WO2011108955A1 - Multi -targeting system comprising a nanocarrier, nucleic acid(s) and non-nucleic acid based drug(s) - Google Patents

Multi -targeting system comprising a nanocarrier, nucleic acid(s) and non-nucleic acid based drug(s) Download PDF

Info

Publication number
WO2011108955A1
WO2011108955A1 PCT/PT2011/000005 PT2011000005W WO2011108955A1 WO 2011108955 A1 WO2011108955 A1 WO 2011108955A1 PT 2011000005 W PT2011000005 W PT 2011000005W WO 2011108955 A1 WO2011108955 A1 WO 2011108955A1
Authority
WO
WIPO (PCT)
Prior art keywords
peg
targeting system
liposomes
sirna
dlin
Prior art date
Application number
PCT/PT2011/000005
Other languages
French (fr)
Inventor
LILIANA SIMÕES MENDONçA
João Nuno SERENO DE ALMEIDA MOREIRA
Maria Da Conceição Pedroso De Lima
Sérgio Paulo DE MAGALHÃES SIMÕES
Original Assignee
Universidade De Coimbra
CENTRO DE NEUROCiÉNCIAS E BIOLOGIA CELULAR
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Universidade De Coimbra, CENTRO DE NEUROCiÉNCIAS E BIOLOGIA CELULAR filed Critical Universidade De Coimbra
Publication of WO2011108955A1 publication Critical patent/WO2011108955A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6905Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion
    • A61K47/6907Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion the form being a microemulsion, nanoemulsion or micelle
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6905Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion
    • A61K47/6911Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion the form being a liposome
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/88Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation using microencapsulation, e.g. using amphiphile liposome vesicle
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy

Definitions

  • the present invention relates to the fields of therapy- arid diagnosis of human cancer and other diseases, including but not limited to inflammation, neurodegenerative diseases,
  • Ribozymes DNA decoys and Aptamers .
  • DNAzymes are catalytic DNA molecules, not naturally occurring, that bind to and cleave their target mRNA in a sequence specific manner (Bhindi et al . 2007; Kalota et al . 2004) .
  • Ribozymes are naturally occurring catalytic RNA molecules, capable of sequence-specific cleavage of target mRNA, which catalytic activity is highly dependent on their structure (Bhindi et al . 2007; Rayburn and Zhang 2008) .
  • Decoys are small double stranded DNA molecules that contain binding sites for a variety of protein targets, namely to transcription factors.
  • aptamers have specific three dimensional structures that allow them to bind their target protein with high affinity and specificity, forming complexes and blocking their activity (Bhindi et al . 2007; Rayburn and Zhang 2008).
  • asODN are stretches of usually 18-25 nucleotides in length that hybridize in a sequence specific manner to the target mRNA through Watson-Crick base pairing (Bhindi et al . 2007; Galderisi et al . 1999; Stahel and Zangemeister-Wittke
  • Inhibition of gene expression by antisense ODN is likely to occur by two mechanisms i) the formation of a hybrid complex mRNA:asODN that prevents the ribosomes from reading along the mRNA message by steric hindrance and ii) nuclease degradation of the target mRNA, mediated by the
  • RNAi is a naturally occurring and evolutionarily conserved mechanism (Chiu and Rana 2003; Kumar and Clarke 2007; Shrivastava and Sriyastava 2008) employed by cells to mediate gene regulation, protecting the genome from invading elements such as virus and transposons (Dykxhoorn et al . 2006; Shrivastava and Srivastava 2008).
  • RNAi is thought to be evolved as a part of cell's innate
  • RNA molecules such as small interfering RNA (siRNA) , microRNA (miRNA) , repeated- associated siRNA (rasiRNA) , short-hairpin RNA (shRNA) , small- modulatory RNA (smRNA) , tiny non-coding RNA (tncRNA) and piwi- interacting RNA (piRNA) were .identified as effectors of RNAi mechanism (Martin and Caplen 2007; Shrivastava and Srivastava 2008) .
  • siRNA small interfering RNA
  • miRNA microRNA
  • rasiRNA repeated- associated siRNA
  • shRNA short-hairpin RNA
  • smRNA small- modulatory RNA
  • tncRNA tiny non-coding RNA
  • piRNA piwi- interacting RNA
  • SiRNA are double stranded RNA molecules with 21-23 nucleotides (nts) in length, with a phosphate group at the 5' end and a hydroxyl group on the 3 ' end of each strand and with a two-nucleotide overhang on the 3' end of both strands (Chiu and Rana 2003; Huang et al . 2008; Martin and Caplen 2007), with a preference for uridine residues in the 3 ' overhangs. It is very common to replace uridine residues for 2 ' -deoxythymidine to confer enhanced nuclease resistance (Kumar and Clarke 2007) . SiRNA molecules can be exogenously introduced into cell
  • siRNA can be endogenously produced from long dsRNA (-200 nts) which are processed by the Dicer into siRNA with 21-23 nts and 3 Overhangs (Huang et al . 2008; Martin and Caplen 2007; Rayburn and Zhang 2008) . Then, siRNA are incorporated into a multiprotein RNA induced silencing complex (RISC) that recognizes these dsRNA molecules and becomes active (Bantounas et al r 2004; Huang et al . 2008; Lee and Sinko 2006; Martin and Caplen 2007; Rayburn and Zhang 2008) .
  • RISC multiprotein RNA induced silencing complex
  • siRNA are unwinded through an ATP-dependent process by a helicase enzyme present in the RISC complex and the strand with lower thermodynamic stability at its 5' end remains in the complex and guide it to the complementary mRNA.
  • the antisense strands (guide strand) remains in the RISC complex in opposition to the sense strand (passenger strand) which is eliminated from the RISC.
  • the target mRNA is then cleaved by the nuclease Argonaute 2 protein of the RISC complex at a single site in the center of the duplex region, the phosphodiester bound of 10 nts from the 5' end of the siRNA (Bantounas et al . 2004; Dykxhoorn and Lieberman 2006; Lee and Sinko 2006) .
  • nucleic acid protection from nuclease degradation Even though a large number of clinical trials have been conducted with naked nucleic acids, it is well-known that the use of delivery agents have several advantages, such as nucleic acid protection from nuclease degradation and increased intracellular delivery.
  • Liposomes are micro or nanoparticles composed of one or more lipid bilayers, with an aqueous core (Drummond et al . 2008; Lasic 1998) . Liposomes were introduced as drug delivery vehicles in the 70s (Lasic 1998) , and their application in drug delivery depends on
  • the pharmacological profile of the drug entrapped in the liposomes is a function of the pharmacokinetic, biodistribution, and drug release characteristics of the
  • the lipid-based system must be designed considering a systemic application, thus being stable, exhibiting extended circulation life-times, and not interacting with blood components.
  • these particles must be efficiently internalized and should have the ability to destabilize cell membranes promoting intracellular delivery of the carried nucleic acids (Leonetti et al . 2001; Wheeler et al . 1999) .
  • SALP stabilized antisense lipid particles
  • SNALP stabilized nucleic acids lipid particles
  • SALP SNALP
  • SNALP SNALP
  • siRNA and asODN are nucleic acids
  • SNALP are composed of lipid bilayers containing a mixture of cationic and fusogenic lipids coated with
  • PEG-lipid polyethylene glycol-lipid
  • hydrophilic shield crucial for long circulation times in the blood stream and, that stabilizes the particles during their formation avoiding aggregation and fusion (Holland et al . 1996;
  • lipids used include cholesterol, a bilayer- forming lipid [such as 1,2- distearoyl -sn-glycero-3 - phosphatidylcholine (DSPC) ] , a protonable amino lipid [such as 1,2- d.ioleoyl-3-dimethylammonium-propane (DODAP) ] and a steric barrier lipid conjugate (PEG-lipid) .
  • DSPC 1,2- distearoyl -sn-glycero-3 - phosphatidylcholine
  • DODAP 1,2- d.ioleoyl-3-dimethylammonium-propane
  • PEG-lipid steric barrier lipid conjugate
  • the protonable lipid becomes positively charged and complexes the negatively charged nucleic acids, resulting in liposomes entrapping the nucleic acids. Then, the external pH is raised to physiological values at which the protonable lipid turns to neutral (Leonetti et al . 2001; Mui et al . 2001; Semple et al . 2001).
  • liposomes In order to reach all the requirements to cancer therapy, liposomes have to be engineered in a way to exhibit: i) prolonged circulation in the blood stream; ii) ability to specifically recognize and bind to target tissues or cells; iii) ability to provide an enhanced intracellular delivery of drugs and gene silencing tools, namely upon external or local stimulus (Torchilin 2009; Torchilin 2007) .
  • liposomes In order to increase circulation times of liposomes, the physicochemical properties of liposomes, such as charge, hydrophobicity, size, fluidity and packing of the lipid bilayer, influence their stability and biodistribution and have to be considered (Immordino et al . 2006) . Large, negative or positively charged liposomes have shorter half -life in the blood stream than small and neutral particles.
  • the liposome clearance mediated by the mononuclear phagocyte system ⁇ MPS is triggered by the binding of opsonins, which are serum proteins such as immunoglobulins, fibronectin, beta 2 -glycoprotein, C-reactive protein, beta 2 -macroglobulin and complement components (Immordino et al . 2006; Owens and Peppas 2006) .
  • opsonins are serum proteins such as immunoglobulins, fibronectin, beta 2 -glycoprotein, C-reactive protein, beta 2 -macroglobulin and complement components
  • Opsonin proteins quickly bind to conventional non- stealth nanoparticles, allowing macrophages of the MPS to recognize and remove these particles before they can reach their target organ and exert their therapeutic function.
  • the most used method to mask or camouflage the liposomes from the MPS is the adsorption or grafting of
  • biocompatible, soluble and hydrophilic polymers with a highly flexible main chain used to prepare long circulating liposomes are poly (aer 1 amide), poly (vinyl pyrrolidone) , poly (acryloyl morpholine) , poly (2 -ethyl -2 -oxazoline) , poly (2 -methyl - oxazoline), phosphatidyl polyglycerols , polyvinyl alcohols, polysaccharides, PEG and PEG- containing copolymers (as
  • EG lipososmes
  • PEG protein or cellular adsorption of any known polymer and has been FDA-approved for many injected biotech products.
  • PEG is eliminated by a combination of hepatic and renal pathways (Immordino et al . 2006; Ryan et al . 2008).
  • PEG molecular weight and structure can be freely modulated for specific purposes, and the process of lipid conjugation is easy and cheap (Immordino et al . 2006; Owens and Peppas 2006) .
  • PEG-lipid conjugates to be used as stealth coatings for liposomes are: i) PEG-phosphatidylethanolamine ; ii) PEG-ceramide; iii) PEG-diacylglycerol ; iv) PEG- dialkyloxypropylamine and v) PEG- 1 -methyl -4 - (cis-9- dioleyl) methyl -pyridinium chloride ( PEG-SAINT) .
  • PEG-SAINT PEG- 1 -methyl -4 - (cis-9- dioleyl) methyl -pyridinium chloride
  • Targeting to specific sites or cell surface markers is performed by coupling cell surface-directing ligands in the targeted therapeutics (Baker et al . 2003; Torchilin 2006).
  • the basic principle behind ligand- targeted therapeutics is that the delivery of drugs to cancer cells can be selectively enhanced by associating the drugs or the drug vehicles with molecules that specifically bind to antigens or receptors, which are either uniquely expressed or over-expressed on the target cells as compared to normal cells (Allen 2002; Baker et al . 2003; Sapra and Allen 2003) .
  • Targeting moieties may include, but are not limited, antibodies, antibody fragments, naturally occurring or synthetic ligands like peptides, carbohydrates, glycoproteins, or receptor ligands, i.e. essentially a y molecule that selectively
  • a classical target is the folate receptor, which has high affinity for the folic acid and is upregulated in many human cancers.
  • the transferrin receptor also over-expressed on the surface of many tumor cells, can be targeted with antibodies as well as transferrin (Torchilin
  • Monoclonal antibodies or antibody fragments can be selected in order to exhibit a high degree of specificity for the target tissue.
  • Some antibodies that bind to a specific surface receptor or antigen have intrinsic cytotoxicity, because they are able to interfere with cell proliferation and
  • Transferrin is a serum glycoprotein (80 KDa) responsible for iron transport (Baker et al . 2003; Li and Qian 2002) .
  • the iron-linked transferrin is designated by holo- transferrin.
  • the Trf receptor (TrfR) also designated as CD71, is a membrane glycoprotein, a homodimer composed of two
  • Trf binding site is localized on the extracellular domain of the receptor, and each receptor subunit binds one transferrin molecule (Li and Qian 2002; Ponka and Lok 1999) .
  • Trf attaches to the receptors on the cell surface, in a temperature- and energy- independent process (Ponka and Lok 1999) .
  • Holo-Trf binds to TrfR, and the resulting complex undergoes endocytosis via clathrin-coated pits, by a temperature- and energy-dependent process.
  • endosomal maturation the endosomal lumen is acidified to pH - 5.5. At this pH, the binding of iron to Trf is weakened leading to iron release from the protein.
  • TrfR may play an
  • TrfR also changes its conformation at low pH and, thus forcing the
  • D T1 divalent metal transporter
  • TrfR dissociates from TrfR due to its low affinity at pH 7.4, being released into the circulation, and reutilized (Li and Qian 2002; Ponka and Lok 1999; Qian et al . 2002).
  • TrfR is ubiquitously expressed in all nucleated cells in the body, however differs in levels of expression. It is highly expressed on rapidly dividing cells such as cells of the basal epidermis and intestinal epithelium and very low or frequently undetectable in non-proliferating cells.
  • Various studies have shown raised TrfR expression on cancer cells when compared to their normal counterparts, this being attributed to the increased need of rapidly dividing cells for iron as a cofactor of the ribonucleotide reductase enzyme involved in DNA synthesis (Daniels et al . 2006; Li and Qian 2002; Qian et al . 2002) .
  • Increased TrfR expression has been correlated with tumor grade and stage or prognosis (Daniels et al . 2006) .
  • TrfR TrfR
  • TrfR can be mediated by coupling Trf or antibodies to the delivery vector, such as liposomes, lipoplexes or viral vectors (Nobs et al . 2004; Qian et al . 2002) .
  • the coupling of ligands to the liposome surface can be achieved by covalent or non-covalent bonds.
  • Non-covalent methods have the great advantage of being easy to be carried out without the need of aggressive reagents, A simple method is to merely add the ligand to the phospholipids during the liposome
  • a way to overcome this limitation consists in coupling ligands to stealth liposomes by the "post-insertion” technique.
  • ligands are coupled to end-functionalized groups in PEG micelles and then ligand-PEG conjugates are transferred in a simple incubation step into the outer monolayer of pre -formed, drug/nucleic acid loaded
  • the multi-target therapeutic strategies comprise different modalities: i) the components act on separate targets to create a combination effect; ii) one component alters the ability of the other to reach its target and therefore the modulation of one target facilitates action at a second target, iii) the components act at the same target to create a
  • the pharmacological interactions between the drugs can be classified as synergism, antagonism and additivity.
  • a non- interactive or additive combina ion is observed when the
  • the golden goal of combinatory therapy is to obtain synergistic drug combinations in order to achieve more favorable outcomes, such as enhanced efficacy, decreased dosage at equal level of efficacy, minimal or slower development of drug resistance, reduction of adverse effects (Chou 2006; Jia et al . 2009; Merlin 1994) .
  • the dose reduction index (D I) is a measure of how many- fold the dose of a drug in a combination may be reduced to produce a given effect level compared with the dose of this drug used per se (Chou 2006) .
  • the antitumor activity of drug combinations can be significantly dependent on the molecular ratio of the combined drugs. For the same drug combination, some ratios can be
  • Liposomes can overcome the uncoordinated
  • pharmacokinetics of individual drugs utilized in the drug combinations because a single liposomal formulation can entrap the combined drugs in the desired ratio, allowing in vivo tumor cells to be exposed to the optimal drug/drug ratio. It is important to keep in mind that the pharmacokinetic behavior of the co- formulated drugs will be dictated by the pharmacokinetics of the drug carrier used, and thus the plasma elimination and tissue distribution of the combined agents can be coordinated adequately.
  • active loading drug is loaded into preformed vesicles, in response to specific transmembrane gradients, such as pH gradient and transition metal ions gradient (Li et al . 2008) .
  • the establishment of metal ions gradient across the liposomal membrane allows the efficient drug loading based on the formation of drug /metal complexes trapped inside liposomes (Abraham et al . 2004; Li et al . 2008) .
  • the pH gradient method to encapsulate drugs is based on the fact that neutral forms of weak acids and weak bases can permeate through lipid bilayer membranes at much faster rates than the charged forms.
  • the neutral form permeates the liposomal membrane and _ is subsequently protonated in the liposomal internal acidic buffer.
  • the charged (protonated) form of the drug permeates much less than the neutral form, the drug becomes trapped inside.
  • the drug can also be precipitated by anions such as sulphate and citrate, thus increasing the drug loading (Cullis et al. 1997; Ishida et al. 1999; Li et al . 2008).
  • not limiting examples of generation of a pH gradient are i) preparation of vesicles in acidic buffer and then exchange of external buffer or vice versa; ii) employment of a self-generating system such as ammonium sulphate gradient and, iii) using an ionophore (Abraham et al. 2004; Li et al . 2008; Mayer et al. 1986; Wang et al.
  • the efficiency of drug trapping and the capability of the encapsulated drug to be retained by the liposomes are dependent on a variety of factors, such as stability of the pH gradient (which is dependent on the buffering capacity within the liposome and the amount of loaded drug) , the chemical characteristics of the drug (as the potential to form insoluble salt products) , and the permeability of the liposomal membrane (which is affected by lipid composition and temperature)
  • siRNA and imatinib were quantified and the encapsulation yield of imatinib (A) and of siRNA (B) as well as the siRNA/ imatinib molar ratio (C) were assessed. No symbol p>0.05; *p ⁇ 0.05;
  • the present invention provides a single drug delivery nanocarrier allowing triple targeting to human disease and disorders.
  • a single drug delivery nanocarrier co-encapsulates two or more molecules in well established therapeutic molar ratios.
  • Long circulation times in the blood stream are conferred by hydrophilic polymers and the ability to target a specific cell population (cellular targeting) is conferred by coupling a targeting ligand to the surface of this nanocarrier.
  • Multi-molecular targeting is attained through the therapeutic agents encapsulated or
  • the nanocarrier is capable to transport the therapeutic agents to the target cells, avoiding degradation or biotransformation. Subsequently, it binds to the target cells, is internalized and delivers their content intracellulary .
  • this invention provides, the possibility to target a cell population and two or more molecular targets simultaneously using one single nanocarrier, containing a specific molar ratio of two or more therapeutic agents.
  • targeting ligands are coupled to the nanocarrier surface in order to target leukemia cells.
  • Ant i - BCR-ABL siR A and imatinib are co-encapsulated into the targeted nanocarrier, which allows the simultaneous
  • the active agent or therapeutic agent is fully encapsulated within the lipid particle such that the active agent or therapeutic agent are protected from enzymatic degradation, e.g., by a nucleases or proteases.
  • the antitumor activity of drug combinations can be significantly dependent on the molar ratio of the combined drugs.
  • some ratios can be synergistic, whereas other ratios can be additive or even antagonistic, which highlights the need to control drug ratios being exposed to tumor cells.
  • ratios of drug combinations exposed to tumor cells can be tightly controlled. This is something that upon systemic administration is extremely difficult to achieve, due to different pharmacokinetic profile of each one of the drugs entering the combination. Under the circumstances, tumor cells are therefore exposed to sub-optimal drug ratios with a concomitant loss in therapeutic activity. Such problem can be overcome upon incorporation/encapsulation of the drug combination into nanocarriers, like PEGylated
  • liposomes able to maintain the drug ratio from the site of administration until it reaches the tumor cells.
  • Targeting the therapeutic agents to a specific cell population allows improvement of the therapeutic activity, while lowering the side effects promoted by these agents.
  • targeting is possible through coupling of specific targeting ligands to the therapeutic agents or to the nanocarriers that carry and mediate the intracellular delivery of the therapeutic agents .
  • the lipid particles of the present invention provides, the possibility to target a cell population and two or more molecular targets simultaneously using one single nanocarrier, containing a specific molar ratio of two or more therapeutic agents.
  • the invention provides a single drug delivery
  • the lipid particles and compositions of the present invention may be used for a variety of purposes, including the delivery of associated or encapsulated therapeutic agents to cells, both in vitro and in vivo. Accordingly, the present invention provides methods for treating human diseases or disorders, by treating the subject or the subject cells/tissues with the lipid particles described here containing one or more therapeutic agents.
  • RNA interfering refers to single stranded RNA fe g mature micro RNA (miRNA) ) or double stranded RNA (e.g. siRMA) that is capable of reducing or inhibiting the expression of a target gene (e.g., by promoting the degradation or inhibiting the translation of the mRNA which are complementary to the siRNA/miRNA sequence) .
  • RNA interference thus refers to the single stranded RNA that is complementary to a target mRNA sequence, or to the double stranded RNA formed by two strands, with one of the strand complementary to the target mRNA.
  • RNA interference may have complete complementarity to the target gene or sequence or may comprise a region of mismatch (i e, an uncomplementary motif) .
  • RNA interference includes "small
  • interfering RNA or "siRNA, " e g , siRNA of about 15-60 (duplex) nucleotides in length, more typically about 15-30 (duplex) nucleotides in length, and is preferably about 20-27 (duplex) nucleotides in length (e.g., each complementary sequence of the double stranded siRNA is 15-60 nucleotides in length, preferably about 20-27 nucleotides in length.
  • siRNA duplexes may comprise 3' overhangs of about 1 to 4 nucleotides and 5' phosphate termini. Examples of siRNA include, but are not limited to, a double stranded polynucleotide molecule assembled from two separate stranded molecules, wherein one strand is the sense strand and the other is the complementary antisense strand.
  • SiRNA are chemically synthesized, or may be generated by
  • siRNA may be encoded by a plasmid (e.g., transcribed from the plasmid
  • nucleic acid refers to a polymer containing at least two deoxyribonucleotides or
  • ribonucleotides in either single or double stranded form it includes both DNA and RNA molecules.
  • DNA molecules are antisense molecules and plasmid DNA.
  • RNA molecules are siRNA, asymmetrical interfering RNA (aiRNA) , microRNA.
  • Nucleic acids may include nucleotide analogs or modified backbone residues or linkages, which are synthetic, naturally occurring, and non-naturally occurring. Not limiting examples of such analogs are phosphorothioates , phosphoramidates , methyl phosphonates and peptide-nucleic acids (PNAs) .
  • lipid refers to a group of organic water insoluble compounds which are the basic components of biological membranes. Lipids are a broad group of molecules which includes fats, waxes, sterols, phospholipids. Lipids may be broadly defined as hydrophobic or amphiphilic molecules; the amphiphilic nature of some lipids allows them to form structures such as vesicles, liposomes, or membranes in an aqueous environment.
  • a “lipid particle” or “liposomes” are micro or nanoparticles composed of one or more lipid bilayers, with an aqueous core, which is used herein to refer to a lipid formulation that can be used to deliver an active agent or therapeutic agent, such as a nucleic acid (e.g., a siRNA) , to a target site of interest.
  • an active agent or therapeutic agent such as a nucleic acid (e.g., a siRNA)
  • the therapeutic agents and/or diagnosis agent may be encapsulated in the liposomes, thereby protecting the agents from enzymatic degradation and improving the pharmacokinetics features.
  • SALP refers to stabilized antisense lipid particles
  • SNALP refers to a stabilized nucleic acid lipid particle.
  • SALP is the term used to refer to a nucleic acid-lipid particle encapsulating asODN within the lipid particles.
  • SNALP is the term used to refer to a nucleic acid-lipid particle
  • SALP/SNALP represent particles made from lipids (e.g., a cationic lipid, a neutral lipid, and a conjugated lipid that prevents aggregation and increases blood stream circulation times) , encapsulating nucleic acids (e.g., siRNA, aiRNA, mlRNA, ssDNA, dsDNA, ssRNA, short hairpin RNA (shRNA) , dsRNA, or a plasmid, including plasmids from which an interfering RNA is transcribed) .
  • SNALP includes SPLP (stabilized plasmid particles; whenever the lipid particles encapsulate plasmid molecules) ; SNALP and SALP .
  • SNALP can exhibit extended
  • circulation lifetimes following intravenous (I.V.) injection they can accumulate at distal sites (e.g., sites physically separated from the administration site) , they can mediate expression of the transfected gene or silencing of target gene expression at these distal sites, they can mediate the efficient delivery of the non- nucleic acid-based therapeutic agents co- encapsulated with the nucleic acids.
  • distal sites e.g., sites physically separated from the administration site
  • fusogenic refers to the ability of a liposome, or other drug delivery system to fuse with membranes of a cell .
  • cationic lipid refers to any lipid that carries a net positive charge at a selected pH.
  • cationic lipids are 1 , 2 -dioleoyl-3 -dimethylammonium- propane (DODAP) , 1 , 2 -dilinoleyloxy- 3 - (2 -N, - dimethylamino) ethoxypropane (DLin-EG-D A) , N, -dioleyl -N, - dimethylammonium chloride (DODAC) , 1 , 2 -dioleyloxy-N, N-dimethyl - 3 -aminopropane (DODMA) , 1 , 2 -distearyloxy-N, N-dimethyl -3 - aminopropane (DSDMA) , N- (1- (2 , 3 -dioleyloxy) propyl) -N, N, - trimethylammonium chloride (DODAC)
  • neutral lipid refers to any lipid that exist either in an uncharged or neutral z itterionic form at a selected pH.
  • the neutral lipid components may be cholesterol or a derivative, phospholipids, or a mixture of phospholipids and cholesterol or a derivative.
  • cholesterol derivatives include, but are not limited to, cholestanol, cholestanone , cholestenone , coprostanol, cholesteryl-2 1 -hydroxyethyl ether, cholesteryl -4 1 - hydroxybutyl ether, and mixtures thereof.
  • neutral lipids examples include but are not limited to dipalmitoylphosphatidylcholine (DPPC) ,
  • DSPC distearoylphosphatidylcholine
  • dioleoylphosphatidylethanolamine DOPE
  • palmitoyloleoyl - phosphatidylcholine POPC
  • palmitoyloleoyl - phosphatidylethanolamine POPE
  • palmitoyloleyol - phosphatidylglycerol POPG
  • dipalmitoyl- phosphatidylethanolamine DPPE
  • dimyristoyl- phosphatidylethanolamine DMPE
  • distearoyl- phosphatidylethanolamine DSPE
  • monomethyl- phosphatidylethanolamine dimethyl -phosphatidylethanolamine
  • DEPE dielaidoyl- phosphatidylethanolamine
  • SOPE stearoyloleoyl- phosphatidylethanolamine
  • EPC egg phosphatidylcholine
  • “Increased blood stream circulation,” as used herein, refers to a broad biodistribution of a therapeutic agent such as siRNA within an organism through the improvement of the time in circulation of the delivery vehicle.
  • the enhancement of the amount of the active agent available to be exposed to most parts of the body is generally achieved by decreasing degradation and/or blood clearance (such as by first pass organs (liver, spleen, lung, etc.) and nonspecific cell binding.
  • cancer is the designation adopted for a group of more than 100 human diseases that have in common the uncontrolled cell growth (division beyond the normal cell control), invasion (intrusion of adjacent tissues), and
  • Cancers are caused by abnormalities in the genetic material of the transformed cells. These abnormalities may be due to the effects of carcinogens, such as tobacco smoke, radiation, chemicals, or infectious agents. Cancer promoting genetic abnormalities may be caused by random errors in DNA replication, or are inherited.
  • the term "leukemia” refers to a broad group of
  • Leukemias can be classified in four main types, the acute and chronic leukemias, which are subdivided into lymphoid and myeloid leukemias. Chronic leukemias are distinguished from acute by their slower progression in opposition to the
  • lymphoid leukemias differ in the hematopoietic lineage affected by the malignant transformation; myeloid leukemias are
  • Chronic myeloid leukemia is a myeloproliferative disease originated from hematopoietic stem cells, and was the first human malignancy to be linked with a dominant acquired genetic mutation.
  • Chronic myeloid leukemia is caused by a translocation between chromosomes 9 and 22 which create the oncogene BCR-ABL and an abnormal 22 chromosome (Philadelphia chromosome; Ph) .
  • stressing refers to the at least partial suppression of the expression of the target gene as assessed by the reduction of the amount of the target mRNA.
  • lipid conjugate refers to a lipid conjugated to a shielding group that inhibits aggregation of nucleic acid lipid particles and increase blood stream
  • carbohydrates or polymers, as shielding groups, on the liposome surface extends the liposome half-life from a few minutes
  • Such lipid conjugates include, but are not limited to,
  • poly (acrylamide) poly (vinylpyrrolidone) ,
  • poly (aeryloylmorpholine) pol (2-ethyl-2-oxazoline) , poly (2- methyl -oxazoline) , phosphatidyl polyglycerols , polyvinyl
  • PEG is a polyether diol which provides a very attractive combination of properties, such as solubility in aqueous and organic media, high
  • Torchilin 2006 presents the lowest level of protein or cellular adsorption of any known polymer and has been FDA-approved for many injected biotech products.
  • PEG- lipid conjugates are: i) PEG- phosphatidylethanolamine ; ii) PEG-ceramide ; iii) PEG- diacylglycerol ; iv) PEG-dialkyloxypropylamine and v) PEG-1- methyl-4- (cis-9-dioleyl) methyl -pyridinium chloride (PEG-SAINT) (Romberg et al . 2008).
  • post insertion refers to a 1 igand coupling method in which 1 igands attached to a conjugated lipid are introduced into preformed liposomes.
  • micelles refers to an aggregate of surfactant molecules dispersed in a liquid. Generally, micelles in aqueous solution form an aggregate with the hydrophilic regions "head” in contact with the surrounding solvent (water) , whereas, the hydrophobic single tail regions are in the micelle cent e .
  • antisense oligonucleotides or “asODN” are single stranded molecules of RNA or DNA usually with 18-25 nucleotides in length that hybridize in a sequence specific manner to the target mRNA/DNA and down regulate the target gene, Description of the embodiments
  • the lipid particles of the present invention provides, the possibility to target a cell population and two or more molecular targets simultaneously using one single nanocarrier.
  • the drug delivery nanocarrier is targeted for specific organ, tissue or cells by coupling of targeting ligands to its surface.
  • targeting moieties include peptides, polypeptides, antibodies, polyclonal antibodies, monoclonal antibodies, antibody fragments, humanized antibodies, recombinant antibodies, recombinant human antibodies, proteins and cell surface ligands.
  • the ligands are linked to the surface of the nanocarrier in a way that it is able to interact with a specific molecule, protein, glycoprotein and/or cell surface receptor that is overexpressed or specifically expressed on the cell surface of a specific cellular target.
  • An appropriate spacer can be positioned between the nanocarrier and the ligand to avoid hindrance on the interaction between the ligand and its target.
  • the nanocarrier can allow multivalent cellular
  • the nanocarrier should have a diameter comprised between 100 and 200 nm to travel in the blood stream circulation without occluding circulation and without being rapidly removed from circulation by first passage organs; being available to specifically interact with the target cells in therapeutic dosages .
  • the drug delivery nanocarrier of the present invention provides a method for simultaneous encapsulation, adsorption or complexation of two or more gene silencing agents (nucleic acids) and two or more non-nucleic acid based active agents (therapeutic drugs) .
  • the gene silencing agents (nucleic acids) may comprise, but not limited to plasrnids, antisense
  • oligonucleotides oligonucleotides
  • siRNA mi NA, shRNA, aiRNA
  • DNAzymes DNA enzymes
  • Ribozymes DNA decoys, Aptamers and mixtures thereof.
  • the gene silencing agents may comprise modified nucleotides including, but not limited to phosphorothioate linkages, 2 ' -O-methyi (2'OMe) nucleotides, 2 ' -deoxy-2 ' - fluoro (2'F) nucleotides, 2 1 -deoxy nucleotides, 2 ' -0- (2 -methoxyethyl ) (MOB) nucleotides, locked nucleic acid (LNA) nucleotides, and mixtures thereof.
  • phosphorothioate linkages 2 ' -O-methyi (2'OMe) nucleotides
  • the gene silencing agents may comprise at least one or a cocktail (e.g., at least two, three, four, five, six, seven or more) of unmodified and/or modified gene silencing agents.
  • the cocktail of gene silencing agents may comprise sequences which are directed to the same region or domain and/or to different regions or domains of one or more target genes.
  • the gene silencing may be performed by any viral vector capable of accepting the coding sequences for the gene silencing agent, including but not limited to retrovirus, herpes virus,
  • adenovirus and adenoassociated virus.
  • Non-nucleic acid based active agents may include, but not limited to chemotherapy drugs, hormonal therapeutic agents, immunotherapeutic agents, antiviral drugs, anti - inflammatory compounds, antidepressants, stimulants, analgesics, antibiotics, antipyretics, vasodilators, an i -angiogenics, c o ascular agents, signal transduction inhibitors, anti -arrhythmic agents, hormones, vasoconstrictors, and steroids .
  • Non-limiting examples of chemotherapy drugs include platinum-based drugs (e.g. cisplatin, carboplat in, etc) ;
  • alkylating agents e.g., cyclophosphamide, chlorambucil, busulfan, melphalan, lomustine, carmustine, estramust ine , treosulfan, thiotepa, mitobronitol , etc
  • anti-metabolites e.g., 5-fluorouracil , methotrexate, capecitabine, cytarabine, fludarabine, gemcitabine, cladribine, raltitrexed,
  • plant alkaloids e.g., vincristine, vinblastine, vindesine, paclitaxel, docetaxel, etc.
  • topoisomerase inhibitors e.g., irinotecan, topotecan
  • cytotoxic antibiotics e.g., doxorubicin, adriamycin, daunorubicin, epirubicin, actinomycin, bleomycin, mitomycin, mitoxantrone , aclarubicin, idarubicin, dactinomycin, etc
  • taxanes e.g., docetaxel, paclitaxel
  • tyrosine kinase inhibitors e.g., gefitinib, sunitinib, erlotinib, lapatinib, canertinib, semaxinib, vatalanib, sorafenib, imatinib,
  • anti-inflamatory agents such as but not limited to ibuprofen, aceclofenac, acemetacin, acid acetilsalicilic, azapropazone , celecoxib, diclofenac sodium, diflunisal, cetodolac, fenbufen, fenoprofen, flubiprofen, indomethacin, acetaminocin, piroxicam, rofecoxib, sulindac, tenoxicam,- antiangiogenic agents or angiolytic agents such as but not limited to angiostatin (plasminogen fragment) , antiangiogenic antithrombin III, vasculostatin, vasostatin and mixtures thereof.
  • the nanocarrier is composed, but not limited, by liposomes and/or polymers.
  • liposomes are stabilized nucleic acid lipid particles (SNALP) .
  • SNALP liposomes comprise one or more cat ionic lipids, one or more neutral lipids and one or more conjugated lipid that inhibits aggregation of particles and provide long circulation times to the liposomes.
  • the cationic lipids may comprise from 10 to 60 mol%
  • the neutral lipid may comprise from 10 to 70 mol%
  • the conjugated lipid that inhibits aggregation and provides long circulation times may comprise from 1 to 10 mol%.
  • cationic lipids are 1,2- dioleoyl-3-dimethylammonium-propane (DODAP) , 1 , 2 -dilinoleyloxy- 3-(2-N,N- dimethylamino) ethoxypropane (DLin-EG-DMA) , N,N- dioleyl-N,N-dimethylammonium chloride (DODAC) , 1 , 2 -dioleyloxy- N, -dimethyl -3 -aminopropane (DODMA) , 1 , 2 -distearyloxy-N, N- dimethyl -3 -aminopropane (DSDMA) , N- (1 - ⁇ 2,3 -dioleyloxy) propyl) - ⁇ , ⁇ , ⁇ - trimethyl ammonium chloride (DOTMA) , N-(l-(2,3- dioleoyloxy) propyl) -N, N, N, N, N
  • dioctadecylamidoglycylspermine DOGS
  • DMOBA dioleyloxybenzylamme
  • DOcarbDAP dioleyloxybenzylamme
  • DOcarbDAP dioleyloxybenzylamme
  • 2-N 2-N, ' -dilmoleylcarbamyl-3 - dimethylaminopropane
  • DLincarbDAP dioctadecylamidoglycylspermine
  • DLin-DAC dimethylamino acetoxypropane
  • DLin-MA dimethylaminopropane
  • DLinDAP 1 , 2 -dilinoleoyl -3 - dimethylaminopropane
  • DLin-S- DMA 1-linoleoyl -2 -linoleyloxy-3- dimethylaminopropane
  • DLin-2 -DMAP 1-linoleoyl -2 -linoleyloxy-3- dimethylaminopropane
  • DLin-2 -DMAP 1-linoleoyl -2 -linoleyloxy-3- dimethylaminopropane
  • DLin-2 -DMAP 1-linoleoyl -2 -linoleyloxy-3- dimethylaminopropane
  • DLin-2 -DMAP 1-linoleoyl -2 -linoleyloxy-3- dimethylaminopropan
  • the neutral lipid components may be cholesterol or a derivative; phospholipids; or a mixture of phospholipids and cholesterol or a derivative thereof.
  • cholesterol derivatives include, but are not limited to, cholestanol, cholestanone, cholestenone , coprostanol, cholesteryl -2 ' - hydroxyethyl ether, cholesteryl-4 1 - hydroxybutyl ether, and mixtures thereof.
  • neutral lipids include but are not limited to dipalmitoylphosphatidylchol ine (DPPC) ,
  • DSPC distearoylphosphatidylcholine
  • dioleoylphosphatidylethanolamine DOPE
  • palmitoyloleoyl - phosphatidylcholine POPC
  • palmitoyloleoyl - phosphatidylethanolamine POPE
  • palmitoyloleyol- phosphatidylglycerol POPG
  • dipalmitoyl- phosphatidylethanolamine DPPE
  • dimyristoyl- phosphatidylethanolamine DMPE
  • distearoyl- phosphat idylethanolamine DSPE
  • monomethyl- phosphatidylethanolamine dimethyl -phosphatidylethanolamine
  • dielaidoyl- phosphatidylethanolamine DEPE
  • stearoyloleoyl - phosphatidylethanolamine SOPE
  • egg phosphatidylcholine EPC
  • biocompatible, soluble and hydrophilic polymers with a highly flexible main chain used to prepare long circulating liposomes and avoid particles
  • poiy acryl amide
  • poly vinyl pyrrol idone
  • poly acryloyl morpholine
  • poly (2 -ethyl -2 - oxazoline)
  • poly (2 -methyl -oxazoline) phosphatidyl
  • PEG-lipid particles include but are not limited to PEG-diacylglycerol (DAG) , PEG
  • PEG-SAINT dialkyloxypropyl
  • PEG-ceramide Cer
  • PEG-phosphatidylethanolami e PEG- 1 -methyl -4 - ( cis - 9 -dioleyl ) methyl - pyridinium chloride (PEG-SAINT) or mixtures thereof.
  • PEG-Cer conjugate include but are not limited to PEG- dilauryloxypropyl (C 12), a PEG- dimyristyloxypropyl (C 14), a PEG-dipalmityloxypropyl (C 16) , a PEG-distearyloxypropyl (C 18) , or mixtures thereof.
  • the PEG moiety of the PEG-lipid conjugates described herein may compose an average molecular weight ranging from about 550 da1tons to about 10 000 daltons.
  • Trf or BSA protein was modified with the addition of thiol groups through reaction with 2 -iminothiolane hydrochloride (2-IT) .
  • Trf or BSA and 2-IT freshly dissolved in HEPES buffer (20 mM HEPES, 145 mM NaCl , pH 8) were mixed in a
  • a lipid film of DSPE-PEG-MAL was prepared by solvent evaporation under a mild stream of N 2 and further dried under vacuum for 2 h. This dried lipid film was then hydrated with MES buffer (20 mM HEPES, 20 mM MES, pH 6.5), at a concentration above 2.3 ⁇ , the critical micellar concentration of the lipid (Ishida et al . 1999) .
  • Micelles were formed by strong vortex followed by 15 s heating in a water bath at 38 °C, followed by a second vortex shaking. Then, the freshly thiolated protein was coupled to the freshly prepared DSPE-PEG-MAL micelles by a thioesther linkage (protein to micelles molar ratio of 1:1) . The coupling reaction was performed overnight, ih the dark at room temperature with gentle stirring. The remaining free MAL groups in the micelles were quenched by the addition of ⁇
  • liposomes preparation Upon their formation the liposomes were extruded, 21 times, in a LipoFast mini extruder (Lipofast,
  • imatinib was encapsulated into the liposomes, by addition of imatinib to the siRNA- containing liposomes at different imatinib/total lipid molar ratios (1/3, 1/8, 1/16, 1/32, 1/42; initial imatinib/total lipid) and incubation for 1 h, at 60 °C, in a water bath.
  • the liposomes were then allowed to reach the room temperature and 4 mol % of Trf -micelles was added and incubated for 17 h, at 38 -C, in a water bath under dark.
  • Trf -liposomes were purified by size exclusion
  • SiRNA quantification The amount of siRNA entrapped inside liposomes was assessed by the Quant -iT RiboGreen RNA Assay (Molecular Probes, Invitrogen, Düsseldorf, Germany) against a siRNA standard curve. Liposomes were dissolved upon addition of 0.6 m of octaethylene glycol monododecyl ether (C 12 E 8 ) and the RiboGreen fluorescence (Xex 485 nm, Xem 530 nm, cut off 515 nm) was measured using a Spectra Max Gemini EM plate reader fluorimeter (Molecular Devices, Sunnyvale, CA) .
  • C 12 E 8 octaethylene glycol monododecyl ether
  • RiboGreen fluorescence Xex 485 nm, Xem 530 nm, cut off 515 nm
  • Imatinib quantification The method for imatinib quantification was developed by adaptation of the Dharmacon RNA Technologies (Lafayette, CO, USA) protocol for siRNA
  • microfuge tubes 0.1 ⁇ of total lipid was added to the precipitation reagent (400 ⁇ destilled water, 40 ⁇ of 10 M ammonium acetate, pH 7 and 1.5 ml absolute ethanol) up to 800 ⁇ , samples were then submitted to 30 s of strong vortex and transferred to -80 °C/2 h or -20 °C/overnight .
  • the precipitation reagent 400 ⁇ destilled water, 40 ⁇ of 10 M ammonium acetate, pH 7 and 1.5 ml absolute ethanol
  • Imatinib concentration was determined in the supernatant by measuring the absorbance at 259 nm against a standard curve of imat inib. This quantification method was optimized to eliminate any interference by the other components of the formulation.
  • Example 2- Cell viability of targeted nanocarriers co- encapsulating siRNA and imatinib Cell lines Two human chronic myeloid leukemia cell lines in blast crisis, positive for BCR-ABL oncogene, with the b3a2 translocation (K562 and LAMA- 84 cells) purchased from DSMZ (Braunschweig, Germany) were maintained in culture at 37 °C, 5% C0 2 under humidified atmosphere in RPMI-1640 ⁇ supplemented with 10% (v/v) heat -inactivated fetal bovine serum (FBS) (Gibco, invitrogeil, California, USA) , penicillin (100 U/ml) and
  • FBS heat -inactivated fetal bovine serum
  • streptomycin 100 ⁇ g/ml (Cambrex, NJ, USA) (Cambrex, NJ, USA) .
  • imatinib-resistant cell line K562 cells maintained in culture as previously described, were incubated with increasing concentrations of imatinib, starting at 0,05 ⁇ and with 0.05 ⁇ increments every 4 days of culture, until cells acquired the ability to grow at 1 ⁇ . At this time point, drug resistance was assessed and cells were designated as IRK562. The new cell line was maintained continuously in culture in the presence of 1 ⁇ of imatinib and was washed with drug- free medium before all experimental procedures.
  • Cell transfection K562 and LAMA- 8 cells (20 000 cells/well) and IRK562 cells (25 000 cells/well) in RPMI-1640 culture medium supplemented with 10% FBS and antibiotics were seeded in 96-round well plates.
  • Cells were transfected with Trf- associated liposomes co-encapsulating siRNA and imatinib at different molar ratios at 37 °C for 4 h. After incubation with liposomes, the medium was replaced with fresh medium and cells further incubated for 44 h.
  • Cell viability Cell viability was evaluated by the resazurin reduction assay (O'Brien et al . 2000). The assay measures the chemical reduction of the resazurin dye resulting from cellular metabolic activity, and allows the determination of viability over the culture period without harming the cells. Briefly, the culture medium was replaced with 10% (v/v)
  • IC 50 and DRI The required drug concentration to promote reduction of 50% in cell viability (IC 50 ) and dose reduction index (DRI), which is the magnitude of dose reduction allowed for a drug when given in a drug
  • TrfR TrfR-liposomes
  • Trf-PEG-DSPE conjugates were then inserted onto the pre-formed liposomes by the post - insertion method.
  • the siRNA encapsulation yield is also significantly affected by the presence of imatinib, namely by the imatinib : total lipid ratio used in the co-encapsulation process.
  • the formulations prepared with higher amounts of imatinib resulted in lower siRNA encapsulation yields, as compared to the formulations prepared with lower amounts of imatinib (lower imatinib : total lipid ratios, e.g. 1/42 ⁇ .
  • siRNA is encapsulated alone, under the same
  • the lowest imatinib/lipid ratio tested (1/42) resulted in the highest siRNA/imatinib ratio (0.63). At this ratio it is therefore possible to obtain therapeutic concentrations of both imatinib and siRNA inside the same liposome. In contrast, the 1/3 ratio at which the imatinib loading is too high as compared to that of siRNA, did not allow to reach therapeutic
  • Trf-liposomes co-encapsulating imatinib and siRNA molecules prepared from imatinib total lipid ratios of 1/16, 1/32 and 1/42 (1/16; 1/32 and 1/42 formulations) , resulted in 0.15, 0.35 and 0.63 siRNA/imatinib molar ratio, respectively (formulations are codenamed by the resulting siRNA/imatinib molar ratios) .
  • a remarkable decrease in the siRNA yield of encapsulation was observed for the highest ratios of
  • siRNA enhances the imatinib
  • results also revealed that the imatinib-resistant cell line IRK562 required higher siRNA/imatinib ratios, as compared to non-resistant cell line LAMA- 8 .
  • a correlation between the cellular response and the expression of Trf receptor and BCR-ABL mRNA levels could be established.
  • the cell line with higher Trf receptor expression and lower BCR-ABL mRNA levels demonstrated higher response to the tested
  • siRNA/ imatinib ratio is 0.63, a higher DRI (3.43) was obtained.
  • lower siRNA/ imatinib ratios can be employed to induce a certain degree of cytotoxicity as compared to the co- treatment of the cells with encapsulated siRNA and free imatinib.
  • Both free imatinib and imatinib encapsulated in Trf- liposomes promoted similar cytotoxicity, suggesting that these Trf -coupled liposomes loading imatinib allow efficient
  • Cy3 - siRNA is dependent on the lipid concentration incubated with the cells up to 0.34 mM of total lipid which is correspondent to 1.0 ⁇ siRNA, since no significant difference on the RFU of cells treated with 1.0 ⁇ or 2.0 ⁇ siRNA encapsulated in Trf- liposomes was observed.
  • imatiiiib total lipid ratio imatinib (nmol/anol TL) siRNA. (nmol/ ⁇ TU)
  • siRNA/imatinib molar ratios on imatinib IC 50 and DRI siRNA/imatinib molar ratios on imatinib IC 50 and DRI
  • Tf -lipoplexes for neuronal siRNA delivery a promising system to mediate gene
  • siRNAs as small molecule drugs.
  • RNA small interfering RNA
  • Ratiometric dosing of anticancer drug combinations controlling drug ratios after systemic administration regulates therapeutic activity in tumor-bearing mice. Mol Cancer Ther 5(7) :1854- 63.
  • Liposomal anticancer drugs as agents to be used in combination with other anticancer agents studies on a liposomal formulation with two
  • Liposomal anticancer drugs as agents to be used in combination with other anticancer agents studies on a liposomal formulation with two
  • RNA interference an emerging generation of biologicals. Biotechnol J 3(3) :339-53.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Genetics & Genomics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biomedical Technology (AREA)
  • Dispersion Chemistry (AREA)
  • Epidemiology (AREA)
  • Wood Science & Technology (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Molecular Biology (AREA)
  • Hospice & Palliative Care (AREA)
  • Plant Pathology (AREA)
  • Biochemistry (AREA)
  • Nanotechnology (AREA)
  • Cardiology (AREA)
  • Physics & Mathematics (AREA)
  • Microbiology (AREA)
  • Biophysics (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Psychiatry (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

MuIti-component therapeutic strategies are frequently indicated for the treatment of heterogeneous diseases as cancer. However, the antitumor activity of drug combinations is extremely dependent of the molecular ratio of the combined drugs. This patent describes a multi -targeting system comprising a nanocarrier that was engineered in order to exhibit small size, high stability over time, high encapsulation yields of nucleic acids and the ability to specifically bind to receptors overexpressed at the surface of cancer cells. Further developments were performed in order to simultaneously encapsulate nucleic acids (gene silencing agents) and antitumural drugs (imatinib) in molar ratios allowing both molecules to be used in therapeutic doses. Therefore, with this product it is possible to mediate specific delivery to cancer cells and to address two specific molecular targets (a transcript and a protein) This unique properties thus render the system a great potential for cancer treatment, namely when resistance to chemotherapy is associated.

Description

DESCRIPTION
"MUL I -TARGETING SYSTEM COMPRISING A NANOCARRIER, NUCLEIC
ACID(S) AND NON-NUCLEIC ACID BASED DRUG(S)"
Field of the invention The present invention relates to the fields of therapy- arid diagnosis of human cancer and other diseases, including but not limited to inflammation, neurodegenerative diseases,
infectious diseases, and cardiovascular disorders; more
specifically to methods of selectively targeting therapeutic or diagnostic molecules to specific cells, combining cellular and molecular targeting.
Background information Many cancers are associated with abnormal expression of some genes. The specific silencing of these genes constitutes a targeted therapy leading to high specificity and therefore minimal toxicity. There are several strategies to inhibit gene expression. Not limiting examples are antisense oligonucleotides (asODN) ; RNA interference (RNAi) ; DNA enzymes (DNAzymes) ,
Ribozymes, DNA decoys and Aptamers . DNAzymes are catalytic DNA molecules, not naturally occurring, that bind to and cleave their target mRNA in a sequence specific manner (Bhindi et al . 2007; Kalota et al . 2004) . Ribozymes are naturally occurring catalytic RNA molecules, capable of sequence-specific cleavage of target mRNA, which catalytic activity is highly dependent on their structure (Bhindi et al . 2007; Rayburn and Zhang 2008) . Decoys are small double stranded DNA molecules that contain binding sites for a variety of protein targets, namely to transcription factors. Therefore, decoys compete with endogenous DNA for the transcription factors, which results in the decrease of the transcription rate of genes that are dependent of those transcription factors (Bhindi et al . 2007; Brennan et al . 2008; Kalota et al . 2004) . Aptamers are small stretches of RNA or DNA to which gene complementarity is not important because their activity is determined by their tertiary and quaternary
structures. In fact, aptamers have specific three dimensional structures that allow them to bind their target protein with high affinity and specificity, forming complexes and blocking their activity (Bhindi et al . 2007; Rayburn and Zhang 2008). asODN are stretches of usually 18-25 nucleotides in length that hybridize in a sequence specific manner to the target mRNA through Watson-Crick base pairing (Bhindi et al . 2007; Galderisi et al . 1999; Stahel and Zangemeister-Wittke
2003; Tamm 2006) . Inhibition of gene expression by antisense ODN is likely to occur by two mechanisms i) the formation of a hybrid complex mRNA:asODN that prevents the ribosomes from reading along the mRNA message by steric hindrance and ii) nuclease degradation of the target mRNA, mediated by the
endogenous enzyme RNase H, triggered by the recognition and selective destruction of the mRNA of the mRNA : asODN complex (Figure 1.2) (Bhindi et al . 2007; Galderisi et al . 1999; Mahato et al. 2005; Rayburn and Zhang 2008) . Other mechanisms were observed including interference with mRNA processing and
transport and, inhibition of gene transcription by forming DNA triple helices in a sequence specific manner (Rayburn and Zhang 2008; Stahel and Zangemeister-Wittke 2003) . RNAi is a naturally occurring and evolutionarily conserved mechanism (Chiu and Rana 2003; Kumar and Clarke 2007; Shrivastava and Sriyastava 2008) employed by cells to mediate gene regulation, protecting the genome from invading elements such as virus and transposons (Dykxhoorn et al . 2006; Shrivastava and Srivastava 2008). Thus, RNAi is thought to be evolved as a part of cell's innate
immunity, and mediates its activity through small double stranded RNA molecules, which complementarity to the target mRNA triggers the gene silencing by mRNA degradation, either by preventing translation or by silencing the chromatin (Dykxhoorn and Lieberman 2006; Dykxhoorn et al . 2006; Shrivastava and
Srivastava 2008) . Various types of small RNA molecules, such as small interfering RNA (siRNA) , microRNA (miRNA) , repeated- associated siRNA (rasiRNA) , short-hairpin RNA (shRNA) , small- modulatory RNA (smRNA) , tiny non-coding RNA (tncRNA) and piwi- interacting RNA (piRNA) were .identified as effectors of RNAi mechanism (Martin and Caplen 2007; Shrivastava and Srivastava 2008) . SiRNA are double stranded RNA molecules with 21-23 nucleotides (nts) in length, with a phosphate group at the 5' end and a hydroxyl group on the 3 ' end of each strand and with a two-nucleotide overhang on the 3' end of both strands (Chiu and Rana 2003; Huang et al . 2008; Martin and Caplen 2007), with a preference for uridine residues in the 3' overhangs. It is very common to replace uridine residues for 2 ' -deoxythymidine to confer enhanced nuclease resistance (Kumar and Clarke 2007) . SiRNA molecules can be exogenously introduced into cell
cytoplasm. These synthetic siRNA are designed to mimic Dicer
(RNase I II -like enzyme) products enabling them to enter the RNAi machinery. Alternatively, siRNA can be endogenously produced from long dsRNA (-200 nts) which are processed by the Dicer into siRNA with 21-23 nts and 3 Overhangs (Huang et al . 2008; Martin and Caplen 2007; Rayburn and Zhang 2008) . Then, siRNA are incorporated into a multiprotein RNA induced silencing complex (RISC) that recognizes these dsRNA molecules and becomes active (Bantounas et al r 2004; Huang et al . 2008; Lee and Sinko 2006; Martin and Caplen 2007; Rayburn and Zhang 2008) . Therefore, siRNA are unwinded through an ATP-dependent process by a helicase enzyme present in the RISC complex and the strand with lower thermodynamic stability at its 5' end remains in the complex and guide it to the complementary mRNA. Thus, the antisense strands (guide strand) remains in the RISC complex in opposition to the sense strand (passenger strand) which is eliminated from the RISC. The target mRNA is then cleaved by the nuclease Argonaute 2 protein of the RISC complex at a single site in the center of the duplex region, the phosphodiester bound of 10 nts from the 5' end of the siRNA (Bantounas et al . 2004; Dykxhoorn and Lieberman 2006; Lee and Sinko 2006) .
Consequently, the targeted mRNA is degraded and protein
expression is abolished or reduced.
In order to enhance the stability and improve the potency of the above mentioned gene silencing tools several structural modifications can be performed.
Even though a large number of clinical trials have been conducted with naked nucleic acids, it is well-known that the use of delivery agents have several advantages, such as nucleic acid protection from nuclease degradation and increased intracellular delivery.
Lipid-based gene delivery systems can be used to efficiently deliver gene silencing agents. Liposomes are micro or nanoparticles composed of one or more lipid bilayers, with an aqueous core (Drummond et al . 2008; Lasic 1998) . Liposomes were introduced as drug delivery vehicles in the 70s (Lasic 1998) , and their application in drug delivery depends on
physicochemical characteristics, such as composition, size, net charge, loading efficiency and stability (Drummond et al . 2008; Lasic 1998) . Therefore, the pharmacological profile of the drug entrapped in the liposomes is a function of the pharmacokinetic, biodistribution, and drug release characteristics of the
carrier .
For cancer gene silencing the access to metastasis disease sites, as well as primary tumors is vital. To fulfill these biodistribution demands, the lipid-based system must be designed considering a systemic application, thus being stable, exhibiting extended circulation life-times, and not interacting with blood components. On the other hand, these particles must be efficiently internalized and should have the ability to destabilize cell membranes promoting intracellular delivery of the carried nucleic acids (Leonetti et al . 2001; Wheeler et al . 1999) . The stabilized antisense lipid particles (SALP) or stabilized nucleic acids lipid particles (SNALP) lipid-based formulations incorporate some features that make them promising for systemic delivery applications, including minimal surface charge, small size and long circulation times, nucleic acid entrapment within lipid bilayers at high efficiencies,
protection of the nucleic acids and efficient delivery into tumor cells. Therefore, these lipid-based systems have been successfully used by others to mediate nucleic acid delivery (Leonetti et al . 2001; Mui et al . 2001; Semple et al . 2001;
Takasaki et al . 2006).
The only difference between SALP and SNALP is the nature of the entrapped nucleic acid. The SALP designation is used when the liposomes entrap asODN, whereas SNALP is used for liposomes entrapping siRNA. As both siRNA and asODN are nucleic acids, we decided to use indistinctively the term SNALP for this type of liposomes. SNALP are composed of lipid bilayers containing a mixture of cationic and fusogenic lipids coated with
polyethylene glycol-lipid (PEG-lipid) , which provides a
hydrophilic shield crucial for long circulation times in the blood stream and, that stabilizes the particles during their formation avoiding aggregation and fusion (Holland et al . 1996;
Morrissey et al . 2005; Semple et al . 2001). These particles are formed through a self-assembled process after addition of an ethanolic solution of lipids to an aqueous buffered solution of nucleic acids (Mui et al . 2001; Semple et al . 2001). Not
limiting examples of lipids used include cholesterol, a bilayer- forming lipid [such as 1,2- distearoyl -sn-glycero-3 - phosphatidylcholine (DSPC) ] , a protonable amino lipid [such as 1,2- d.ioleoyl-3-dimethylammonium-propane (DODAP) ] and a steric barrier lipid conjugate (PEG-lipid) . When the lipids are
injected into the acidic aqueous buffer, the protonable lipid becomes positively charged and complexes the negatively charged nucleic acids, resulting in liposomes entrapping the nucleic acids. Then, the external pH is raised to physiological values at which the protonable lipid turns to neutral (Leonetti et al . 2001; Mui et al . 2001; Semple et al . 2001).
The interaction of negatively charged nucleic acids with positively charged liposomes promotes membrane
destabilization which is enhanced by ethanol, a component of the SNALP preparation. This destabilizing effect induces the
formation of multilamellar liposomes with concentric bilayer shells from large unilamellar liposomes, thereby trapping nucleic acids between the lamellae of these multilamellar liposomes. Thus, concerning morphology, it was observed that unilamellar liposomes co-exist with bi- and multilamellar liposomes (Maurer et al . 2001) .
In order to reach all the requirements to cancer therapy, liposomes have to be engineered in a way to exhibit: i) prolonged circulation in the blood stream; ii) ability to specifically recognize and bind to target tissues or cells; iii) ability to provide an enhanced intracellular delivery of drugs and gene silencing tools, namely upon external or local stimulus (Torchilin 2009; Torchilin 2007) .
In order to increase circulation times of liposomes, the physicochemical properties of liposomes, such as charge, hydrophobicity, size, fluidity and packing of the lipid bilayer, influence their stability and biodistribution and have to be considered (Immordino et al . 2006) . Large, negative or positively charged liposomes have shorter half -life in the blood stream than small and neutral particles.
The liposome clearance mediated by the mononuclear phagocyte system {MPS ) is triggered by the binding of opsonins, which are serum proteins such as immunoglobulins, fibronectin, beta 2 -glycoprotein, C-reactive protein, beta 2 -macroglobulin and complement components (Immordino et al . 2006; Owens and Peppas 2006) . Indeed, the MPS does not recognize the liposomes themselves but, rather, recognize opsonins bound to the
liposomes. Opsonin proteins quickly bind to conventional non- stealth nanoparticles, allowing macrophages of the MPS to recognize and remove these particles before they can reach their target organ and exert their therapeutic function. This
sequestration in the MPS organs is very rapid, typically a matter of minutes, and usually is concentrated in the liver and spleen (Immordino et al . 2006; Owens and Peppas 2006) .
The most used method to mask or camouflage the liposomes from the MPS is the adsorption or grafting of
shielding groups which can block the attractive forces between the liposomes and the opsonins (Immordino et al . 2006; Owens and Peppas 2006) . These masked liposomes are named as stealth liposomes. The incorporation of hydrophilic carbohydrates or polymers on the liposome surface extends the liposome half-life from a few minutes (classical non-stealth liposomes) to several hours (stealth liposomes) (Immordino et al . 2006; Sapra and Allen 2003) . There are several biocompatible, soluble and hydrophilic polymers with a highly flexible main chain used to prepare long circulating liposomes, not limiting examples are poly (aer 1 amide), poly (vinyl pyrrolidone) , poly (acryloyl morpholine) , poly (2 -ethyl -2 -oxazoline) , poly (2 -methyl - oxazoline), phosphatidyl polyglycerols , polyvinyl alcohols, polysaccharides, PEG and PEG- containing copolymers (as
poloxamers and poloxamines) (Owens and Peppas 2006; Torchilin 2009; Torchilin 2006). Still, by far, the most successful and used approach is the covalent coupling of PEG chains to
lipososmes (PEGylation) . EG is a polyether diol which provides a very attractive combination of properties, such as solubility in aqueous and organic media, high flexibility of its polymer chain, very low toxicity, immunogenicity , and antigenicity
(Immordino et al . 2006; Torchilin 2006) . In addition, it
presents the lowest level of protein or cellular adsorption of any known polymer and has been FDA-approved for many injected biotech products. PEG is eliminated by a combination of hepatic and renal pathways (Immordino et al . 2006; Ryan et al . 2008). PEG molecular weight and structure can be freely modulated for specific purposes, and the process of lipid conjugation is easy and cheap (Immordino et al . 2006; Owens and Peppas 2006) . Not limiting examples of PEG- lipid conjugates to be used as stealth coatings for liposomes are: i) PEG-phosphatidylethanolamine ; ii) PEG-ceramide; iii) PEG-diacylglycerol ; iv) PEG- dialkyloxypropylamine and v) PEG- 1 -methyl -4 - (cis-9- dioleyl) methyl -pyridinium chloride ( PEG-SAINT) . The choice is dependent on the compromise between prolongation of the
circulation times, sufficient interaction of the carrier with the target and efficient drug delivery (Romberg et al . 2008) .
Targeting to specific sites or cell surface markers is performed by coupling cell surface-directing ligands in the targeted therapeutics (Baker et al . 2003; Torchilin 2006). The basic principle behind ligand- targeted therapeutics is that the delivery of drugs to cancer cells can be selectively enhanced by associating the drugs or the drug vehicles with molecules that specifically bind to antigens or receptors, which are either uniquely expressed or over-expressed on the target cells as compared to normal cells (Allen 2002; Baker et al . 2003; Sapra and Allen 2003) . Targeting moieties may include, but are not limited, antibodies, antibody fragments, naturally occurring or synthetic ligands like peptides, carbohydrates, glycoproteins, or receptor ligands, i.e. essentially a y molecule that selectively
recognizes and binds to target antigens or receptors (Sapra and Allen 2003; Torc ilin 2007). A classical target is the folate receptor, which has high affinity for the folic acid and is upregulated in many human cancers. The transferrin receptor, also over-expressed on the surface of many tumor cells, can be targeted with antibodies as well as transferrin (Torchilin
2007) .
Monoclonal antibodies or antibody fragments can be selected in order to exhibit a high degree of specificity for the target tissue. Some antibodies that bind to a specific surface receptor or antigen have intrinsic cytotoxicity, because they are able to interfere with cell proliferation and
differentiation. Thus, an added advantage of using these molecules for targeting purposes is the possibility of synergy interaction between the antibodies and the drug (Allen 2002; Sapra and Allen 2003) .
Transferrin (Trf) is a serum glycoprotein (80 KDa) responsible for iron transport (Baker et al . 2003; Li and Qian 2002) . The iron-linked transferrin is designated by holo- transferrin. The Trf receptor (TrfR) , also designated as CD71, is a membrane glycoprotein, a homodimer composed of two
identical transmembrane subunits. The Trf binding site is localized on the extracellular domain of the receptor, and each receptor subunit binds one transferrin molecule (Li and Qian 2002; Ponka and Lok 1999) . Trf attaches to the receptors on the cell surface, in a temperature- and energy- independent process (Ponka and Lok 1999) . Holo-Trf binds to TrfR, and the resulting complex undergoes endocytosis via clathrin-coated pits, by a temperature- and energy-dependent process. Upon endosomal maturation, the endosomal lumen is acidified to pH - 5.5. At this pH, the binding of iron to Trf is weakened leading to iron release from the protein. Although the observed acidification is not enough for efficient iron release, TrfR may play an
important role in this process presumably because the TrfR also changes its conformation at low pH and, thus forcing the
transferrin into an open conformation, facilitating release of iron. The free Fe3+ is reduced to Fe2+ on the endosomal membrane, which is subsequently transported out of endosome to the cytosol by the divalent metal transporter (D T1) to be used as a
cofactor or stored on ferritin. The resulting apo-Trf -TrfR complex is then recruited through exocytic vesicles back to the cell surface. At extracellular physiological pH, apo-Trf
dissociates from TrfR due to its low affinity at pH 7.4, being released into the circulation, and reutilized (Li and Qian 2002; Ponka and Lok 1999; Qian et al . 2002).
The TrfR is ubiquitously expressed in all nucleated cells in the body, however differs in levels of expression. It is highly expressed on rapidly dividing cells such as cells of the basal epidermis and intestinal epithelium and very low or frequently undetectable in non-proliferating cells. Various studies have shown raised TrfR expression on cancer cells when compared to their normal counterparts, this being attributed to the increased need of rapidly dividing cells for iron as a cofactor of the ribonucleotide reductase enzyme involved in DNA synthesis (Daniels et al . 2006; Li and Qian 2002; Qian et al . 2002) . Increased TrfR expression has been correlated with tumor grade and stage or prognosis (Daniels et al . 2006) .
Thus the elevated levels of TrfR in cancer cells, the extracellular accessibility of this molecule and its
constitutive internalization render this receptor with ideal properties for targeting therapeutics to cancer cells. Targeting TrfR can be mediated by coupling Trf or antibodies to the delivery vector, such as liposomes, lipoplexes or viral vectors (Nobs et al . 2004; Qian et al . 2002) .
The coupling of ligands to the liposome surface can be achieved by covalent or non-covalent bonds. Non-covalent methods have the great advantage of being easy to be carried out without the need of aggressive reagents, A simple method is to merely add the ligand to the phospholipids during the liposome
preparation. However, the coupling efficacy is relatively low (4-40%) and aggregation of the liposomes is frequently observed. Furthermore, the amount of ligand linked to the liposome is not easily controllable, and the correct orientation of the ligands is not ensured. Finally, detachment of the ligands in in vivo might occur (Nobs et al . 2004) . Covalent reactions are an effective way to irreversibly attach ligands to the carriers, because the linkage formed is much more stable and reproducible when compared to non-covalent methods (Nobs et al . 2004) .
However, covalent reactions require the need of chemical
reagents and when the coupling reaction is performed in preformed liposomes, the risk of altering the structure of the particles and in some cases of the encapsulated compounds must be considered. Therefore, it is essential to choose non- aggressive reagents and to work under mild, controlled
conditions (Nobs et al . 2004) . A way to overcome this limitation consists in coupling ligands to stealth liposomes by the "post- insertion" technique. In the "post-insertion" method ligands are coupled to end-functionalized groups in PEG micelles and then ligand-PEG conjugates are transferred in a simple incubation step into the outer monolayer of pre -formed, drug/nucleic acid loaded
liposomes (Allen et al . 2002; Ishida et al . 1999; Sapra and Allen 2003; Us r et al . 1996). Not limiting examples of end- functional ized derivatives of PEG have been synthesized for coupling ligands to the PEG-terminus are pyridyldiotiopropionoylamino (PDP) -PEG, hydrazide (Hz) -PEG and maleimide (Mai) -PEG . The reaction between thiol groups introduced in the ligands and the maleimide groups coupled to PEG is highly efficient leading to a stable thioether bond (Sapra and Allen 2003; Uster et al . 1996) . The combination therapy with multiple drugs or
multiple modalities is now a widespread practice in the
treatment of cancer with outcomes that would be unattainable with single drug treatments. The rationale for the use of multiple agents in chemotherapy relies on the heterogeneity of tumor cells and consequent differences in tumor cell sensitivity to individual drugs (Ishida et al . 1999; Saxon et al . 1999a; Zoli et al . 2001). Multi-target therapeutics can be more
efficacious and less vulnerable to adaptative resistance, because the biological system is less able to compensate for the action of two or more drugs simultaneously. Consequently, it is possible to better control complex diseases with less
probability to drug resistance development, accomplishing this way higher cytotoxicity and durable anticancer activity
(Zimmermann et al , 2007; Zoli et al . 2001).
The multi-target therapeutic strategies comprise different modalities: i) the components act on separate targets to create a combination effect; ii) one component alters the ability of the other to reach its target and therefore the modulation of one target facilitates action at a second target, iii) the components act at the same target to create a
combination effect and increase the pharmacological action
(Cullis et al . 1997) . Generally, the cellular targets and mechanisms of action are well-known for most drugs however their interference with each other when used in combination has not been
sufficiently investigated. It is well established that different drugs can interact with each other at the pharmacological level and modify the respective effects (Merlin 1994; Zoli et al .
2001) . The pharmacological interactions between the drugs can be classified as synergism, antagonism and additivity. A non- interactive or additive combina ion is observed when the
compounds act without any interaction among them and
consequently the observed effect is not statistically different from the expected, theoretically calculated from the response of every agent administered individually (Goldoni and Johansson 2007; Merlin 1994) . Synergism occurs when the observed effect is statistically higher than the calculated effect, and antagonism is observed when the obtained effect is statistically lower than the calculated effect (Chou 2006; Goldoni and Johansson 2007; Jia et al . 2009; Merlin 1994) .
Thus, the golden goal of combinatory therapy is to obtain synergistic drug combinations in order to achieve more favorable outcomes, such as enhanced efficacy, decreased dosage at equal level of efficacy, minimal or slower development of drug resistance, reduction of adverse effects (Chou 2006; Jia et al . 2009; Merlin 1994) . The dose reduction index (D I) is a measure of how many- fold the dose of a drug in a combination may be reduced to produce a given effect level compared with the dose of this drug used per se (Chou 2006) .
The antitumor activity of drug combinations can be significantly dependent on the molecular ratio of the combined drugs. For the same drug combination, some ratios can be
synergistic, whereas other ratios can be additive or even antagonistic. This highlighted the heed to control drug ratios being exposed to tumor cells (Mayer et al . 2006; Tardi et al . 2007) . In in vivo applications, in contrast to in vitro systems, where drug concentrations exposed to tumor cells can be tightly controlled, the individual agents of an anticancer drug combination will be distributed and eliminated independently of each other disrupting the original drug ratio. Therefore, uncoordinated pharmacokinetics of individual drugs utilized in the drug cocktails results in exposure to sub-optimal drug ratios with a concomitant loss in therapeutic activity (Mayer et al . 2006; Tardi et al . 2007).
Liposomes can overcome the uncoordinated
pharmacokinetics of individual drugs utilized in the drug combinations, because a single liposomal formulation can entrap the combined drugs in the desired ratio, allowing in vivo tumor cells to be exposed to the optimal drug/drug ratio. It is important to keep in mind that the pharmacokinetic behavior of the co- formulated drugs will be dictated by the pharmacokinetics of the drug carrier used, and thus the plasma elimination and tissue distribution of the combined agents can be coordinated adequately.
There are two drug loading approaches, passive and active loading. In active loading, drug is loaded into preformed vesicles, in response to specific transmembrane gradients, such as pH gradient and transition metal ions gradient (Li et al . 2008) . The establishment of metal ions gradient across the liposomal membrane allows the efficient drug loading based on the formation of drug /metal complexes trapped inside liposomes (Abraham et al . 2004; Li et al . 2008) . On the other hand, the pH gradient method to encapsulate drugs is based on the fact that neutral forms of weak acids and weak bases can permeate through lipid bilayer membranes at much faster rates than the charged forms. Therefore, for drugs with protonable amines, as imatinib mesylate, the neutral form permeates the liposomal membrane and _ is subsequently protonated in the liposomal internal acidic buffer. As the charged (protonated) form of the drug permeates much less than the neutral form, the drug becomes trapped inside. The drug can also be precipitated by anions such as sulphate and citrate, thus increasing the drug loading (Cullis et al. 1997; Ishida et al. 1999; Li et al . 2008).
In the pH gradient method, not limiting examples of generation of a pH gradient are i) preparation of vesicles in acidic buffer and then exchange of external buffer or vice versa; ii) employment of a self-generating system such as ammonium sulphate gradient and, iii) using an ionophore (Abraham et al. 2004; Li et al . 2008; Mayer et al. 1986; Wang et al.
2005) .
The efficiency of drug trapping and the capability of the encapsulated drug to be retained by the liposomes are dependent on a variety of factors, such as stability of the pH gradient (which is dependent on the buffering capacity within the liposome and the amount of loaded drug) , the chemical characteristics of the drug (as the potential to form insoluble salt products) , and the permeability of the liposomal membrane (which is affected by lipid composition and temperature)
(Abraham et al. 2004; Cullis et al. 1997; Ishida et al. 1999; Saxon et al . 1999a) .
Brief description of the drawings
Figure 1. Encapsulation yields of imatinib and siRNA and siRNA/imatinib molar ratio in Trf-liposomes co-encapsulating both drugs. Different imatinib : total lipid molar ratios (1/3; 1/5; 1/8; 1/16; 1/32; 1/42) were incubated with SNALP liposomes loaded with an i-BC.R-.4I5I· siRNA as described in Materials and Methods. In addition, imatinib at 1/8 imatinib: total lipid ratio [referenced in the figure as 1/8 (imatj] was incubated with Trf- liposomes without siRNA. After liposome purification, the final siRNA and imatinib were quantified and the encapsulation yield of imatinib (A) and of siRNA (B) as well as the siRNA/ imatinib molar ratio (C) were assessed. No symbol p>0.05; *p<0.05;
**p<0.01 and ***p<0.001 when the comparison was established with the 1/3 formulation or between the conditions indicated by the lines (A and B) or with the 1/16 formulation (C) .
Figure 2- Extent of association of Trf-coupled
liposomes loaded with Cy3 - siRNA to LAMA- 84 cells. A) Cells were incubated at 37 bC during 4 h with different concentrations of Tr -coupled liposomes encapsulating 0.2-2.0 μΜ Gy3 - siRNA (0.2- 2.0 μΜ Trf), BSA-coupled liposomes encapsulating 0.5-2.0 μΜ Cy3 - siRNA (0.5-2.0 μΜ BSA) or non-targeted liposomes encapsulating 0.5-2.0 μΜ Cy3 - siRNA (0.5-2.0 μΜ NT) . B) Cells were incubated with different concentrations of Trf -liposomes loading Cy3 -siRNA at 37 DC in the presence (37 °C Trf RS) or absence (37 °C) of an excess of free Trf or incubated at 4 °C in the presence (4 °C Trf RS) or absence (4 °C) of free Trf. The fluorescence of the Cy3 -siRNA associated to LAMA- 84 cells was assessed by flow cytometry. A) One-way ANOVA analysis of variance combined with Tukey post test and B) regular two-way ANOVA analysis of
variance combined with Bonferroni post test for multiple
comparisons were used. ***p<0.001, **p<0.01, *p<0.05 and no symbol p>0.05 when the comparison was established between the conditions indicated by the lines (A) or when comparison Was established with cells treated at 37 °C in the absence of free Trf (B) . ###p<0.001 when the comparison was established with the corresponding concentration of the Trf-liposome formulation (A) .
Figure 3- Confocal microscopy images from studies performed in LAMA- 84 cells incubated with Trf-coupled liposomes loaded with Cy3 -siRNA. Cells were incubated for 4 h, at 37 °C with 1 μΜ of Cy3 -siRNA encapsulated in A,F) Trf -coupled
liposomes, B) BSA-coupled liposomes C) N -liposomes or D) Trf- coupled liposomes in the presence of an excess of free Trf . E) LAMA- 84 cells incubated with Trf-coupled liposomes at 4 °C.
Confocal images were acquired in a point scanning confocal microscope Zeiss LSM 510 Meta (Zeiss, Germany) , A-E) with a 4 Ox EC Plan-Neofluar . F) Differential interference contrast (DIG) images were taken with a 63x Plan-Apochromat oil immersion obj ectives .
Summary of the invention
The present invention provides a single drug delivery nanocarrier allowing triple targeting to human disease and disorders. Such a system co-encapsulates two or more molecules in well established therapeutic molar ratios. Long circulation times in the blood stream are conferred by hydrophilic polymers and the ability to target a specific cell population (cellular targeting) is conferred by coupling a targeting ligand to the surface of this nanocarrier. Multi-molecular targeting is attained through the therapeutic agents encapsulated or
incorporated into the nanocarrier. The nanocarrier is capable to transport the therapeutic agents to the target cells, avoiding degradation or biotransformation. Subsequently, it binds to the target cells, is internalized and delivers their content intracellulary .
Overall, as a major benefit, this invention provides, the possibility to target a cell population and two or more molecular targets simultaneously using one single nanocarrier, containing a specific molar ratio of two or more therapeutic agents. For example, for CML treatment, targeting ligands are coupled to the nanocarrier surface in order to target leukemia cells. Ant i - BCR-ABL siR A and imatinib are co-encapsulated into the targeted nanocarrier, which allows the simultaneous
knockdown of the BCR-ABL oncogene and the inhibition of the tyrosine kinase activity of the Bcr-Abl oncoprotein. The active agent or therapeutic agent is fully encapsulated within the lipid particle such that the active agent or therapeutic agent are protected from enzymatic degradation, e.g., by a nucleases or proteases.
Detailed description of the invention Introduction
The combination of different strategies directed to the same molecular target or to different molecular targets emerges as a promising therapeutic strategy for some diseases, allowing to reach outcomes unattainable with single therapeutic approaches. The rationale to the use of drug combinations for cancer treatment, is based on the heterogeneity of tumor cells and consequently on differences in tumor cell responses to individual drugs. One not limiting example of combined
strategies to achieve a more effective treatment for drug- sensitive or drug-resistant cancer cells is to combine imatinib mesylate with gene silencing tools, such as siRNA, targeting the oncogene BCR-ABL aiming at chronic myeloid leukemia (CML)
treatment. However, the antitumor activity of drug combinations can be significantly dependent on the molar ratio of the combined drugs. In fact, for the same drug combination some ratios can be synergistic, whereas other ratios can be additive or even antagonistic, which highlights the need to control drug ratios being exposed to tumor cells.
In in vitro cell culture systems, ratios of drug combinations exposed to tumor cells can be tightly controlled. This is something that upon systemic administration is extremely difficult to achieve, due to different pharmacokinetic profile of each one of the drugs entering the combination. Under the circumstances, tumor cells are therefore exposed to sub-optimal drug ratios with a concomitant loss in therapeutic activity. Such problem can be overcome upon incorporation/encapsulation of the drug combination into nanocarriers, like PEGylated
liposomes, able to maintain the drug ratio from the site of administration until it reaches the tumor cells.
Targeting the therapeutic agents to a specific cell population allows improvement of the therapeutic activity, while lowering the side effects promoted by these agents. Such
targeting is possible through coupling of specific targeting ligands to the therapeutic agents or to the nanocarriers that carry and mediate the intracellular delivery of the therapeutic agents .
The lipid particles of the present invention provides, the possibility to target a cell population and two or more molecular targets simultaneously using one single nanocarrier, containing a specific molar ratio of two or more therapeutic agents. The invention provides a single drug delivery
nanocarrier allowing triple targeting to human disease and disorders. The lipid particles and compositions of the present invention may be used for a variety of purposes, including the delivery of associated or encapsulated therapeutic agents to cells, both in vitro and in vivo. Accordingly, the present invention provides methods for treating human diseases or disorders, by treating the subject or the subject cells/tissues with the lipid particles described here containing one or more therapeutic agents.
Definitions
As used herein, the following terms have the meanings attributed to them unless specified otherwise.
The term "RNA interfering" or "RNAi" refers to single stranded RNA fe g mature micro RNA (miRNA) ) or double stranded RNA (e.g. siRMA) that is capable of reducing or inhibiting the expression of a target gene (e.g., by promoting the degradation or inhibiting the translation of the mRNA which are complementary to the siRNA/miRNA sequence) . RNA interference, thus refers to the single stranded RNA that is complementary to a target mRNA sequence, or to the double stranded RNA formed by two strands, with one of the strand complementary to the target mRNA. RNA interference may have complete complementarity to the target gene or sequence or may comprise a region of mismatch (i e, an uncomplementary motif) . RNA interference includes "small
interfering RNA" or "siRNA, " e g , siRNA of about 15-60 (duplex) nucleotides in length, more typically about 15-30 (duplex) nucleotides in length, and is preferably about 20-27 (duplex) nucleotides in length (e.g., each complementary sequence of the double stranded siRNA is 15-60 nucleotides in length, preferably about 20-27 nucleotides in length. siRNA duplexes may comprise 3' overhangs of about 1 to 4 nucleotides and 5' phosphate termini. Examples of siRNA include, but are not limited to, a double stranded polynucleotide molecule assembled from two separate stranded molecules, wherein one strand is the sense strand and the other is the complementary antisense strand.
SiRNA are chemically synthesized, or may be generated by
cleavage of longer dsRNA (e.g., dsRNA greater than about 25 nucleotides in length) with the endogenous enzyme Dicer, which process the dsRNA into biologically active siRNA, or siRNA may be encoded by a plasmid (e.g., transcribed from the plasmid
(circular dsDNA) encoded information as single nucleotide strand that automatically fold into duplexes with hairpin loops) .
The term "nucleic acid" as used herein refers to a polymer containing at least two deoxyribonucleotides or
ribonucleotides in either single or double stranded form, it includes both DNA and RNA molecules. Not limiting forms of DNA molecules are antisense molecules and plasmid DNA. Not limiting examples of RNA molecules are siRNA, asymmetrical interfering RNA (aiRNA) , microRNA. Nucleic acids may include nucleotide analogs or modified backbone residues or linkages, which are synthetic, naturally occurring, and non-naturally occurring. Not limiting examples of such analogs are phosphorothioates , phosphoramidates , methyl phosphonates and peptide-nucleic acids (PNAs) .
The term "lipid" refers to a group of organic water insoluble compounds which are the basic components of biological membranes. Lipids are a broad group of molecules which includes fats, waxes, sterols, phospholipids. Lipids may be broadly defined as hydrophobic or amphiphilic molecules; the amphiphilic nature of some lipids allows them to form structures such as vesicles, liposomes, or membranes in an aqueous environment. A "lipid particle" or "liposomes" are micro or nanoparticles composed of one or more lipid bilayers, with an aqueous core, which is used herein to refer to a lipid formulation that can be used to deliver an active agent or therapeutic agent, such as a nucleic acid (e.g., a siRNA) , to a target site of interest. In the liposomes of the invention, which are formed from a cationic lipid, a neutral lipid, and a conjugated lipid (e.g., PEG-lipid) that prevents aggregation of the liposomes and increases blood stream circulation times, the therapeutic agents and/or diagnosis agent may be encapsulated in the liposomes, thereby protecting the agents from enzymatic degradation and improving the pharmacokinetics features. As used herein, the term "SALP" refers to stabilized antisense lipid particles and "SNALP" refers to a stabilized nucleic acid lipid particle. SALP is the term used to refer to a nucleic acid-lipid particle encapsulating asODN within the lipid particles. SNALP is the term used to refer to a nucleic acid-lipid particle
encapsulating siRNA within the lipid particles. SALP/SNALP represent particles made from lipids (e.g., a cationic lipid, a neutral lipid, and a conjugated lipid that prevents aggregation and increases blood stream circulation times) , encapsulating nucleic acids (e.g., siRNA, aiRNA, mlRNA, ssDNA, dsDNA, ssRNA, short hairpin RNA (shRNA) , dsRNA, or a plasmid, including plasmids from which an interfering RNA is transcribed) . As used herein, the term "SNALP" includes SPLP (stabilized plasmid particles; whenever the lipid particles encapsulate plasmid molecules) ; SNALP and SALP . SNALP can exhibit extended
circulation lifetimes following intravenous (I.V.) injection, they can accumulate at distal sites (e.g., sites physically separated from the administration site) , they can mediate expression of the transfected gene or silencing of target gene expression at these distal sites, they can mediate the efficient delivery of the non- nucleic acid-based therapeutic agents co- encapsulated with the nucleic acids.
The term "fusogenic" refers to the ability of a liposome, or other drug delivery system to fuse with membranes of a cell .
The term "cationic lipid" refers to any lipid that carries a net positive charge at a selected pH. Not limiting examples of cationic lipids are 1 , 2 -dioleoyl-3 -dimethylammonium- propane (DODAP) , 1 , 2 -dilinoleyloxy- 3 - (2 -N, - dimethylamino) ethoxypropane (DLin-EG-D A) , N, -dioleyl -N, - dimethylammonium chloride (DODAC) , 1 , 2 -dioleyloxy-N, N-dimethyl - 3 -aminopropane (DODMA) , 1 , 2 -distearyloxy-N, N-dimethyl -3 - aminopropane (DSDMA) , N- (1- (2 , 3 -dioleyloxy) propyl) -N, N, - trimethylammonium chloride (DOTMA) , N-(l-(2,3- dioleoyloxy) propyl ) -N, N, N-trimethylammonium chloride (DOTAP) , 3- (N- ( ' , ' -dimethylaminoethane) -carbamoyl ) cholesterol (DC-Choi) , N- (1, 2-dimyristyloxyprop-3-yl) -N, -dimethyl -N-hydroxyethyl ammonium bromide (D RIE) , 2 , 3 -dioleyloxy-N- [2 (spermine- carboxamido) ethyl] - , -dimethyl - 1 -propanaminium trifluoroacetate (DOSPA) , dioctadecylamidoglycylspermine (DOGS) , N, N-dimethyl - 3 , 4-didleyloxybenzylamme (DMOBA) , 1 , 2 -N, N ' -dioleylcarbamyl -3 - dimethylaminopropane (DOcarbDAP) , 1 , 2 -N, N ' -dilmoleylcarbamyl - 3 - dimethylaminopropane (DLincarbDAP) , 1 , 2 -dilinoleyloxy-N, N- dimethylaminopropane (DLinDMA) , 1 , 2 -dilinolenyloxy-N, N- dimethylaminopropane (DLenDMA) , 2 , 2 -dilinoleyl -4 - (2 - dimethylaminoethyl ) - [1,3] -dioxolane (DLin-K C2 -DMA) , 2,2- dilinoleyl -4 - (3 -dimethylarninopropyl ) - [1,3] -dioxolane (DLin-K- C3 -DMA) , 2 , 2 -dilinoleyl -4 - (4 -dimethylaminobutyl ) - [1,3] -dioxolane (DLin-K-C -DMA) , 2 , 2 -dilinoleyl - -dimethylaminomethyl - [1,3] - dioxolane (DLin-K-DMA) ; 1 , 2 -dilinoleylcarbamoyioxy-3 - dimethylaminopropane (DLin-C-DAP) , 1 , 2 -dilinoleyoxy- (dimethylamino) acetoxypropane (DLin-DAC) , 1 , 2 -dilinoleyoxy-3 - morpholinopropane (DLin-MA) , 1 , 2 -dilinoleoyl -3 - dimethylaminopropane (DLinDAP) , 1 , 2-dilinoleylthio-3- dimethylaminopropane (DLin-S-DMA) , l-linoleoyl-2 -linoleyloxy-3 - dimethylaminopropane (DLin-2 -DMAP) , 1 , 2 -dilinoleyloxy-3 - trimethylaminopropane chloride salt (DLin-TMA. Cl ) , 1,2- dil inoleoyl -3 - trimethylaminopropane chloride salt (DLin-TAP . Cl ) , 1 , 2 -dilinoleyloxy-3 - (N-methylpiperazino) propane (DLin-MPZ) , 3- (N, N-dilinoleylamino) -1 , 2 -propanediol (DLinAP) , or mixtures thereof .
The term "neutral lipid" refers to any lipid that exist either in an uncharged or neutral z itterionic form at a selected pH. The neutral lipid components may be cholesterol or a derivative, phospholipids, or a mixture of phospholipids and cholesterol or a derivative. Examples of cholesterol derivatives include, but are not limited to, cholestanol, cholestanone , cholestenone , coprostanol, cholesteryl-21 -hydroxyethyl ether, cholesteryl -41 - hydroxybutyl ether, and mixtures thereof.
Examples of neutral lipids include but are not limited to dipalmitoylphosphatidylcholine (DPPC) ,
distearoylphosphatidylcholine (DSPC) ,
dioleoylphosphatidylethanolamine (DOPE), palmitoyloleoyl - phosphatidylcholine (POPC) , palmitoyloleoyl - phosphatidylethanolamine (POPE) , palmitoyloleyol - phosphatidylglycerol (POPG) , dipalmitoyl- phosphatidylethanolamine (DPPE) , dimyristoyl- phosphatidylethanolamine (DMPE) , distearoyl- phosphatidylethanolamine (DSPE) , monomethyl- phosphatidylethanolamine, dimethyl -phosphatidylethanolamine , dielaidoyl- phosphatidylethanolamine (DEPE) , stearoyloleoyl- phosphatidylethanolamine (SOPE) , egg phosphatidylcholine (EPC) , and mixtures thereof .
"Increased blood stream circulation," as used herein, refers to a broad biodistribution of a therapeutic agent such as siRNA within an organism through the improvement of the time in circulation of the delivery vehicle. The enhancement of the amount of the active agent available to be exposed to most parts of the body is generally achieved by decreasing degradation and/or blood clearance (such as by first pass organs (liver, spleen, lung, etc.) and nonspecific cell binding.
The term "cancer" is the designation adopted for a group of more than 100 human diseases that have in common the uncontrolled cell growth (division beyond the normal cell control), invasion (intrusion of adjacent tissues), and
sometimes metastasis (spread to other locations in the body) . Cancers are caused by abnormalities in the genetic material of the transformed cells. These abnormalities may be due to the effects of carcinogens, such as tobacco smoke, radiation, chemicals, or infectious agents. Cancer promoting genetic abnormalities may be caused by random errors in DNA replication, or are inherited. The term "leukemia" refers to a broad group of
diseases characterized by accumulation of malignant white cells in the bone marrow and blood that do not tend to form solid masses of cells (Hoff'brand et al . 2006a; Ottensmeier 2001). This accumulation of abnormal white cells leads to bone marrow failure and infiltration of leukemia cells in different organs (e.g. liver, brain, skin, spleen) {Hoff-brand et al . 2006a) .
Leukemias can be classified in four main types, the acute and chronic leukemias, which are subdivided into lymphoid and myeloid leukemias. Chronic leukemias are distinguished from acute by their slower progression in opposition to the
aggressive character Of the acute leukemias. Myeloid and
lymphoid leukemias differ in the hematopoietic lineage affected by the malignant transformation; myeloid leukemias are
originated in the myeloid lineage and lymphoid leukemias in the lymphoid lineage (Hoffbrand et al . 2006a) .
Chronic myeloid leukemia is a myeloproliferative disease originated from hematopoietic stem cells, and was the first human malignancy to be linked with a dominant acquired genetic mutation. Chronic myeloid leukemia is caused by a translocation between chromosomes 9 and 22 which create the oncogene BCR-ABL and an abnormal 22 chromosome (Philadelphia chromosome; Ph) . There are no known predisposing factors, except the exposure to ionizing radiation, since the incidence of CML was greatly increased in survivors of the atomic bomb exposures in Japan. No inheritable factors have been identified, although it is assumed that an acquired genetically chromosomal
instability might be the precondition for the translocation (Hoffbrand et al . 2006b; Pasternak et al . 19.98) .
The terms "silencing", "knockdown the expression of " and "down -regulation the expression of," herein refer to the at least partial suppression of the expression of the target gene as assessed by the reduction of the amount of the target mRNA.
The term "lipid conjugate" refers to a lipid conjugated to a shielding group that inhibits aggregation of nucleic acid lipid particles and increase blood stream
circulation times. The incorporation of hydrophilic
carbohydrates or polymers, as shielding groups, on the liposome surface extends the liposome half-life from a few minutes
(classical non-stealth liposomes) to several hours (stealth liposomes) (Immordino et al . 2006; Sapra and Allen 2003) . Such lipid conjugates include, but are not limited to,
poly (acrylamide) , poly (vinylpyrrolidone) ,
poly (aeryloylmorpholine) , pol (2-ethyl-2-oxazoline) , poly (2- methyl -oxazoline) , phosphatidyl polyglycerols , polyvinyl
alcohols, polysaccharides, PEG and PEG-containing copolymers (as poloxamers and poloxamines) (Owens and Peppas 2006; Torchilin 2009; Torchilin 2006), and mixtures thereof. PEG is a polyether diol which provides a very attractive combination of properties, such as solubility in aqueous and organic media, high
lity of its polymer chain, very low toxicity,
immunogenicity, and antigenicity (Immordino et al . 2006;
Torchilin 2006) . In addition, it presents the lowest level of protein or cellular adsorption of any known polymer and has been FDA-approved for many injected biotech products. Not limiting examples of PEG- lipid conjugates are: i) PEG- phosphatidylethanolamine ; ii) PEG-ceramide ; iii) PEG- diacylglycerol ; iv) PEG-dialkyloxypropylamine and v) PEG-1- methyl-4- (cis-9-dioleyl) methyl -pyridinium chloride (PEG-SAINT) (Romberg et al . 2008).
The term "post insertion" refers to a 1 igand coupling method in which 1 igands attached to a conjugated lipid are introduced into preformed liposomes.
The term "micelles" refers to an aggregate of surfactant molecules dispersed in a liquid. Generally, micelles in aqueous solution form an aggregate with the hydrophilic regions "head" in contact with the surrounding solvent (water) , whereas, the hydrophobic single tail regions are in the micelle cent e .
The term "antisense oligonucleotides" or "asODN" are single stranded molecules of RNA or DNA usually with 18-25 nucleotides in length that hybridize in a sequence specific manner to the target mRNA/DNA and down regulate the target gene, Description of the embodiments
The lipid particles of the present invention provides, the possibility to target a cell population and two or more molecular targets simultaneously using one single nanocarrier. Thus, the drug delivery nanocarrier is targeted for specific organ, tissue or cells by coupling of targeting ligands to its surface. Not limiting examples of targeting moieties include peptides, polypeptides, antibodies, polyclonal antibodies, monoclonal antibodies, antibody fragments, humanized antibodies, recombinant antibodies, recombinant human antibodies, proteins and cell surface ligands. The ligands are linked to the surface of the nanocarrier in a way that it is able to interact with a specific molecule, protein, glycoprotein and/or cell surface receptor that is overexpressed or specifically expressed on the cell surface of a specific cellular target. An appropriate spacer can be positioned between the nanocarrier and the ligand to avoid hindrance on the interaction between the ligand and its target. The nanocarrier can allow multivalent cellular
targeting, presenting more than one homing ligand that
selectively homes the delivery agent to specific molecules on the target cells.
The nanocarrier should have a diameter comprised between 100 and 200 nm to travel in the blood stream circulation without occluding circulation and without being rapidly removed from circulation by first passage organs; being available to specifically interact with the target cells in therapeutic dosages .
The drug delivery nanocarrier of the present invention provides a method for simultaneous encapsulation, adsorption or complexation of two or more gene silencing agents (nucleic acids) and two or more non-nucleic acid based active agents (therapeutic drugs) . The gene silencing agents (nucleic acids) may comprise, but not limited to plasrnids, antisense
oligonucleotides (asODN) , siRNA, mi NA, shRNA, aiRNA, DNA enzymes (DNAzymes) , Ribozymes, DNA decoys, Aptamers and mixtures thereof. The gene silencing agents may comprise modified nucleotides including, but not limited to phosphorothioate linkages, 2 ' -O-methyi (2'OMe) nucleotides, 2 ' -deoxy-2 ' - fluoro (2'F) nucleotides, 21 -deoxy nucleotides, 2 ' -0- (2 -methoxyethyl ) (MOB) nucleotides, locked nucleic acid (LNA) nucleotides, and mixtures thereof. The gene silencing agents may comprise at least one or a cocktail (e.g., at least two, three, four, five, six, seven or more) of unmodified and/or modified gene silencing agents. The cocktail of gene silencing agents may comprise sequences which are directed to the same region or domain and/or to different regions or domains of one or more target genes. The gene silencing may be performed by any viral vector capable of accepting the coding sequences for the gene silencing agent, including but not limited to retrovirus, herpes virus,
adenovirus and adenoassociated virus.
Non-nucleic acid based active agents (therapeutic drugs) may include, but not limited to chemotherapy drugs, hormonal therapeutic agents, immunotherapeutic agents, antiviral drugs, anti - inflammatory compounds, antidepressants, stimulants, analgesics, antibiotics, antipyretics, vasodilators, an i -angiogenics, c o ascular agents, signal transduction inhibitors, anti -arrhythmic agents, hormones, vasoconstrictors, and steroids .
Non-limiting examples of chemotherapy drugs include platinum-based drugs (e.g. cisplatin, carboplat in, etc) ;
alkylating agents (e.g., cyclophosphamide, chlorambucil, busulfan, melphalan, lomustine, carmustine, estramust ine , treosulfan, thiotepa, mitobronitol , etc); anti-metabolites (e.g., 5-fluorouracil , methotrexate, capecitabine, cytarabine, fludarabine, gemcitabine, cladribine, raltitrexed,
mercaptopurine, etc); plant alkaloids (e.g., , vincristine, vinblastine, vindesine, paclitaxel, docetaxel, etc) ,
topoisomerase inhibitors (e.g., irinotecan, topotecan,
etoposide , etc); cytotoxic antibiotics (e.g., doxorubicin, adriamycin, daunorubicin, epirubicin, actinomycin, bleomycin, mitomycin, mitoxantrone , aclarubicin, idarubicin, dactinomycin, etc ), taxanes (e.g., docetaxel, paclitaxel); tyrosine kinase inhibitors (e.g., gefitinib, sunitinib, erlotinib, lapatinib, canertinib, semaxinib, vatalanib, sorafenib, imatinib,
dasatinib, leflunomide, vandetanib, pharmaceutically acceptable salts thereof, stereoisomers thereof, analogs thereof,
derivatives thereof, and mixtures thereof; anti-inflamatory agents such as but not limited to ibuprofen, aceclofenac, acemetacin, acid acetilsalicilic, azapropazone , celecoxib, diclofenac sodium, diflunisal, cetodolac, fenbufen, fenoprofen, flubiprofen, indomethacin, acetaminocin, piroxicam, rofecoxib, sulindac, tenoxicam,- antiangiogenic agents or angiolytic agents such as but not limited to angiostatin (plasminogen fragment) , antiangiogenic antithrombin III, vasculostatin, vasostatin and mixtures thereof.
The nanocarrier is composed, but not limited, by liposomes and/or polymers. Not limiting examples of liposomes are stabilized nucleic acid lipid particles (SNALP) . SNALP liposomes comprise one or more cat ionic lipids, one or more neutral lipids and one or more conjugated lipid that inhibits aggregation of particles and provide long circulation times to the liposomes. The cationic lipids may comprise from 10 to 60 mol%, the neutral lipid may comprise from 10 to 70 mol%, the conjugated lipid that inhibits aggregation and provides long circulation times may comprise from 1 to 10 mol%. Not limiting examples of cationic lipids are 1,2- dioleoyl-3-dimethylammonium-propane (DODAP) , 1 , 2 -dilinoleyloxy- 3-(2-N,N- dimethylamino) ethoxypropane (DLin-EG-DMA) , N,N- dioleyl-N,N-dimethylammonium chloride (DODAC) , 1 , 2 -dioleyloxy- N, -dimethyl -3 -aminopropane (DODMA) , 1 , 2 -distearyloxy-N, N- dimethyl -3 -aminopropane (DSDMA) , N- (1 -{2,3 -dioleyloxy) propyl) - Ν,Ν,Ν- trimethyl ammonium chloride (DOTMA) , N-(l-(2,3- dioleoyloxy) propyl) -N, N, N-trimethylammonium chloride (DOTAP) , 3- (N- (Ν' ,Ν1 -dimethylaminoethane) -carbamoyl ) cholesterol (DC-Choi) , N- (1 , 2-dimyristyloxyprop-3-yl) -N, N-dimethyl -N- hydroxyethylammonium bromide (DMRIE) , 2 , 3 -dioleyloxy-N- [2 (spennine-carboxamido) ethyl] -N, -dimethyl - 1 - propanaminiumtrifluoroacetate (DOSPA) ,
dioctadecylamidoglycylspermine (DOGS), N, -dimethyl -3 , - dioleyloxybenzylamme (DMOBA) , 1 , 2 -N, 1 -dioleylcarbamyl -3 - dimethylammopropane (DOcarbDAP) , 1 , 2-N, ' -dilmoleylcarbamyl-3 - dimethylaminopropane (DLincarbDAP) , 1 , 2 -dilinoleyloxy-N, N- dimethylaminopropane (DLinDMA) , 1 , 2 -dilinolenyloxy-N, N- dimethylaminopropane (DLenDMA) , 2 , 2-dilinoleyl-4- (2- dimethylaminoethyl) - [1, 3] -dioxolane (DLin-K-C2 -DMA) , 2,2- dilinoleyl-4- (3-dimethylarninopropyl) - [1,3] -dioxolane (DLin-K- C3 -DMA) , 2 , 2 -dilinoleyl -4- (4 -dimethylaminobutyl ) - [1,3] -dioxolane (DLin-K-C4 -DMA) , 2 , 2 -dilinoleyl -4 -dimethylaminomethyl - [1,3] - dioxolane (DLin- -DMA) , 1 , 2 -dilinoleylcarbamoyloxy-3 - dimethylaminopropane (DLin-C-DAP) , 1 , 2 -dilinoleyoxy-
(dimethylamino) acetoxypropane (DLin-DAC) , 1 , 2 -dilinoleyoxy-3 - morpholinopropane (DLin-MA) , 1 , 2 -dilinoleoyl -3 - dimethylaminopropane (DLinDAP) , 1 , 2 -dilinoleylthio-3 - dimethylaminopropane (DLin-S- DMA), 1-linoleoyl -2 -linoleyloxy-3- dimethylaminopropane (DLin-2 -DMAP) ., 1 , 2 -dilinoleyloxy-3 - trimethylaminopropane chloride salt (DLin-TMA. CI) , 1,2- dilinoleoyl-3-trimethylaminopropane chloride salt (DLin-TAP . Cl ) , 1, 2-dilinoleylbxy-3- (N-methylpiperazino) propane (DLin-MPZ) , 3- (N,N-dilinoleylamino) -1 , 2 -propanediol (DLinAP) , or mixtures thereof. The neutral lipid components may be cholesterol or a derivative; phospholipids; or a mixture of phospholipids and cholesterol or a derivative thereof. Examples of cholesterol derivatives include, but are not limited to, cholestanol, cholestanone, cholestenone , coprostanol, cholesteryl -2 ' - hydroxyethyl ether, cholesteryl-41 - hydroxybutyl ether, and mixtures thereof. Examples of neutral lipids include but are not limited to dipalmitoylphosphatidylchol ine (DPPC) ,
distearoylphosphatidylcholine (DSPC) ,
dioleoylphosphatidylethanolamine (DOPE) , palmitoyloleoyl - phosphatidylcholine (POPC) , palmitoyloleoyl - phosphatidylethanolamine (POPE) , palmitoyloleyol- phosphatidylglycerol (POPG) , dipalmitoyl- phosphatidylethanolamine (DPPE) , dimyristoyl- phosphatidylethanolamine (DMPE) , distearoyl- phosphat idylethanolamine (DSPE) , monomethyl- phosphatidylethanolamine , dimethyl -phosphatidylethanolamine , dielaidoyl- phosphatidylethanolamine (DEPE) , stearoyloleoyl - phosphatidylethanolamine (SOPE) , egg phosphatidylcholine (EPC) , and mixtures thereof.
Not limiting examples of biocompatible, soluble and hydrophilic polymers with a highly flexible main chain used to prepare long circulating liposomes and avoid particles
aggregation during preparation are poiy (acryl amide), poly (vinyl pyrrol idone ) , poly (acryloyl morpholine) , poly (2 -ethyl -2 - oxazoline) , poly (2 -methyl -oxazoline) , phosphatidyl
polyglycerols, polyvinyl alcohols, polysaccharides, PEG and PEG- containing copolymers (as poloxamers and poloxamines) , or mixtures therefore. Examples of PEG-lipid particles include but are not limited to PEG-diacylglycerol (DAG) , PEG
dialkyloxypropyl (DAA) , PEG-ceramide (Cer) , PEG- phosphatidylethanolami e , PEG- 1 -methyl -4 - ( cis - 9 -dioleyl ) methyl - pyridinium chloride (PEG-SAINT) or mixtures thereof. Examples of PEG-Cer conjugate include but are not limited to PEG- dilauryloxypropyl (C 12), a PEG- dimyristyloxypropyl (C 14), a PEG-dipalmityloxypropyl (C 16) , a PEG-distearyloxypropyl (C 18) , or mixtures thereof. The PEG moiety of the PEG-lipid conjugates described herein may compose an average molecular weight ranging from about 550 da1tons to about 10 000 daltons.
The present invention will be described in detail through specific examples. Examples are presented for
illustrative purposes, and do not limit the invention in any manner, since some noncritical parameters can be modified to yield essentially the same results.
Examples ^Materials and Methods
Example 1- Preparation of targeted nanocarrxers co-encapsulating siRNA and imati ib
Preparation of Trf -coupled or BSA-coupled micelles:
Coupling of Trf or BSA to PEG2ooo~DSPE micelles was performed accordingly to Ishida et al (Ishida et al . 1999) . Briefly, Trf or BSA protein was modified with the addition of thiol groups through reaction with 2 -iminothiolane hydrochloride (2-IT) . For this purpose, Trf or BSA and 2-IT freshly dissolved in HEPES buffer (20 mM HEPES, 145 mM NaCl , pH 8) were mixed in a
protein/2-IT molar ratio of 1/10 and gently stirred for 1 h, in the dark at room temperature.
A lipid film of DSPE-PEG-MAL was prepared by solvent evaporation under a mild stream of N2 and further dried under vacuum for 2 h. This dried lipid film was then hydrated with MES buffer (20 mM HEPES, 20 mM MES, pH 6.5), at a concentration above 2.3 μΜ, the critical micellar concentration of the lipid (Ishida et al . 1999) . Micelles were formed by strong vortex followed by 15 s heating in a water bath at 38 °C, followed by a second vortex shaking. Then, the freshly thiolated protein was coupled to the freshly prepared DSPE-PEG-MAL micelles by a thioesther linkage (protein to micelles molar ratio of 1:1) . The coupling reaction was performed overnight, ih the dark at room temperature with gentle stirring. The remaining free MAL groups in the micelles were quenched by the addition of β
mercaptoethanol at a maleimide : β mercaptoethanol molar ratio Of 1:5, under stirring for 30 min at room temperature. Encapsulation of siRNA into the nanocarriers : a solution containing 13 μπιοΐ of total lipid composed of Choi:
DSPC:DODAP: mPEG 2000 Ci6Ceramide (45:22:25:8, mol%) in 200 μΐ of absolute ethanol , and a solution of 0.041 μτηοΐ of siRNA in 300 μΐ of 20 mM citrate buffer, pH 4, were heated at 60 °C. The lipids were then slowly added under strong vortex to the siRNA solution. In some experiments, empty liposomes were used. In this case, lipids were added to 300 μΐ of citrate buffer under similar conditions as those used for siRNA-encapsulating
liposomes preparation. Upon their formation the liposomes were extruded, 21 times, in a LipoFast mini extruder (Lipofast,
Avestin, Toronto, Canada) through 100 nm diameter polycarbonate filters (Avestin, Toronto, Canada) . Then, a dialysis was
performed in HBS, pH 7.4, through regenerated cellulose tubular membrane with MWCO 6000-8000 (Cellu Sep T2 , Membrane Filtration Products, Inc Seguin, TX, USA) during 3 h at room temperature to remove ethanol and raise the external pH. Subsequently, the total lipid concentration was assessed by cholesterol
quantification. For this purpose, samples were added to absolute ethanol (1/6, v/v) and Infinity™ Cholesterol Liquid Stable
Reagent (Thermo Electron; Melbourne, Australia) . Absorbance was measured at 500 nm in a spectrophotometer and the concentration assessed against a cholesterol standard curve. The cholesterol quantification allowed the determination of the total lipid that remained at this stage and consequently the determination of the amount of imatinib and micelles to be added. Encapsulation of imatinib in siRNA-containing liposomes and post-insertion of Trf at the liposome surface:
Immediately after the dialysis of the liposomes containing siRNA (performed as described previously) , imatinib was encapsulated into the liposomes, by addition of imatinib to the siRNA- containing liposomes at different imatinib/total lipid molar ratios (1/3, 1/8, 1/16, 1/32, 1/42; initial imatinib/total lipid) and incubation for 1 h, at 60 °C, in a water bath. The liposomes were then allowed to reach the room temperature and 4 mol % of Trf -micelles was added and incubated for 17 h, at 38 -C, in a water bath under dark.
Purification of liposomes: After incubation with micelles, Trf -liposomes were purified by size exclusion
chromatography on a Sepharose CL-4B column, using HBS , pH 7.4, as running buffer to remove external siRNA and imatinib as well as chemical reagents used during the liposomal preparation.
SiRNA quantification: The amount of siRNA entrapped inside liposomes was assessed by the Quant -iT RiboGreen RNA Assay (Molecular Probes, Invitrogen, Karlsruhe, Germany) against a siRNA standard curve. Liposomes were dissolved upon addition of 0.6 m of octaethylene glycol monododecyl ether (C12E8) and the RiboGreen fluorescence (Xex 485 nm, Xem 530 nm, cut off 515 nm) was measured using a Spectra Max Gemini EM plate reader fluorimeter (Molecular Devices, Sunnyvale, CA) .
Imatinib quantification: The method for imatinib quantification was developed by adaptation of the Dharmacon RNA Technologies (Lafayette, CO, USA) protocol for siRNA
precipitation. In microfuge tubes, 0.1 μτηοΐ of total lipid was added to the precipitation reagent (400 μΐ destilled water, 40 μΐ of 10 M ammonium acetate, pH 7 and 1.5 ml absolute ethanol) up to 800 μΐ , samples were then submitted to 30 s of strong vortex and transferred to -80 °C/2 h or -20 °C/overnight .
Subsequently, frozen samples were slightly thawed at room temperature and centrifuged at 18 000 g for 20 min at 4 °C.
Imatinib concentration was determined in the supernatant by measuring the absorbance at 259 nm against a standard curve of imat inib. This quantification method was optimized to eliminate any interference by the other components of the formulation.
Example 2- Cell viability of targeted nanocarriers co- encapsulating siRNA and imatinib Cell lines: Two human chronic myeloid leukemia cell lines in blast crisis, positive for BCR-ABL oncogene, with the b3a2 translocation (K562 and LAMA- 84 cells) purchased from DSMZ (Braunschweig, Germany) were maintained in culture at 37 °C, 5% C02 under humidified atmosphere in RPMI-1640 ιηεάίμΓη supplemented with 10% (v/v) heat -inactivated fetal bovine serum (FBS) (Gibco, invitrogeil, California, USA) , penicillin (100 U/ml) and
streptomycin (100 μg/ml) (Cambrex, NJ, USA) .
Development of imatinib-resistant cell line: K562 cells maintained in culture as previously described, were incubated with increasing concentrations of imatinib, starting at 0,05 μΜ and with 0.05 μΜ increments every 4 days of culture, until cells acquired the ability to grow at 1 μΜ. At this time point, drug resistance was assessed and cells were designated as IRK562. The new cell line was maintained continuously in culture in the presence of 1 μΜ of imatinib and was washed with drug- free medium before all experimental procedures.
Cell transfection: K562 and LAMA- 8 cells (20 000 cells/well) and IRK562 cells (25 000 cells/well) in RPMI-1640 culture medium supplemented with 10% FBS and antibiotics were seeded in 96-round well plates. Cells were transfected with Trf- associated liposomes co-encapsulating siRNA and imatinib at different molar ratios at 37 °C for 4 h. After incubation with liposomes, the medium was replaced with fresh medium and cells further incubated for 44 h. Cell viability: Cell viability was evaluated by the resazurin reduction assay (O'Brien et al . 2000). The assay measures the chemical reduction of the resazurin dye resulting from cellular metabolic activity, and allows the determination of viability over the culture period without harming the cells. Briefly, the culture medium was replaced with 10% (v/v)
resazurin dye in RPMI-1640 medium without serum and antibiotics, which was added to each well. After 2.5 h of incubation at 37 °C, the absorbance at 540 nm (reduced form) and 630 nm (oxidized form) was measured in a microplate reader Multiskan Ex (Thermo Labsystems, Vantaa, Finland) . Cell viability was calculated as percentage of control cells using the equation:
[ (A54o-A63o) treated cells xlOO) / (A540-A63o) control cells]
Assessment of imatinib IC50 and DRI : The required drug concentration to promote reduction of 50% in cell viability (IC50) and dose reduction index (DRI), which is the magnitude of dose reduction allowed for a drug when given in a drug
combination (IC50 of the drug when administered alone/ ICS0 of the drug when administered in the combination) were assessed. For IC50 determination non-linear curve fit assuming sigmoidal dose- response was performed. Example 3- Cell association and cell internalization of Trf - or BSA- associated nanocarriers
Flow Cytometry studies: To evaluate the extent of cell association of Trf- or BSA-associa ed liposomes or NT-liposomes , cells were transfected with fluorescently labelled siRNA (Cy3- siRNA) encapsulated in each one of the mentioned liposomes.
Cells were seeded in RPMI-1640 culture medium (200 000
cells/well) in 48-well plates. To demonstrate that the receptor- mediated internalization is temperature -dependent , cells were incubated 1 h at 4 ° or at 37 °C, prior to liposome addition. To demonstrate that the internalization of Trf -liposomes is mediated by TrfR, this receptor was saturated by incubating the cells during 30 min with 125 μΜ of free holo-transferrin
(dissolved in HBS, pH 7.4), as performed by others (Cardoso et al. 2008; Chiu et al . 2006) , After these pre -treatments , cells were incubated with the formulations under study for h at 37 CC or 4 °C. Subsequently, cells were collected in conic tubes (BD, Biosciences) , washed twice with cold PBS and re-suspended in 500 L of cold PBS. All samples were immediately analyzed in a FAGS Calibur flow cytometer (BD, Biosciences) . Cy3
fluorescence was evaluated in the FL-2 channel. The data were then analyzed in the Cell Quest software (BD) . Cell association was only assessed in viable cells, these being gated based on the morphological features and on Trypan Blue staining (FL-3 channel) . The cell association efficiency of each formulation was assessed by comparing the relative fluorescence units (RFU) increase relative to non-treated cells (RFU of cells treated with the formulation under study/RFU of non-treated cells) . Confocal microscopy studies: In order to assess the extent of cellular internalization of the developed liposomes, confocal microscopy studies on transfected cells were performed. Cells were transfected with the formulations under study as described in the previous section. Then, cells were collected in conic tubes (BD, Biosciences) , washed with cold PBS, fixed with
4% paraformaldehyde for 20 min in the dark, at room temperature, washed with cold PBS, stained with the fluorescent DNA-binding dye Hoechst 33342 (Molecular Probes, Oregon, USA) (1 /xg/mL) for 5 min, in the dark, washed with cold PBS, and mounted in Mowiol mounting medium (Fluka, Sigma) . Confocal images were acquired in a point scanning confocal microscope Zeiss LSM 510 Meta (Zeiss, Germany) , with a 40x EC Plan-Neofluar or with a 63x Plan- Apochromat oil immersion objectives, and an argon (488nm) , a DPSS (561 nm) , a diode (405 nm) and a helium-neon (633 nm) lasers. Preparations were excited at 561 nm for the Cy3 (em: >560 nm) and at 405 nm for Hoechst 33342 (em: 420-480 nm) . Differential interference contrast (DIC) images were obtained using the helium-neon (633 nm) laser. Digital images were acquired using the LS 510 Meta software. All instrumental parameters pertaining to fluorescence detection and image analyses were held constant to allow sample comparison.
Results :
As described in Example 1, after the encapsulation of siRNA in SNALP liposomes, a transmembrane pH gradient between the aqueous content of the liposome (citrate buffer, pH 4) and the external liposome milieu (HBS, pH 7.4) was generated, which is the driving force for the active encapsulation of the imatinib. Trf-PEG-DSPE conjugates were then inserted onto the pre-formed liposomes by the post - insertion method. Using this methodology, the effect of imatinib on siRNA encapsulation yields and the effect of siRNA on imatinib encapsulation was assessed upon incubation with different imatinib : lipid ratios (1/3; 1/8; l/l6; 1/32; 1/42). As illustrated in Figure 1A, the encapsulation yields of imatinib increase with decreasing of imatinib : total lipid ratios, being 11.88 ± 2.09% for the 1/3 ratio and of 19.8 ± 2.32% for the 1/8 ratio. For ratios above 1/16, the encapsulation yields were very similar, being around 25%. As can be observed in Table I, for ratios exhibiting the same yield, the higher loading of imatinib is obtained with increasing imatinib/lipid ratios (i.e. 1/16 > 1/32 > 1/42). It is important to notice that the presence of siRNA significantly enhances the imatinib encapsulation yields. For example, at the ratio of 1/8 the encapsulation yield of imatinib increase from 5.96 ± 2.39%, in liposomes without siRNA, to 19.80 ± 2.32% when imatinib is co-encapsulated with siRNA.
As can be observed in Figure IB, the siRNA encapsulation yield is also significantly affected by the presence of imatinib, namely by the imatinib : total lipid ratio used in the co-encapsulation process. Thus, the formulations prepared with higher amounts of imatinib (higher imatinib : total lipid ratios, e.g. 1/3) resulted in lower siRNA encapsulation yields, as compared to the formulations prepared with lower amounts of imatinib (lower imatinib : total lipid ratios, e.g. 1/42} . When siRNA is encapsulated alone, under the same
conditions as those in this co-encapsulating process, the encapsulation yield is 92.17 + 1.39%, (Mendonca et al . 2010). Overall, these results demonstrate that when imatinib is co- encapsulated with siRNA, the siRNA encapsulation yield decreases in a manner dependent on the imatinib/total lipid ratio pre- incubated with the liposomes. However, for ratios lower than 1/16, no significant difference in the siRNA loading of the formulations prepared was observed (Table I) . Nevertheless, the imatinib loading is smaller for the lower ratios (Table I), allowing to reach higher siRNA/imatinib ratios (Figure 1C) .
Thus, the lowest imatinib/lipid ratio tested (1/42) resulted in the highest siRNA/imatinib ratio (0.63). At this ratio it is therefore possible to obtain therapeutic concentrations of both imatinib and siRNA inside the same liposome. In contrast, the 1/3 ratio at which the imatinib loading is too high as compared to that of siRNA, did not allow to reach therapeutic
concentrations for both agents, in fact, with this formulation the achievement of therapeutic concentrations for siRNA will lead to extremely cytotoxic imatinib concentration, which would block the possible therapeutic contribution from the siRNA molecules. Trf-liposomes co-encapsulating imatinib and siRNA molecules prepared from imatinib : total lipid ratios of 1/16, 1/32 and 1/42 (1/16; 1/32 and 1/42 formulations) , resulted in 0.15, 0.35 and 0.63 siRNA/imatinib molar ratio, respectively (formulations are codenamed by the resulting siRNA/imatinib molar ratios) . In what concerns to the role of imatinib in the siRNA entrapment yield, a remarkable decrease in the siRNA yield of encapsulation was observed for the highest ratios of
imatinib/total lipid, whereas no impact was observed for the lower imatinib/lipid ratios, as compared to the encapsulation of siRNA in the absence of imatinib. Thus, as observed by others, we have shown that the inclusion of a second drug may induce leakage of the first encapsulated drug (Saxon et al . 1999b;
Waterhouse et al . 2001), in a drug amount -dependent manner. In opposition, the presence of siRNA enhances the imatinib
encapsulation yields. This can be explained by interactions between the negatively charged siRNA and the positively charged imatinib (at the intra-liposomal milieu) , which may increase the amount of imatinib entrapped inside the liposomes. As described in Example 2, the formulations that allow to obtain therapeutic concentrations for both imatinib and siRNA within the same liposome formulation were tested against cell lines sensitive to imatinib ( 562 and LAMA-84) as well as against the imatinib-resistant IR 562 cell line. As can be observed in Table II, for all tested cell lines the formulation with higher amount of anti - BCR -ABL siRNA (0.63) led to higher imatinib ICS0 reduction and, consequently, to higher dose reduction index (DRI) . IRK562 cells were more sensitive to the increment of the siRNA dose in the combination of siRNA and imatinib than the imatinib sensitive cell line LAMA-84, since for the 1/16 and 1/42 formulations the DRI was of 1.16 and 3.84, respectively, whereas for LAMA-84 the difference between the DRI obtained for the different formulations is not so evident. In fact, lowe siRNA/ imatinib ratios were required to reach the same DRI in LAMA-8 cells as compared to K562 and IRK562 cells, indicating that cell lines with higher BCR-ABL oncogene levels are more dependent on the gene silencing agent concentration (levels of BCR -ABL mRNA : IR 562 > K562 >LAMA-84, as assessed by qRT-PCR, data not shown) . For all tested cell lines, the 1/42 formulation (highest relative siRNA contribution in the drug combination) led to the highest imatinib ICSo reduction,
demonstrating the importance of achieving a level for both molecules within the range of therapeutic concentrations.
Results also revealed that the imatinib-resistant cell line IRK562 required higher siRNA/imatinib ratios, as compared to non-resistant cell line LAMA- 8 . A correlation between the cellular response and the expression of Trf receptor and BCR-ABL mRNA levels could be established. In fact, the cell line with higher Trf receptor expression and lower BCR-ABL mRNA levels ( LAMA-84) demonstrated higher response to the tested
formulations. Therefore, even considering that other cellular features may play relevant roles in the response to targeted therapy, our results strongly suggest that both the levels of Trf receptors and BCR-ABL mRNA can be used as biomarkers to predict the efficacy of the developed therapies. Cytotoxicity of encapsulated and free imatinib was also tested. LAMA- 84 cells were also treated with the combination of different
concentrations of free imatinib with 1 μΜ of siRNA encapsulated in Trf -liposomes (Table II) . Such treatments require higher siRNA/ imatinib ratios than that required by the strategy in which both molecules are co-encapsulated in the same liposome. In fact, for the combination of imatinib and encapsulated siRNA, the siRNA/ imatinib ratios used were 2.5 to 200, which led to a 2.85 DRI, whereas with the 1/42 formulation, in which
siRNA/ imatinib ratio is 0.63, a higher DRI (3.43) was obtained. Thus, upon co-encapsulation of siRNA and imatinib in the same liposomal particle, lower siRNA/ imatinib ratios can be employed to induce a certain degree of cytotoxicity as compared to the co- treatment of the cells with encapsulated siRNA and free imatinib. Both free imatinib and imatinib encapsulated in Trf- liposomes promoted similar cytotoxicity, suggesting that these Trf -coupled liposomes loading imatinib allow efficient
intracellular drug delivery (Table II) . As described in Example 3, the extent of association of liposomes bearing Trf or BSA attached at the end of PEG chains or of NT- liposomes , encapsulating Cy3 -labelled siRNA, to LAMA- 84 cells was assessed by flow cytometry. As can be observed in Figure 2A, the presence of Trf attached to the end of PEG- grafted lipids is the main responsible for the observed extent of iiposome-cell association. Cells incubated with BSA- liposomes or NT-liposomes revealed no significant cell association, illustrated by the low increase of the fluorescence intensity of the transfected cells as compared to non-treated cells. As can be observed, the RFU fold increase for cells treated with 2.0 μΜ siRNA encapsulated in Trf-liposomes (2.0 μΜ siRNA Trf) was 7.20 ± 0.40, in contrast to 3.20 ± 0.07 and 1.57 ± 0.22 fold increase observed upon treatment of the cells with BSA-liposomes and NT- liposomes, respectively. Our results also indicate that the extent of cell association of Trf-coupled liposomes containing
Cy3 - siRNA is dependent on the lipid concentration incubated with the cells up to 0.34 mM of total lipid which is correspondent to 1.0 μΜ siRNA, since no significant difference on the RFU of cells treated with 1.0 μΜ or 2.0 μΜ siRNA encapsulated in Trf- liposomes was observed.
To clearly demonstrate that the ligand Trf coupled at the surface of the liposomes trigger their cell internalization through theTrfR, competitive inhibition studies were performed. Cells were incubated with 125 μΜ of free human holo-Trf to block the TrfR. As illustrated in Figure 2B, a drastic decrease of cell association of Trf -liposomes for all tested concentrations was observed. At 1.0 μΜ siRNA and incubation at 37 °C, the observed RFU was 7.84 ± 0.54, whereas with pre -saturation of the Trf receptor, the RFU decreased to 2.85 + 0.54. The effect of the temperature on the extent of liposome-cell association was examined through the incubation of tumor cells with Trf- liposomes at 4 °C and 37 °C. As expected, incubation at 4 °C resulted in a significant reduction of the extent of liposome- cell association as compared to the incubation at 37 °C (at 1.0 μΜ siRNA, the RFU was reduced from 7.84 ± 0.54 to 3.72 ± 1.25). It was also observed that Trf receptor saturation performed at 4°C, resulted in the highest reduction observed for the extent of cellular association, with a RFU of 1.48 ± 0.15, indicating that binding of the liposomes to the cell surface is also affected by the saturation of TrfR. As illustrated in Figure 3, confocal microscopy studies were performed to assess the transferrin-receptor dependency of cell internalization of the Trf -associated nanocarriers . Upon incubation of the cells with liposomes, at 37 °C, cell internalization of Cy3-siR A was only detected for the cells incubated with Trf -liposomes (Figure 3A and F) . For BSA- (Figure 3B) or NT- liposomes (Figure 3G) no Cy3- siRNA inside the cells was observed. This same observation was registered upon saturation of the Trf receptor at 37 °C (Figure 3D) or upon treatment of the cells at 4 °C (Figure 3E) . Thus, the cell association and internalization is temperature- and TrfR- dependent.
Tables Table I. Imatinib and siRNA loading parameters of Trf-liposomes co -encapsulating imatinib and siRNA
imatiiiib: total lipid ratio imatinib (nmol/anol TL) siRNA. (nmol/μποΐ TU)
1/16 20.91 ± 7.0 3.19 ± 1.49
1/32 9.70 + 4.42 3.38 + 1.5
1/42 5.99 + 0.82 3.90 + 2.02
Table II. Effect of Trf-liposomes loading different
siRNA/imatinib molar ratios on imatinib IC50 and DRI,
imatinib-resistant and non-resistant leukemia cells
siRNA/ijj-itinib free free imatinib 0.15 0.35 0.63 imtinib + imtinib encaps
Cell line
imatinib/lipid ~ ~~ ' "siRNA
(1/16) (1/32) ' (1/42) eneaps
IRKS62 ICso ίμΜ) 3 .03 ± 0.91 ±
1.49 ± 0 .19 3 .49 ± 0 .32 n.a .
0 .95 0.09
DRI 1.16 2.34 3' 84
K562 IC¾0 (nM) 85.90 ± 138 .0 + 122 .0 ±
60.40 + 4.60 n.a . n.a.
6.78 34 .0 14.0
DRI 1.61 2.28
LSMA-84 ICjo (nM) 50.40 + 35.60 ± 54 . 10 ±
36 . 80 ± 2 .50 84 .0 ± 8 .0 70.0 ± 4 .3.0
4 .2 16.20
DRI 2 .42 .32 3 .43 2.85 References :
Abraham SA, McKenzie C, Masin D, Ng R, Harasym TO, Mayer LD, Bally MB. 2004. In vitro and in vivo characterization of doxorubicin and vincristine coencapsulated within liposomes through use of transition metal ion complexation and pH gradient loading. Clin Cancer Res 10 (2) : 728-38.
Allen TM. 2002. Ligand- targeted therapeutics in anticancer
therapy . Nat Rev Cancer 2 (10) : 750-63.
Allen TM, Sapra P, Moase E. 2002. Use of the post-insertion
method for the formation of ligand-coupled liposomes. Cell Moi Biol Lett 7(3) : 889-94.
Baker HM , Anderson BF, Baker EN. 2003. Dealing with iron: common structural principles in proteins that transport iron and heme. Proc Natl Acad Sci U S 100(7) : 3579-83.
Bantounas I, Phylactou LA, Uney JB. 2004. RNA interference and the use of small interfering RNA to study gene function in mammalian systems. J Mol Endocrinol 33 (3 ): 545-57.
Bhindi R, Fahmy RG, Lowe HC, Chesterman CN, Dass CR, Cairns MJ, Saravolac EG, Sun LQ, Khachigian LM. 2007. Brothers in arms: DNA enzymes, short interfering RNA, and the emerging wave of small -molecule nucleic acid-based gene- silencing strategies. Am J Pathol 171 (4) : 1079- 88.
Brennan P, Donev R, Hewamana S. 2008. Targeting transcription factors for therapeutic benefit, Mol Biosyst 4(9) :909-19.
Cardoso AL, Simoes S, de Almeida LP, Plesnila N, Pedroso de Lima MC, Wagner E, Culmsee C. 2008. Tf -lipoplexes for neuronal siRNA delivery: a promising system to mediate gene
silencing in the CNS . J Control Release 132 (2) : 113-23.
Chiu SJ, Liu S, Perrotti D, Marcueci G, Lee RJ. 2006. Efficient delivery of a Bel -2 -specific antisense
oligodeoxyribonucleotide (G3139) via transferrin receptor- targeted liposomes. J Control Release 112 (2) : 199-207.
Chiu YL, Rana TM. 2003. siRNA function in RNAi : a chemical
modification analysis. Rna 9 ( 9) : 1034-48. Chou TC. 2006. Theoretical basis, experimental design, and computerized simulation of synergism and antagonism in drug combination studies. Pharmacol Rev 58 (3) : 621-81.
Cullis PR, Hope MJ, Bally MB, Madden TD, Mayer LD, Fenske DB.
1997. Influence of pH gradients on the transbilayer
transport of drugs, lipids, peptides and metal ions into large unilamellar vesicles. Biochim Biophys Acta
1331 (2) : 187-211.
Daniels TR, Delgado T, Rodriguez JA, Helguera G, Penichet ML.
2006. The transferrin receptor part I: Biology and
targeting with cytotoxic antibodies for the treatment of cancer. Clin Immunol 121 (2) : 144-58.
Drummond DC, Noble CO, Hayes ME, Park JW, Kirpotin DB. 2008.
Pharmacokinetics and in vivo drug release rates in
liposomal nanocarrier development. J Pharm Sci 97(11) :4696-
740.
Dykxhoorn DM, Lieberman J. 2006. Running interference: prospects and obstacles to using small interfering RNAs as small molecule drugs. Annu Rev Biomed Eng 8:377-402.
Dykxhoorn DM, Palliser D, Lieberman J. 2006. The silent
treatment: siRNAs as small molecule drugs. Gene Ther
13 (6) : 541-52.
Galderisi U, Cascino A, Giordano A. 1999. Antisense
oligonucleotides as therapeutic agents. J Cell Physiol 181 (2) -251-7.
Goldoni M, Johansson C. 2007. A mathematical approach to study combined effects of toxicants in vitro: evaluation of the Bliss independence criterion and the Loewe additivity model. Toxicol In Vitro 21 (5) : 759-69.
Hoffbrand AV, Moss PAH, Pettit JE, editors. 2006a. The aetiology and genetics of haematological malignancies. 5 ed:
Blackwell Publishing. 129-136 p.
Hoff rand AV, Moss PAH, Pettit JE, editors. 2006b. Chronic
myeloid leukaemia. 5 ed: Blackwell publishing, 174-181 p. Holland JW, Hui C, Cullis PR, Madden TD . 1996. Poly (ethylene glycol) - -lipid conjugates regulate the calcium-induced fusion of liposomes composed of phosphatidylethanolamine and phosphatidylserine . Biochemistry 35 (8) : 2618-24.
Huang C, Li M, Chen C, Yao Q. 2008. Small interfering RNA
therapy in cancer: mechanism, potential targets, and clinical applications. Expert Opin Ther Targets 12(5) :637- 45.
Immordino ML, Dosio F, Cattel L. 2006. Stealth liposomes: review of the basic science, rationale, and clinical applications, existing and potential. Int J Nanomedicine 1 (3 ) : 297-315.
Ishida T, Iden DL, Allen TM. 1999. A combinatorial approach to producing sterically stabilized (Stealth) immunoliposomal drugs. FEBS Lett 460 (1) : 129-33.
Jia J, Zhu F, Ma X, Cao ZW, Li YX, Chen YZ . 2009. Mechanisms of drug combinations: interaction and network perspectives.
Nat Rev Drug Discov 8(2) : 111-28.
Kalota A, Shetzline SE, Gewirtz AM. 2004. Progress in the
development of nucleic acid therapeutics for cancer. Cancer Biol Ther 3(1) : 4-12.
Kumar LD, Clarke AR. 2007. Gene manipulation through the use of small interfering RNA (siRNA) : from in vitro to in vivo applications. Adv Drug Deliv Rev 59 (2 -3 ) : 87 - 100.
Lasic DD. 1998. Novel applications of liposomes. Trends
Biotechnol 16 (7) : 307-21.
Lee SH, Sinko PJ. 2006. siRNA- -get ing the message out. Eur J Pharm Sci 27 (5 ) : 401- 10.
Leonetti C, Biroccio A, Benassi B, Stringaro A, Stoppacciaro A, Semple SC, Zupi G. 2001. Encapsulation of c -myc antisense oligodeoxynucleotides in lipid particles improves
antitumoral efficacy in vivo in a human melanoma line.
Cancer Gene Ther 8(6) :459-68.
Li C, Cui J, Li Y , Wang C, Li Y, Zhang L, Zhang L, Guo W, Wang J, Zhang H and others. 2008. Copper ion-mediated liposomal encapsulation of mitoxantrone : the role of anions in drug loading, retention and release. Eur J Pharm Sci 34(4-
5) : 333-44. Li H, Qian ZM . 2002. Transferrin/transferrin receptor-mediated drug delivery. Med Res Rev 22 (3) : 225-50.
Mahato RI, Cheng K, Guntaka RV. 2005. Modulation of gene
expression by antisense and antigene oligodeoxynucleot ides and small interfering R A. Expert Opin Drug Deliv 2(1) : 3 -
28.
Martin SE, Caplen NJ . 2007. Applications of RNA interference in mammalian systems. Annu Rev Genomics Hum Genet 8:81-108.
Maurer N, Wong KF, Stark H, Louie L , Mcintosh D , Wong T,
Scherrer P, Semple SC, Cullis PR. 2001. Spontaneous entrapment of polynucleotides upon electrostatic
interaction with ethanol -destabilized cationic liposomes. Biophys J 80 (5) :2310-26.
Mayer LD, Bally MB, Cullis PR. 1986. Uptake of adriamycin into large unilamellar vesicles in response to a pH gradient . Biochim Biophys Acta 857(1) : 123-6.
Mayer LD, Harasym TO, Tardi PG, Harasym NL, Shew CR, Johnstone SA, Ramsay EC, Bally MB, Janoff AS. 2006. Ratiometric dosing of anticancer drug combinations: controlling drug ratios after systemic administration regulates therapeutic activity in tumor-bearing mice. Mol Cancer Ther 5(7) :1854- 63.
Mendonca LS, Firmino F, Moreira JN, Pedroso de Lima MC, Simoes S. 2010. Transferrin receptor- targeted liposomes
encapsulating anti-BCR-ABL siRNA or asODN for chronic myeloid leukemia treatment. Bioconjug Chem 21(1) : 157-68.
Merlin JL . 1994. Concepts of Synergism and Antagonism.
ANTICANCER RESEARCH 14:2315-2320.
Morrissey DV, Lockridge JA, Shaw L, Blanchard K, Jensen K, Breen W, Hartsough K, Machemer L, Radka S, Jadhav V and others. 2005, Potent and persistent in vivo anti-HBV activity of chemically modified siRNAs. Nat Biotechnol 23 "( 8 ) : 1002 -7.
Mui B, Raney SG, Semple SC, Hope MJ. 2001. Immune stimulation by a CpG-containing oligodeoxynucleotide is enhanced when encapsulated and delivered in lipid particles. J Pharmacol Exp Ther 298 (3 ) : 1185 - 92. Nobs L, Buchegger F, Gurny R, Allemanri E . 2004. Current methods for attaching targeting ligands to liposomes and
nandparticles . J Pharm Sci 93 (8) : 1980-92 ,
O'Brien J, Wilson I, Orton T, Pognan F. 2000. Investigation of the Alamar Blue (resazurin) fluorescent dye for the assessment of mammalian cell cytotoxicity. Eur J Biochem
267 (17) :5421-6.
Ottensmeier G. 2001. The classification of lymphomas and
leukemias . Chem Biol Interact 135-136:653-64.
Owens DE, 3rd, Peppas NA. 2006. Opsonization, biodistribut ion, and pharmacokinetics of polymeric nanoparticles. Int J
Pharm 307 (1) : 93-102.
Pasternak G, Hochhaus A, Schultheis B, Hehlmann R. 1998. Chronic myelogenous leukemia: molecular and cellular aspects. J
Cancer Res Clin Oncol 124 ( 12 ) : 643 -60.
Ponka P, Lok CN . 1999. The transferrin receptor: role in health and disease. Int J Biochem Cell Biol 31(10) : 1111-37.
Qian ZM, Li H, Sun H, Ho K. 2002. Targeted drug delivery via the transferrin receptor-mediated endocytosis pathway.
Pharmacol Rev 54 ( ) : 561 -87.
Rayburn ER, Zhang R. 2008. Antisense, RNAi , and gene silencing strategies for therapy: mission possible or impossible?
Drug Discov Today 13(11-12) ; 513-21.
Romberg B, Hennink WE, Storm G. 2008. Sheddable coatings for long-circulating nanoparticles. Pharm Res 25(1) : 55-71.
Ryan SM, Mantovarii G, Wang X, Haddleton DM, Brayden DJ. 2008.
Advances in PEGylation of important biotech molecules:
delivery aspects. Expert Opin Drug Deliv 5(4) :371-83.
Sapra P, Allen TM. 2003. Ligand- targeted liposomal anticancer drugs. Prog Lipid Res 42 ( 5 ) : 439 - 62.
Saxon DN, Mayer LD, Bally MB. 1999a. Liposomal anticancer drugs as agents to be used in combination with other anticancer agents: studies on a liposomal formulation with two
encapsulated drugs. J Liposome Res 9 (4) : 507-522.
Saxon DN, Mayer LD, Bally MB. 1999b. Liposomal anticancer drugs as agents to be used in combination with other anticancer agents: studies on a liposomal formulation with two
encapsulated drugs. Journal of liposome research 9(4) :507- 522.
Semple SC, Klimuk SK, Harasym TO, Dos Santos N, Ansel1 SM, Wong KF, Maurer N, Stark H, Gullis PR, Hope J and others. 2001. Efficient encapsulation of antisense oligonucleotides in lipid vesicles using ionizable aminolipids: formation of novel small multilamellar vesicle structures . Biochim
Biophys Acta 1510 (1-2) .-152-66.
Shrivastava N, Srivastava A. 2008. RNA interference: an emerging generation of biologicals. Biotechnol J 3(3) :339-53.
Stahel RA, Zangemeister-Wittke U. 2003. Antisense
oligonucleotides for cancer therapy-an overview. Lung
Cancer 41 Suppl l:S81-8.
Takasaki J, Raney SG, Chikh G, Sekirov L, Brodsky I, Tarn Y,
Ansell SM. 2006. Methods for the preparation of protein- Oligonucleotide-lipid constructs. Bioconjug Chem 17(2) :451- 8.
Tamm I. 2006. Antisense therapy in malignant diseases: status quo and quo vadis? Clin Sci (Lond) 110(4) :427-42,
Tardi PG, Gallagher RC, Johnstone S, Harasym N, Webb M, Bally MB, Mayer LD . 2007. Coeneapsulation of irinotecan and floxuridine into low cholesterol -containing liposomes that coordinate drug release in vivo. Biochim Biophys Acta
1768 (3) :678-87.
Torch.il in V. 2009. Multifunctional and stimuli-sensitive
pharmaceutical nanocarriers. Eur J Pharm Biopharm
71 (3) :431-4 .
Torchilin VP. 2006. Multifunctional nanocarriers. Adv Drug Deliv
Rev 58 (14) : 1532-55.
Torchilin VP, 2007. Targeted pharmaceutical nanocarriers for
cancer therapy and imaging. Aaps J 9(2) : E128 -47.
lister PS, Allen TM, Daniel BE, Mendez CJ, Newman MS, Zhu GZ .
1996. Insertion of poly (ethylene . glycol ) derivatized phospholipid into pre -formed liposomes results in prolonged in vivo circulation time. FEES Lett 386 (2 -3 ) : 243 - 6.
Wang J, Goh B, Lu W, Zhang Q, Chang A, Liu ΧΫ, Tan TM, Lee H.
2005. In vitro cytotoxicity of Stealth liposomes co- encapsulating doxorubicin and verapamil on doxorubicin- resistant tumor cells. Biol Pharm Bull 28(5) : 822-8.
Waterhouse DN, bos Santos N, Mayer LD, Bally MB. 2001. Drug-drug interactions arising from the use of liposomal vincris ne in combination with other anticancer drugs. Pharm Res
18 (9) : 1331-5.
Wheeler JJ, Palmer L, Ossanlou M, MacLachlan I, Graham RW, Zhang YP, Hope MJ, Scherrer P, Cull is PR. 1999. Stabilized plasmid-lipid particles: construction and characterization. Gene Ther 6 (2) :271-81.
Zimmermann GR, Lehar J, Keith GT. 2007. Mul i- target
therapeutics: when the whole is greater than the sum of the parts. Drug Discov Today 12 (1-2) : 34-42.
Zoli W, Ricotti L, Tesei A, Barzanti F , Amadori D. 2001. In
vitro preclinical models for a rational design of
chemotherapy combinations in human umors. Crit Rev Oncol Hematol 37(1) : 69-82.

Claims

1. A multi-targeting system comprising a nanocarrier in which are included one or more nucleic acids and one or more non-nucleic acid based drugs.
2. The multi-targeting system according to claim wherein the inclusion are performed by simultaneous
encapsulation, adsorption or complexation of said one or more nucleic acids and one or more non-nucleic acid based drugs.
3. The multi-targeting system according to claim 1 or 2, wherein the multi-targeting system further comprises one or more ligands coupled to the nanocarrier surface.
4. The multi-targeting system according to any one of claims 1 to 3, wherein the nucleic acids are selected from plasmid, antisense oligonucleotides (asODN) , siRNA, miRNA, shRN ) aiRNA, DNA enzymes (DNAzymes) , Ribozymes, DNA decoys, A amers and mixtures thereof .
5. The multi -targeting system according to claim 4, wherein the nucleic acid comprises from about 15 to about 60 nucleotides.
6. The multi- targeting system according to claim 5 wherein the nucleic acid comprises at least one modified nucleotide ,
7. The multi -targeting system according to any one of claims 1 to 6 , wherein the non-nucleic acid based drugs are selected from chemotherapy drugs, hormonal therapeutic agents, immunotherapeutic agents, anti -viral drugs, anti-inflammatory compounds, antidepressants, stimulants, analgesics, antibiotic antipyretics, vasodilators, anti-angiogenics , cytovascular agents, signal transduction inhibitors, tyrosine kinase
inhibitors, ant i-arrhythmic agents, hormones, vasoconstrictors, and steroids .
8. The multi- targeting system according to claim 7, wherein the chemotherapy drugs include platinum-based drugs (e.g. cisplatin, carboplatin, etc.); alkylating agents (e.g., cyclophosphamide, chlorambucil, busulfan, melphalan, lomustine, carmustine, estramustine , treosulfan, thiotepa, mitobronitol , etc.); anti -metabolites (e.g., 5 - fluofouracil , methotrexate, capecitabine , cytarabine, fludarabine, gemcitabine, cladribine, raltitrexed, mercaptopurine , etc.); plant alkaloids (e.g., vincristine, vinblastine, vindesine, paclitaxel, docetaxel, etc), topoisomerase inhibitors (e.g., irinotecan, topotecan, etoposide, etc.); cytotoxic antibiotics (e.g., doxorubicin, adriamycin, daunorubicin, epirubicin, actinomycin, bleomycin, mitomycin, mitoxantrone , aclarubicin, idarubicin, dactinomycin, etc.), taxanes (e.g., docetaxel, paclitaxel); tyrosine kinase inhibitors (e.g.; gefitinib, sunitinib, erlotinib, lapatinib, canertinib, semaxinib, vatalanib, sorafenib, imatinib,
dasatinib, leflunomide , vandetanib, salts thereof, stereoisomers thereof, analogs thereof, derivatives thereof, and mixtures thereof; the anti- inflamatory agents include ibuprofen,
aceclofenac, acemetacin, acetilsalicilic acid, azapropazone , celecoxib, diclofenac sodium, diflunisal, cetodolac, fenbufen, fenoprofen, flubiprofen, indomethacin, acetaminocin, piroxicam, rofecoxib, sulindac, tenoxicam; the antiangiogenic agents or angiolytic agents include angiostatin (plasminogen fragment) , antiangiogenic antithrombin III, vasculostatin, vasostatin and mixtures thereof .
9. The multi- targeting system according to any one of claims 3 to 8 comprising one or more ligands linked to the rianocarrier , wherein each one of the ligands specifically bind to an overexpressed or to a specifically expressed molecule or receptor or protein of the targeted cell population.
10. The mul i. - targeting system according to claim 9, wherein the ligand comprise peptides, polypeptides, antibodies, polyclonal antibodies, monoclonal antibodies, antibody
fragments, humanized antibodies, recombinant antibodies,
recombinant human antibodies, nanobodies, aptamers, proteins and cell surface ligands.
11. The multi -targeting system of claim 10, wherein the ligands are linked to the surface of the nanocarrier in a way that it is able to interact with a specific molecule, protein, glycoprotein and/or cell surface receptor that is overexpressed or specifically expressed on the cell surface of a specific cellular target, wherein an appropriate spacer can be positioned between the nanocarrier and the ligand to avoid hindrance on the interaction between the ligand and its target.
12. The multi- targeting system according to any one of claims 1 to 11, wherein the nanocarrier presents more than one homing ligand that selectively homes the delivery agent to specific molecules on the target cells in order to allow
multivalent cellular targeting.
13. The multi-targeting system according to any one of claims 2 to 12, wherein the molecular ratios of the co- encapsuiated/adsorbed/complexed molecules remains unaltered during storage and upon in vivo administration.
14. The multi-targeting system according to any one of claims 1 to 13, wherein the nanocarrier is composed by lipids and/or polymers .
15. The multi-targeting system according to claim 14, wherein the nanocarrier is composed by stabilized nucleic acid lipid particles (SNALP) .
16. The multi-targeting system according to claim 15, wherein SNALP liposomes comprise one or more cationic lipids, one or more neutral lipids and one or more conjugated lipid that inhibits aggregation of particles and provide long circulation times to the liposomes.
17. The multi -targeting system according to claim 16, wherein the cationic lipids may comprise from 10 to 60 mol%.
18. The multi-targeting system according to claim 16, wherein the neutral lipid, may comprise from 10 to 70 mol%.
19. The multi-targeting system according to claim 16, wherein the conjugated lipid that inhibits aggregation and provides long circulation times may comprise from 1 to 10 mol%.
20. The multi-targeting system according to claim 16 or 17, wherein the cationic lipids are 1 , 2 -dioleoyl -3 - dimethylammonium-propane (DODAP) , 1 , 2 -dilinoleyloxy-3 - (2 -N, - dimethylamino) ethoxypropane (DLin-EG-DMA) , N, N-dioleyl -N, N- dimethylammonium chloride (DODAC) , 1 , 2-dioleyloxy-N, N-dimethyl - 3 -arninopropane (DODMA) , 1 , 2 -distearyloxy-N, N-dimethyl -3 - aminopropane (DSDMA) , N- ( 1- (2 , 3 -dioleyloxy) propyl ) -N, , - rimethylammonium chloride (DOTMA) , N-(l-(2,3- dioleoyloxy) propyl ) -N, , -trimethylammonium chloride (DOTAP) , 3- (N- (N' , 1 -dimethylaminoethane) -carbamoyl ) cholesterol (DC-Choi) , N- (1, 2-dimyristyloxyprop-3-yl) -N, -dimethyl -N- hydroxyethylammonium bromide (DMRIE) , 2 , 3 -dioleyloxy-N- [2 (spennine-carboxarnido) ethyl] N, N-dime hyl 1 - propanaminiumtrifluoroacetate (DOSPA) , dioctadecylamidoglycyl spermine (DOGS), N, N-dimethyl -3 , 4 -dioleyloxybenzylamme (DMOBA) , 1 , 2 -N, ' -dioleylcarbamyl -3 -dimethylammopropane (DOcarbDAP) , 1,2- Ν,Ν' -dilmoleylcarbamyl-S-dimethylaminopropahe (DLi c bDAP) , 1, 2-dilinoleyloxy-N,N-dimethylaminopropane (DLinDMA) , 1,2- dilinolenyloxy-N, N-dimethylaminopropane (DLenDMA) , 2,2- dilinoleyl -4 - (2-dimethylaminoethyl) - [1,3] -dioxolane (DLin-K-C2- DMA), 2 , 2 -dilinoleyl -4 - (3-dimethylarninopropyl) - [1, 3] - dioxolane
(DLin-K-C3 -DMA) , 2 , 2 -dilinoleyl -4 - (4 -dimethylaminobutyl ) -[1,3]- dioxolane (DLin-K-C4 -DMA) , 2 , 2 -dilinoleyl -4 -dimethylaminomethyl -
[1 , 3 ] -dioxolane (DLin-K-DMA) , 1 , 2 -dilinoleylcarbamoyioxy-3 - dimethylaminopropane (DLin-C-DAP) , 1,2· dilinoleyoxy- (dimethylamino) acetoxypropane (DLin-DAC) , 1 , 2 -dilinoleyoxy-3 - morpholinopropane (DLin-MA) , 1 , 2 -dilinoleoyl -3 - dimethylaminopropane (DLinDAP) , 1 , 2 -dilinoleylthio- 3 - dimethylaminopropane (DLin-S-DMA) , 1 - linoleoyl -2 - linoleyloxy-3 - dimethylaminopropane (DLin-2 -DMAP) , 1 , 2 -dil inoleyloxy-3 - trimethylaminopropane chloride salt (DLin-TMA. CI ) , 1,2L
dilinoleoyl -3 -trimethylaminopropane chloride salt (DLin-TAP . Cl ) , 1 , 2 -dilinoleyloxy-3 - (N-methylpiperazino) propane (DLin-MPZ) , 3-
(N, N-dilinoleylamino) -1, 2 -propanediol (DLinAP) , or mixtures thereof .
21. The multi-targeting system according to claim 16 or 18, wherein the neutral lipid components may be cholesterol or a derivative thereof; phospholipids; or a mixture of
phospholipids and cholesterol or a derivative thereof. Examples of cholesterol derivatives include cholestanol, cholestanone , cholestenone , coprostanol, cholesteryl -2 ' -hydroxyethyl ether, cholesteryl -4 ' - hydroxybutyl ether, and mixtures thereof.
Examples of neutral lipids include but are not limited to dipalmitoylphosphatidylcholine (DPPG) ,
distearoylphosphatidylcholine (DSPC) ,
dioleoylphosphatidylethanolamine (DOPE) , palmitoyloleoyl - phosphatidylcholine (POPC) , palmitoyloleoyl - phosphatidylethanolamine (POPE) , palmitoyloleyol - phosphatidylglycerol (POPG) , dipalmitoyl- phosphatidylethanolamine (DPPE) , dimyristoyl- phosphatidyiethanolamine (D PE) , distearoyl- phosphatidylethanolamine (DSPE) , monomethyl- phosphatidylethanolamine, dimethyl -phosphatidyiethanolamine , dielaidoyl- phosphatidyiethanolamine (DEPE) , stearoyloleoyl - phosphatidyiethanolamine (SOPE) , egg phosphatidylcholine (EPC) , and mixtures thereof .
22. The multi-targeting system according to claim 16 or 19, wherein conjugated lipids are prepared using
biocompatible, soluble and hydrophilic polymers with a highly flexible main chain selected from poly(acryl amide), poly(vinyl pyrrolidone) , poly (acryloyl morpholine) , poly (2 -ethyl -2 - oxazoline), poly (2 -methyl -oxazoline) , phosphatidyl
polyglycerols , polyvinyl alcohols, polysaccharides, PEG and PEG containing copolymers (as poloxamers and poloxamines) , or mixtures therefore.
23. The multi -targeting system according to claim 22 wherein PEG polymer is used in the formulation as PEG-lipids such as PEG-diacylglycerol (DAG) , PEG dialkyloxypropyl (DAA) , PEG-ceramide (Cer) , PEG-phosphatidylethanolamine , PEG- 1 -methyl - 4-(cis-9- dioleyl) methyl -pyridinium chloride ( PEG-SAINT) or mixtures thereof. Examples of PEG-Cer conjugate include but are not limited to PEG-dilauryloxypropyl (C 12) , a PEG- dimyristyloxypropyl (C 14), a PEG-dipalmityloxypropyl (C 16), a PEG-distearyloxypropyl (C 18), or mixtures thereof.
23. The multi- targeting system of claim 22, wherein the PEG moiety of the PEG-lipid conjugates described herein may compose an average molecular weight ranging from about 550 daltons to about 10 000 daltons.
24. The multi-targeting system according to claim 23 wherein the PEG moiety of the PEG-lipid conjugates described herein may compose an average molecular weight ranging from about 550 daltons to about 10 000 daltons.
25. A method of selectively targeting therapeutic or diagnostic molecules to specific cells, combining cellular and molecular targeting, using the the mult i- arge ing system of any one of claims 1 to 2.4.
26. A pharmaceutically acceptable Composition
comprising the multi- targeting system of any one of claims 1 tc 25 and a pharmaceuticall acceptable carrier for in vivo
administration.
27. The multi-targeting system of any one of claims 1 to 2 or a pharmaceutica11y acceptable composition of claim 26, for the treatment, diagnosis and prevention of human cancer and other : diseases , including inflammation, neurodegenerative diseases, infect ious diseases , and cardiovascular disorders .
PCT/PT2011/000005 2010-03-03 2011-03-03 Multi -targeting system comprising a nanocarrier, nucleic acid(s) and non-nucleic acid based drug(s) WO2011108955A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
PT104988 2010-03-03
PT10498810 2010-03-03

Publications (1)

Publication Number Publication Date
WO2011108955A1 true WO2011108955A1 (en) 2011-09-09

Family

ID=44080275

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/PT2011/000005 WO2011108955A1 (en) 2010-03-03 2011-03-03 Multi -targeting system comprising a nanocarrier, nucleic acid(s) and non-nucleic acid based drug(s)

Country Status (1)

Country Link
WO (1) WO2011108955A1 (en)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014003830A1 (en) * 2012-06-25 2014-01-03 Emory University Particle-nucleic acid conjugates and therapeutic uses related thereto
WO2014203189A1 (en) * 2013-06-18 2014-12-24 Rosetta Genomics Ltd. Nanocarrier system for micrornas and uses thereof
EP3087974A1 (en) * 2015-04-29 2016-11-02 Rodos BioTarget GmbH Targeted nanocarriers for targeted drug delivery of gene therapeutics
WO2017205566A1 (en) * 2016-05-27 2017-11-30 Preceres Inc. Oxidized aminolipidoids and uses thereof
EP3319976A4 (en) * 2015-07-09 2019-03-20 Insmed Incorporated Compositions and methods for treating lung diseases and lung injury
US10398719B2 (en) 2014-05-15 2019-09-03 Insmed Incorporated Methods for treating pulmonary non-tuberculous mycobacterial infections
US10471149B2 (en) 2012-11-29 2019-11-12 Insmed Incorporated Stabilized vancomycin formulations
CN114940756A (en) * 2022-06-02 2022-08-26 北京清科胜因生物科技有限公司 Poly (2-oxazoline) lipid and lipid nanoparticle and application
US11491247B2 (en) 2017-05-02 2022-11-08 Cornell University Methods and reagents for tumor targeting with greater efficacy and less toxicity
US11571386B2 (en) 2018-03-30 2023-02-07 Insmed Incorporated Methods for continuous manufacture of liposomal drug products
CN117582512A (en) * 2022-08-09 2024-02-23 湖南健瑞医药科技有限公司 Metal-polyphenol composite particles, and preparation method and application thereof
US11964052B2 (en) 2021-05-24 2024-04-23 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010108934A1 (en) * 2009-03-25 2010-09-30 Novartis Ag Pharmaceutical composition containing a drug and sirna

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010108934A1 (en) * 2009-03-25 2010-09-30 Novartis Ag Pharmaceutical composition containing a drug and sirna

Non-Patent Citations (75)

* Cited by examiner, † Cited by third party
Title
"Chronic myeloid leukaemia", 2006, BLACKWELL PUBLISHING, pages: 174 - 181
"The aetiology and genetics of haematological malignancies", BLACKWELL PUBLISHING, pages: 129 - 136
ABRAHAM SA, MCKENZIE C, MASIN D, NG R, HARASYM TO, MAYER LD, BALLY MB: "In vitro and in vivo characterization of doxorubicin and vincristine coencapsulated within liposomes through use of transition metal ion complexation and pH gradient loading", CLIN CANCER RES, vol. 10, no. 2, 2004, pages 728 - 38, XP002373873, DOI: doi:10.1158/1078-0432.CCR-1131-03
ALEX M. CHEN ET AL: "Co-delivery of Doxorubicin and Bcl-2 siRNA by Mesoporous Silica Nanoparticles Enhances the Efficacy of Chemotherapy in Multidrug-Resistant Cancer Cells", SMALL, vol. 5, no. 23, 4 December 2009 (2009-12-04), pages 2673 - 2677, XP055000464, ISSN: 1613-6810, DOI: 10.1002/smll.200900621 *
ALLEN TM, SAPRA P, MOASE E: "Use of the post-insertion method for the formation of ligand-coupled liposomes", CELL MOL BIOL LETT, vol. 7, no. 3, 2002, pages 889 - 94
ALLEN TM: "Ligand-targeted therapeutics in anticancer therapy", NAT REV CANCER, vol. 2, no. 10, 2002, pages 750 - 63
BAKER HM, ANDERSON BF, BAKER EN.: "Dealing with iron: common structural principles in proteins that transport iron and heme", PROC NATL ACAD SCI U S A, vol. 100, no. 7, 2003, pages 3579 - 83
BANTOUNAS I, PHYLACTOU LA, UNEY JB.: "RNA interference and the use of small interfering RNA to study gene function in mammalian systems", J MOL ENDOCRINOL, vol. 33, no. 3, 2004, pages 545 - 57
BHINDI R, FAHMY RG, LOWE HC, CHESTERMAN CN, DASS CR, CAIRNS MJ, SARAVOLAC EG, SUN LQ, KHACHIGIAN LM.: "Brothers in arms: DNA enzymes, short interfering RNA, and the emerging wave of small-molecule nucleic acid-based gene-silencing strategies", AM J PATHOL, vol. 171, no. 4, 2007, pages 1079 - 88, XP003026452, DOI: doi:10.2353/AJPATH.2007.070120
BRENNAN P, DONEV R, HEWAMANA S.: "Targeting transcription factors for therapeutic benefit", MOL BIOSYST, vol. 4, no. 9, 2008, pages 909 - 19
CARDOSO AL, SIMOES S, DE ALMEIDA LP, PLESNILA N, PEDROSO DE LIMA MC, WAGNER E, CULMSEE C.: "Tf-lipoplexes for neuronal siRNA delivery: a promising system to mediate gene silencing in the CNS", J CONTROL RELEASE, vol. 132, no. 2, 2008, pages 113 - 23, XP025684667, DOI: doi:10.1016/j.jconrel.2008.08.014
CHIU SJ, LIU S, PERROTTI D, MARCUCCI G, LEE RJ.: "Efficient delivery of a Bcl-2-specific antisense oligodeoxyribonucleotide (G3139) via transferrin receptor-targeted liposomes", J CONTROL RELEASE, vol. 112, no. 2, 2006, pages 199 - 207, XP024957500, DOI: doi:10.1016/j.jconrel.2006.02.011
CHIU YL, RANA TM: "siRNA function in RNAi: a chemical modification analysis", RNA, vol. 9, no. 9, 2003, pages 1034 - 48, XP008158876, DOI: doi:10.1261/rna.5103703
CHOU TC: "Theoretical basis, experimental design, and computerized simulation of synergism and antagonism in drug combination studies", PHARMACOL REV, vol. 58, no. 3, 2006, pages 621 - 81, XP055151376, DOI: doi:10.1124/pr.58.3.10
CULLIS PR, HOPE MJ, BALLY MB, MADDEN TD, MAYER LD, FENSKE DB.: "Influence of pH gradients on the transbilayer transport of drugs, lipids, peptides and metal ions into large unilamellar vesicles", BIOCHIM BIOPHYS ACTA, vol. 1331, no. 2, 1997, pages 187 - 211, XP004281716, DOI: doi:10.1016/S0304-4157(97)00006-3
DANIELS TR, DELGADO T, RODRIGUEZ JA, HELGUERA G, PENICHET ML.: "The transferrin receptor part I: Biology and targeting with cytotoxic antibodies for the treatment of cancer", CLIN IMMUNOL, vol. 121, no. 2, 2006, pages 144 - 58, XP024941315, DOI: doi:10.1016/j.clim.2006.06.010
DRUMMOND DC, NOBLE CO, HAYES ME, PARK JW, KIRPOTIN DB: "Pharmacokinetics and in vivo drug release rates in liposomal nanocarrier development", J PHARM SCI, vol. 97, no. 11, 2008, pages 4696 - 740
DYKXHOORN DM, LIEBERMAN J.: "Running interference: prospects and obstacles to using small interfering RNAs as small molecule drugs", ANNU REV BIOMED ENG, vol. 8, 2006, pages 377 - 402, XP009101888, DOI: doi:10.1146/annurev.bioeng.8.061505.095848
DYKXHOORN DM, PALLISER D, LIEBERMAN J.: "The silent treatment: siRNAs as small molecule drugs", GENE THER, vol. 13, no. 6, 2006, pages 541 - 52, XP002562042, DOI: doi:10.1038/sj.gt.3302703
GALDERISI U, CASCINO A, GIORDANO A.: "Antisense oligonucleotides as therapeutic agents", J CELL PHYSIOL, vol. 181, no. 2, 1999, pages 251 - 7, XP009063561, DOI: doi:10.1002/(SICI)1097-4652(199911)181:2<251::AID-JCP7>3.0.CO;2-D
GOLDONI M, JOHANSSON C.: "A mathematical approach to study combined effects of toxicants in vitro: evaluation of the Bliss independence criterion and the Loewe additivity model", TOXICOL IN VITRO, vol. 21, no. 5, 2007, pages 759 - 69, XP022042538
HOLLAND JW, HUI C, CULLIS PR, MADDEN TD: "Poly(ethylene glycol)--lipid conjugates regulate the calcium-induced fusion of liposomes composed of phosphatidylethanolamine and phosphatidylserine", BIOCHEMISTRY, vol. 35, no. 8, 1996, pages 2618 - 24, XP002084425, DOI: doi:10.1021/bi952000v
HUANG C, LI M, CHEN C, YAO Q.: "Small interfering RNA therapy in cancer: mechanism, potential targets, and clinical applications", EXPERT OPIN THER TARGETS, vol. 12, no. 5, 2008, pages 637 - 45
IMMORDINO ML, DOSIO F, CATTEL L.: "Stealth liposomes: review of the basic science, rationale, and clinical applications, existing and potential", INT J NANOMEDICINE, vol. 1, no. 3, 2006, pages 297 - 315, XP009145524
ISHIDA T, IDEN DL, ALLEN TM: "A combinatorial approach to producing sterically stabilized (Stealth) immunoliposomal drugs", FEBS LETT, vol. 460, no. 1, 1999, pages 129 - 33, XP004342732, DOI: doi:10.1016/S0014-5793(99)01320-4
JIA J, ZHU F, MA X, CAO ZW, LI YX, CHEN YZ.: "Mechanisms of drug combinations: interaction and network perspectives", NAT REV DRUG DISCOV, vol. 8, no. 2, 2009, pages 111 - 28
KALOTA A, SHETZLINE SE, GEWIRTZ AM.: "Progress in the development of nucleic acid therapeutics for cancer", CANCER BIOL THER, vol. 3, no. 1, 2004, pages 4 - 12
KUMAR LD, CLARKE AR: "Gene manipulation through the use of small interfering RNA (siRNA): from in vitro to in vivo applications", ADV DRUG DELIV REV, vol. 59, no. 2-3, 2007, pages 87 - 100, XP022087316, DOI: doi:10.1016/j.addr.2007.03.009
LASIC DD: "Novel applications of liposomes", TRENDS BIOTECHNOL, vol. 16, no. 7, 1998, pages 307 - 21, XP004145644, DOI: doi:10.1016/S0167-7799(98)01220-7
LEE SH, SINKO PJ: "siRNA--getting the message out", EUR J PHARM SCI, vol. 27, no. 5, 2006, pages 401 - 10, XP025137267, DOI: doi:10.1016/j.ejps.2005.12.002
LEONETTI C, BIROCCIO A, BENASSI B, STRINGARO A, STOPPACCIARO A, SEMPLE SC, ZUPI G.: "Encapsulation of c-myc antisense oligodeoxynucleotides in lipid particles improves antitumoral efficacy in vivo in a human melanoma line", CANCER GENE THER, vol. 8, no. 6, 2001, pages 459 - 68, XP002260435, DOI: doi:10.1038/sj.cgt.7700326
LI C, CUI J, LI Y, WANG C, LI Y, ZHANG L, ZHANG L, GUO W, WANG J, ZHANG H: "Copper ion-mediated liposomal encapsulation of mitoxantrone: the role of anions in drug loading, retention and release", EUR J PHARM SCI, vol. 34, no. 4-5, 2008, pages 333 - 44, XP022939890, DOI: doi:10.1016/j.ejps.2008.05.006
LI H, QIAN ZM.: "Transferrin/transferrin receptor-mediated drug delivery", MED RES REV, vol. 22, no. 3, 2002, pages 225 - 50
LILIANA S. MENDONÇA ET AL: "Co-encapsulation of anti-BCR-ABL siRNA and imatinib mesylate in transferrin receptor-targeted sterically stabilized liposomes for chronic myeloid leukemia treatment", BIOTECHNOLOGY AND BIOENGINEERING, vol. 107, no. 5, 1 December 2010 (2010-12-01), pages 884 - 893, XP055000380, ISSN: 0006-3592, DOI: 10.1002/bit.22858 *
LILIANA S. MENDONÇA ET AL: "Transferrin Receptor-Targeted Liposomes Encapsulating anti- BCR-ABL siRNA or asODN for Chronic Myeloid Leukemia Treatment", BIOCONJUGATE CHEMISTRY, vol. 21, no. 1, 20 January 2010 (2010-01-20), pages 157 - 168, XP055000390, ISSN: 1043-1802, DOI: 10.1021/bc9004365 *
MAHATO RI, CHENG K, GUNTAKA RV.: "Modulation of gene expression by antisense and antigene oligodeoxynucleotides and small interfering RNA", EXPERT OPIN DRUG DELIV, vol. 2, no. 1, 2005, pages 3 - 28, XP008062634, DOI: doi:10.1517/17425247.2.1.3
MARTIN SE, CAPLEN NJ: "Applications of RNA interference in mammalian systems", ANNU REV GENOMICS HUM GENET, vol. 8, 2007, pages 81 - 108, XP002467153, DOI: doi:10.1146/annurev.genom.8.080706.092424
MAURER N, WONG KF, STARK H, LOUIE L, MCINTOSH D, WONG T, SCHERRER P, SEMPLE SC, CULLIS PR: "Spontaneous entrapment of polynucleotides upon electrostatic interaction with ethanol-destabilized cationic liposomes", BIOPHYS J, vol. 80, no. 5, 2001, pages 2310 - 26, XP002589197
MAYER LD, BALLY MB, CULLIS PR: "Uptake of adriamycin into large unilamellar vesicles in response to a pH gradient", BIOCHIM BIOPHYS ACTA, vol. 857, no. 1, 1986, pages 123 - 6, XP023793244, DOI: doi:10.1016/0005-2736(86)90105-7
MAYER LD, HARASYM TO, TARDI PG, HARASYM NL, SHEW CR, JOHNSTONE SA, RAMSAY EC, BALLY MB, JANOFF AS: "Ratiometric dosing of anticancer drug combinations: controlling drug ratios after systemic administration regulates therapeutic activity in tumor-bearing mice", MOL CANCER THER, vol. 5, no. 7, 2006, pages 1854 - 63, XP002427287, DOI: doi:10.1158/1535-7163.MCT-06-0118
MENDONCA LS, FIRMINO F, MOREIRA JN, PEDROSO DE LIMA MC, SIMOES S.: "Transferrin receptor-targeted liposomes encapsulating anti-BCR-ABL siRNA or asODN for chronic myeloid leukemia treatment", BIOCONJUG CHEM, vol. 21, no. 1, 2010, pages 157 - 68, XP055000390, DOI: doi:10.1021/bc9004365
MERLIN JL.: "Concepts of Synergism and Antagonism", ANTICANCER RESEARCH, vol. 14, 1994, pages 2315 - 2320
MORRISSEY DV, LOCKRIDGE JA, SHAW L, BLANCHARD K, JENSEN K, BREEN W, HARTSOUGH K, MACHEMER L, RADKA S, JADHAV V: "Potent and persistent in vivo anti-HBV activity of chemically modified siRNAs", NAT BIOTECHNOL, vol. 23, no. 8, 2005, pages 1002 - 7, XP008157520, DOI: doi:10.1038/nbt1122
MUI B, RANEY SG, SEMPLE SC, HOPE MJ.: "Immune stimulation by a CpG-containing oligodeoxynucleotide is enhanced when encapsulated and delivered in lipid particles", J PHARMACOL EXP THER, vol. 298, no. 3, 2001, pages 1185 - 92
NOBS L, BUCHEGGER F, GURNY R, ALLEMANN E: "Current methods for attaching targeting ligands to liposomes and nanoparticles", J PHARM SCI, vol. 93, no. 8, 2004, pages 1980 - 92, XP055055813, DOI: doi:10.1002/jps.20098
NUO CAO ET AL: "The synergistic effect of hierarchical assemblies of siRNA and chemotherapeutic drugs co-delivered into hepatic cancer cells", BIOMATERIALS, vol. 32, no. 8, 1 March 2011 (2011-03-01), pages 2222 - 2232, XP055000456, ISSN: 0142-9612, DOI: 10.1016/j.biomaterials.2010.11.061 *
O'BRIEN J, WILSON I, ORTON T, POGNAN F.: "Investigation of the Alamar Blue (resazurin) fluorescent dye for the assessment of mammalian cell cytotoxicity", EUR J BIOCHEM, vol. 267, no. 17, 2000, pages 5421 - 6, XP002968559, DOI: doi:10.1046/j.1432-1327.2000.01606.x
OTTENSMEIER C.: "The classification of lymphomas and leukemias", CHEM BIOL INTERACT, vol. 135-136, 2001, pages 653 - 64
OWENS DE, 3RD, PEPPAS NA: "Opsonization, biodistribution, and pharmacokinetics of polymeric nanoparticles", INT J PHARM, vol. 307, no. 1, 2006, pages 93 - 102, XP025113094, DOI: doi:10.1016/j.ijpharm.2005.10.010
PASTERNAK G, HOCHHAUS A, SCHULTHEIS B, HEHLMANN R.: "Chronic myelogenous leukemia: molecular and cellular aspects", J CANCER RES CLIN ONCOL, vol. 124, no. 12, 1998, pages 643 - 60
PONKA P, LOK CN.: "The transferrin receptor: role in health and disease", INT J BIOCHEM CELL BIOL, vol. 31, no. 10, 1999, pages 1111 - 37
QIAN ZM, LI H, SUN H, HO K.: "Targeted drug delivery via the transferrin receptor-mediated endocytosis pathway", PHARMACOL REV, vol. 54, no. 4, 2002, pages 561 - 87, XP008054257, DOI: doi:10.1124/pr.54.4.561
RAYBURN ER, ZHANG R.: "Antisense, RNAi, and gene silencing strategies for therapy: mission possible or impossible?", DRUG DISCOV TODAY, vol. 13, no. 11-12, 2008, pages 513 - 21
ROMBERG B, HENNINK WE, STORM G.: "Sheddable coatings for long-circulating nanoparticles", PHARM RES, vol. 25, no. 1, 2008, pages 55 - 71
RYAN SM, MANTOVANI G, WANG X, HADDLETON DM, BRAYDEN DJ.: "Advances in PEGylation of important biotech molecules: delivery aspects", EXPERT OPIN DRUG DELIV, vol. 5, no. 4, 2008, pages 371 - 83, XP008106736, DOI: doi:10.1517/17425247.5.4.371
SAAD M ET AL: "Co-delivery of siRNA and an anticancer drug for treatment of multidrug-resistant cancer", NANOMEDICINE, FUTURE MEDICINE LTD., LONDON, GB, vol. 3, no. 6, 1 December 2008 (2008-12-01), pages 761 - 776, XP008112288, ISSN: 1743-5889, DOI: DOI:10.2217/17435889.3.6.761 *
SAPRA P, ALLEN TM.: "Ligand-targeted liposomal anticancer drugs", PROG LIPID RES, vol. 42, no. 5, 2003, pages 439 - 62, XP002628714, DOI: doi:10.1016/S0163-7827(03)00032-8
SAXON DN, MAYER LD, BALLY MB.: "Liposomal anticancer drugs as agents to be used in combination with other anticancer agents: studies on a liposomal formulation with two encapsulated drugs", J LIPOSOME RES, vol. 9, no. 4, 1999, pages 507 - 522, XP000878068
SAXON DN, MAYER LD, BALLY MB: "Liposomal anticancer drugs as agents to be used in combination with other anticancer agents: studies on a liposomal formulation with two encapsulated drugs", JOURNAL OF LIPOSOME RESEARCH, vol. 9, no. 4, 1999, pages 507 - 522, XP000878068
SEMPLE SC, KLIMUK SK, HARASYM TO, DOS SANTOS N, ANSELL SM, WONG KF, MAURER N, STARK H, CULLIS PR, HOPE MJ: "Efficient encapsulation of antisense oligonucleotides in lipid vesicles using ionizable aminolipids: formation of novel small multilamellar vesicle structures", BIOCHIM BIOPHYS ACTA, vol. 1510, no. 1-2, 2001, pages 152 - 66, XP004248775, DOI: doi:10.1016/S0005-2736(00)00343-6
SHRIVASTAVA N, SRIVASTAVA A.: "RNA interference: an emerging generation of biologicals", BIOTECHNOL J, vol. 3, no. 3, 2008, pages 339 - 53
STAHEL RA, ZANGEMEISTER-WITTKE U.: "Antisense oligonucleotides for cancer therapy-an overview", LUNG CANCER, vol. 41, no. 1, 2003, pages S81 - 8, XP001156189, DOI: doi:10.1016/S0169-5002(03)00147-8
TAKASAKI J, RANEY SG, CHIKH G, SEKIROV L, BRODSKY I, TAM Y, ANSELL SM.: "Methods for the preparation of protein- oligonucleotide-lipid constructs", BIOCONJUG CHEM, vol. 17, no. 2, 2006, pages 451 - 8
TAMM I.: "Antisense therapy in malignant diseases: status quo and quo vadis?", CLIN SCI (LOND), vol. 110, no. 4, 2006, pages 427 - 42, XP055167727, DOI: doi:10.1042/CS20050284
TARDI PG, GALLAGHER RC, JOHNSTONE S, HARASYM N, WEBB M, BALLY MB, MAYER LD.: "Coencapsulation of irinotecan and floxuridine into low cholesterol-containing liposomes that coordinate drug release in vivo", BIOCHIM BIOPHYS ACTA, vol. 1768, no. 3, 2007, pages 678 - 87, XP005900726, DOI: doi:10.1016/j.bbamem.2006.11.014
TORCHILIN V.: "Multifunctional and stimuli-sensitive pharmaceutical nanocarriers", EUR J PHARM BIOPHARM, vol. 71, no. 3, 2009, pages 431 - 44, XP025992165, DOI: doi:10.1016/j.ejpb.2008.09.026
TORCHILIN VP.: "Multifunctional nanocarriers", ADV DRUG DELIV REV, vol. 58, no. 14, 2006, pages 1532 - 55
TORCHILIN VP.: "Targeted pharmaceutical nanocarriers for cancer therapy and imaging", AAPS J, vol. 9, no. 2, 2007, pages E128 - 47, XP035718887, DOI: doi:10.1208/aapsj0902015
USTER PS, ALLEN TM, DANIEL BE, MENDEZ CJ, NEWMAN MS, ZHU GZ.: "Insertion of poly(ethylene glycol) derivatized phospholipid into pre-formed liposomes results in prolonged in vivo circulation time", FEBS LETT, vol. 386, no. 2-3, 1996, pages 243 - 6, XP002236375, DOI: doi:10.1016/0014-5793(96)00452-8
WANG J, GOH B, LU W, ZHANG Q, CHANG A, LIU XY, TAN TM, LEE H.: "In vitro cytotoxicity of Stealth liposomes co-encapsulating doxorubicin and verapamil on doxorubicin- resistant tumor cells", BIOL PHARM BULL, vol. 28, no. 5, 2005, pages 822 - 8, XP002652275
WATERHOUSE DN, DOS SANTOS N, MAYER LD, BALLY MB.: "Drug-drug interactions arising from the use of liposomal vincristine in combination with other anticancer drugs", PHARM RES, vol. 18, no. 9, 2001, pages 1331 - 5
WHEELER JJ, PALMER L, OSSANLOU M, MACLACHLAN I, GRAHAM RW, ZHANG YP, HOPE MJ, SCHERRER P, CULLIS PR.: "Stabilized plasmid-lipid particles: construction and characterization", GENE THER, vol. 6, no. 2, 1999, pages 271 - 81, XP008106998, DOI: doi:10.1038/sj.gt.3300821
YONG WANG ET AL: "Co-delivery of drugs and DNA from cationic core-shell nanoparticles self-assembled from a biodegradable copolymer", NATURE MATERIALS, vol. 5, no. 10, 1 October 2006 (2006-10-01), pages 791 - 796, XP055000458, ISSN: 1476-1122, DOI: 10.1038/nmat1737 *
ZIMMERMANN GR, LEHAR J, KEITH CT.: "Multi-target therapeutics: when the whole is greater than the sum of the parts", DRUG DISCOV TODAY, vol. 12, no. 1-2, 2007, pages 34 - 42, XP005819181, DOI: doi:10.1016/j.drudis.2006.11.008
ZOLI W, RICOTTI L, TESEI A, BARZANTI F, AMADORI D.: "In vitro preclinical models for a rational design of chemotherapy combinations in human tumors", CRIT REV ONCOL HEMATOL, vol. 37, no. 1, 2001, pages 69 - 82

Cited By (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9803197B2 (en) 2012-06-25 2017-10-31 Emory University Particle-nucleic acid conjugates and therapeutic uses related thereto
WO2014003830A1 (en) * 2012-06-25 2014-01-03 Emory University Particle-nucleic acid conjugates and therapeutic uses related thereto
US10155946B2 (en) 2012-06-25 2018-12-18 Emory University Particle-nucleic acid conjugates and therapeutic uses related thereto
US10471149B2 (en) 2012-11-29 2019-11-12 Insmed Incorporated Stabilized vancomycin formulations
WO2014203189A1 (en) * 2013-06-18 2014-12-24 Rosetta Genomics Ltd. Nanocarrier system for micrornas and uses thereof
US10751355B2 (en) 2014-05-15 2020-08-25 Insmed Incorporated Methods for treating pulmonary non-tuberculous mycobacterial infections
US12016873B2 (en) 2014-05-15 2024-06-25 Insmed Incorporated Methods for treating pulmonary non-tuberculous mycobacterial infections
US11446318B2 (en) 2014-05-15 2022-09-20 Insmed Incorporated Methods for treating pulmonary non-tuberculous mycobacterial infections
US11395830B2 (en) 2014-05-15 2022-07-26 Insmed Incorporated Methods for treating pulmonary non-tuberculous mycobacterial infections
US10828314B2 (en) 2014-05-15 2020-11-10 Insmed Incorporated Methods for treating pulmonary non-tuberculous mycobacterial infections
US10398719B2 (en) 2014-05-15 2019-09-03 Insmed Incorporated Methods for treating pulmonary non-tuberculous mycobacterial infections
US10588918B2 (en) 2014-05-15 2020-03-17 Insmed Incorporated Methods for treating pulmonary non-tuberculous mycobacterial infections
CN107683132A (en) * 2015-04-29 2018-02-09 罗得靶点生物技术股份有限公司 Targeted nano carrier for the targeted delivery of drugs of gene therapy
AU2016254729B2 (en) * 2015-04-29 2021-08-05 Rodos Biotarget Gmbh Targeted nanocarriers for targeted drug delivery of gene therapeutics
EP3087974A1 (en) * 2015-04-29 2016-11-02 Rodos BioTarget GmbH Targeted nanocarriers for targeted drug delivery of gene therapeutics
WO2016174250A1 (en) * 2015-04-29 2016-11-03 Rodos Biotarget Gmbh Targeted nanocarriers for targeted drud delivery of gene therapeutics
AU2016291228B2 (en) * 2015-07-09 2020-04-09 Insmed Incorporated Compositions and methods for treating lung diseases and lung injury
EP3319976A4 (en) * 2015-07-09 2019-03-20 Insmed Incorporated Compositions and methods for treating lung diseases and lung injury
WO2017205566A1 (en) * 2016-05-27 2017-11-30 Preceres Inc. Oxidized aminolipidoids and uses thereof
US11491247B2 (en) 2017-05-02 2022-11-08 Cornell University Methods and reagents for tumor targeting with greater efficacy and less toxicity
US11738101B2 (en) 2017-05-02 2023-08-29 Cornell University Methods and reagents for tumor targeting with greater efficacy and less toxicity
US11571386B2 (en) 2018-03-30 2023-02-07 Insmed Incorporated Methods for continuous manufacture of liposomal drug products
US11964052B2 (en) 2021-05-24 2024-04-23 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
CN114940756B (en) * 2022-06-02 2023-11-07 北京清科胜因生物科技有限公司 Poly (2-oxazoline) lipid and lipid nanoparticle and application
CN114940756A (en) * 2022-06-02 2022-08-26 北京清科胜因生物科技有限公司 Poly (2-oxazoline) lipid and lipid nanoparticle and application
CN117582512A (en) * 2022-08-09 2024-02-23 湖南健瑞医药科技有限公司 Metal-polyphenol composite particles, and preparation method and application thereof

Similar Documents

Publication Publication Date Title
WO2011108955A1 (en) Multi -targeting system comprising a nanocarrier, nucleic acid(s) and non-nucleic acid based drug(s)
Yi Xue et al. Lipid-based nanocarriers for RNA delivery
JP5697988B2 (en) Method for silencing polo-like kinase expression using interfering RNA
EP2281041B1 (en) Silencing of csn5 gene expression using interfering rna
Kapoor et al. Physicochemical characterization techniques for lipid based delivery systems for siRNA
Antimisiaris et al. Targeted si-RNA with liposomes and exosomes (extracellular vesicles): How to unlock the potential
US20160081944A1 (en) Oligonucleotide Lipid Nanoparticle Compositions, Methods of Making and Methods of Using the Same
WO2009061515A1 (en) Self-assembling micelle-like nanoparticles for systemic gene delivery
JP2011516586A (en) Lipid formulations for nucleic acid delivery
JP5801055B2 (en) Composition that suppresses expression of target gene
US8802138B2 (en) Methods and compositions for improved deliver, expression or activity of RNA interference agents
WO2011046934A1 (en) Methods and compositions for improved delivery, expression or activity of rna interference agents
CA3215957A1 (en) Tissue-specific nucleic acid delivery by mixed cationic lipid particles
Gujrati et al. Targeted systemic delivery of therapeutic siRNA
MOREIRA et al. Le 18, 1s. 3o3o. 377 covi. A (T) PEDRoso o" º
JP5872898B2 (en) Composition that suppresses expression of target gene
JP5952197B2 (en) Composition that suppresses expression of target gene
US20120207818A1 (en) Composition for suppressing expression of target gene
US20120244210A1 (en) Composition for suppressing expression of target gene
Muralidharan et al. Lipid Nanocarriers for RNAi-Based Cancer Therapy
KR20240072931A (en) Lipid nanoparticle comprising histidine and preparation method thereof
Goodwin et al. Lipids in DNA, RNA, and Peptide Delivery for In Vivo Therapeutic Applications
Arpac Overcoming biological barriers by lipid-based nanocarriers

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 11716085

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11716085

Country of ref document: EP

Kind code of ref document: A1