainly via combination of DNA integration phenomenon and cell membrane disruption showed the notability of this nanosystem for cancer-targeting and photothermal therapy. These nanostructures can be integrated in bioscaffolds for bone repair, thus joining, both the osteogenic effect of the biomaterials and the chemo-photothermal therapeutic synergic effect of this magnetic mesoporous calcium silicate/chitosan nanoplatform, via doxorubicin anticancer effect and tumorreducing photothermic effect[127]. A different approach relies on Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) / Cas 9 genome editing technology applied to tumor-targeting strategies[128], in which a Vascular Endothelial Growth Factor A (VEGFA) guide RNA (gRNA) was developed for tackling tumor growth and angiogenesis mediated by this type of VEGF overexpressed in OS cells[65]. An aptamer specific for OS cells targeting named LC09 was developed and functionalized to a lipopolymer composed by polyethyleneimine (PEI), methoxypolyethyleneglycol (PEG) and cholesterol (CHOL). CRISPR/ CAS 9 plasmids encoding for the referred guide RNA were loaded into the lipopolymeric structure (LC09-PPC-CRISPR/Cas9 nanosystem) and specific targeting was achieved, as well as a genome editing in the portion of the genome codifying to the VEGFA, resulting not only in diminished tumor growth, but also an inhibition of lung metastasis and angiogenic processes, in orthotopic mouse models for OS and lung lesion-metastatic OS tissue[65, 129]. Other studies regarding CRISPR/Cas9-
mediated targeting of OS can be found, namely ligand 1, for programmed cell death of OS cells[130] and as a drug resistance management strategy in chemoresistant OS cells[131]. 3.3. Micelleplexes for smart delivery of nucleic acids and chemotherapy to osteosarcoma As explained above, micelleplexes refer to stable complexes of genetic material with cationic amphiphilic block copolymers that adopt a micelle-like structure[7]. Nucleic acids, such as plasmidic DNA (pDNA)[132], small interfering RNA (siRNA)[133] and micro RNA (miRNA)[134] have been successfully loaded into cancer-targeted micelleplexes, recently. Combination of nucleic acid delivery with chemotherapy agents aiming synergistic anticancer action has been also explored in a wide variety of cancers[135]. Henceforth, special attention will be given to the major RNA interference (RNAi) agents advances in osteosarcoma pathological framework, as well as the exploitation of cancer-targeted micelleplexes as suitable nanocarriers for nucleic acid delivery to the OS tumor microenvironment. 3.3.1. RNA interference agents in osteosarcoma An interesting strategy developed to a great extent in several diseases is undoubtedly the delivery of small ribonucleic acid molecules with inhibitory properties[136]. Ribonucleic acid (RNA) -based therapeutics have witnessed a huge growth and a large increase in scope in the past years, following the discovery of RNA with catalytic properties in the 80’s and the work of scientists in the late 90’s, culminating in the 2006 Nobel Prize win, for the RNA interference discovery[137]. The potential use of RNA molecules as therapeutic molecules has prompted intensive studies on different types of RNA and their different activities, attesting the versatility of the different ribonucleic acid
molecules[138]. A plethora of RNA molecules have assumed relevance in the landscape of RNA-based therapeutics, such as RNA interference agents – miRNAs, siRNAs, shRNAs-, ribozymes, aptamers, antisense oligonucleotides. Antisense oligonucleotides are single-stranded non-coding sequences of a small number of nucleotides which are specific to certain sequences of mRNA, resulting in the inhibition of the mRNA transcript after complementary binding, consequently affecting gene expression. RNA interference is a dynamic cellular process mediating silencing gene expression, most notoriously posttranscriptional gene silencing processes, in which small RNA molecules play a key role, such as endogenous microRNAs (miRNAs), functioning either as translation repressors or mRNA degradation molecules, after limited complementarity with the mRNA strand, and exogenous small interfering RNAs (siRNAs), serving as mRNA cleaving molecules after base pairing. Indeed, miRNA dysregulation has been implied in cancer progression, and the two main miRNA-based therapeutics consist in restoration of tumor suppressive miRNAs, by administration of miRNA mimics or miRNA-coding genes, or by oncogenic miRNA ablation, recurring to antisense oligonucleotides (ASO) or miRNA sponges, to name a few[139]. As for siRNA, the ability to downregulate expression of molecular targets in different signalling pathways has paved the way to siRNA-based potential therapeutics, currently under extensive investigation either alone or in combination with chemotherapy agents to maximize therapeutic outcome[140]. Despite both miRNA and siRNA-mediated gene silencing mechanisms share the same cytosolic silencing complex – RNA-induced silencing complex (RISC), further exerting their functions regarding translation repression, siRNAs have a higher complementary relationship to the target mRNA and in this way these oligonucleotides are more specific in terms of base pairing, whereas miRNAs are less specific and have a lower degree of complementarity with the targeted RNA messenger molecule[141]. RNAi therapy, in particular siRNA therapy has
been described as advantageous when compared to the traditional drug research and therapy undertones; namely, quicker research and development of specific gene inhibitors, thus less time-consuming; and both potency and selectivity are improved due to the complementarity of the target oligonucleotides to the specific mRNA strand, as opposed to conventional chemotherapy [142]. Focusing on RNAi therapeutics and the delivery of therapeutic oligonucleotides, several challenges can be easily traced, bearing in mind that the integrity of the structure is needed and the advantageous use of nanotechnology and nanocarriers as platforms for miRNA and siRNA delivery. Due to their small size and molecular weight, their renal clearance can be augmented as they are easily excreted via urine; protein adsorption and opsonisation of positive charged RNAi nanocarriers is another aspect to take in mind, resulting in the development of new polymeric coatings to overcome unspecific binding and opsonisation processes. Filtration of RNAi-containing nanoparticles by the reticuloendothelial system (RES) is important, and the physicochemical characteristics of the nanocarriers (shape, size, surface charge, hydrophilicity) are crucial for the final outcome. Large sizes, for example, would increase the opsonisation and uptake process by the RES cells; thus the importance of polymeric molecules capable of reducing or at least limiting RES uptake[143]. The nanoparticle should also reach the target tissue, therefore it must trespass tumoral extracellular matrix and this way reaching the cancerous cells. Despite the abnormal proliferation and secretion of certain cells and contents, accumulation of the nanoparticles in the tumour microenvironment is facilitated due to the vascular heterogeneity and discontinuity (EPR effect). Cell membranes need also to be transposed and for these cellular uptake process size and surface charge are pivotal in their roles, once smaller nanoparticles may be preferentially internalized via clathrin-mediated endocytosis, whereas bigger molecules are normally internalized via caveolae-mediated endocytosis (CvME), a clathrin-
independent endocytic pathway. For particles with positive surface charge, internalization is favoured[143]. The ability of loaded nanoparticles to escape the endosomes is another step to consider, for the successful release of miRNA and siRNA molecules, before degradation, via endosome membrane rupture. This endosomal scape is favored for nanocarriers coated with cationic polymers containing protonable amine groups, hence enhancing membrane disruption and facilitating the RNA molecule delivery. Off-target effects can arise, namely due to a partial complementarity of the strands to an unintended target, and are therefore non-desirable, once they can lead to inflammatory responses. These problems could be addressed through nucleobase modifications[144] and recurring to artificial nucleotide analogues with the ability to reduce off-target effects of siRNA[145]. miRNA-based therapies are, in pair with siRNA therapy, promising strategies and have experimented a notable increase namely within the cancer panorama[146]. miRNA have been extensively studied regarding both their pathological and therapeutical role, and the regulation pathways, downregulation (underexpression), upregulation (overexpression) in cancer cells, this way arising the classification of oncomiRs if the miRNA has an oncogenic effect, and tumor suppressor miRNAs if the miRNA functions as a tumor suppressor molecule[147]. These small nucleic acid molecules (17-21 nucleotides approximately) have shown to be key-players in the oncological molecular landscape and have been therefore extensively studied in their ability to supress cancer development, proliferation, metastization, opening new avenues for potential therapeutic targets in OS pathology (Table 3). < Table 3 > MiRNAs can be categorized in 5 main groups, for instance as potential prognosis biomarkers, thus important in OS diagnosis[169-172]; microRNAs functioning as oncogenic molecules, the oncomirs[165, 173-175]; tumor suppressor-microRNAs[176-
179]; microRNAs playing a role in chemoresistance promotion [180-183] , as well as active molecules in chemoresistance repression[184-187] (Figure 4). siRNA-based strategies for OS targeting include mutant p53 gene knockdown, by using a p53-specific siRNA [188] and Che-1 gene silencing, leading to decrease expression levels mutant p53 gene which further promotes apoptosis in OS cells[189]. < Figure 4> Figure 4. General view of major microRNAs as key molecules interplaying different roles in the pathological framework of osteosarcoma.
3.3.2. Micelleplexes for nucleic acid delivery into the tumour microenvironment The therapeutic potential of small RNA molecules has received increasing attention in the last decade and successful strategies for the efficient delivery of these molecules have led to the development of smart nanosystems for diagnosis or therapeutic purposes[190]. Micelleplexes have since emerged as an interesting strategy for not only nucleic acid delivery but also nucleic acid-drug combinations, as the hydrophobic core can function as a reservoir for hydrophobic drugs, such as chemotherapy agents[45]. This way, synergistic effects can be achieved through the co-delivery of chemotherapeutic agents and nucleic acids to target simultaneously different cellular mechanisms with overall benefit concerning cancer therapy, via direct killing of cancer cells and MDR circumvention. Also, surface chemistry modification allow ligand conjugation, thus displaying active targeting functionalities (Figure 5)[7, 9].