WO2013105101A1 - Solid lipid nanoparticles entrapping hydrophilic/ amphiphilic drug and a process for preparing the same - Google Patents

Solid lipid nanoparticles entrapping hydrophilic/ amphiphilic drug and a process for preparing the same Download PDF

Info

Publication number
WO2013105101A1
WO2013105101A1 PCT/IN2012/000154 IN2012000154W WO2013105101A1 WO 2013105101 A1 WO2013105101 A1 WO 2013105101A1 IN 2012000154 W IN2012000154 W IN 2012000154W WO 2013105101 A1 WO2013105101 A1 WO 2013105101A1
Authority
WO
WIPO (PCT)
Prior art keywords
glycerol
glycerides
drug
lipid nanoparticles
solid lipid
Prior art date
Application number
PCT/IN2012/000154
Other languages
French (fr)
Other versions
WO2013105101A8 (en
Inventor
Indu Pal KAUR
Rouit BHANDARI
Original Assignee
Department Of Biotechnology
Panjab University
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 Department Of Biotechnology, Panjab University filed Critical Department Of Biotechnology
Publication of WO2013105101A1 publication Critical patent/WO2013105101A1/en
Publication of WO2013105101A8 publication Critical patent/WO2013105101A8/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
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/5123Organic compounds, e.g. fats, sugars
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4409Non condensed pyridines; Hydrogenated derivatives thereof only substituted in position 4, e.g. isoniazid, iproniazid

Definitions

  • the present disclosure relates to entrapping hydrophilic or an amphiphilic drug within nanoparticles and a process for preparing the same.
  • the present disclosure relates to solid lipid nanoparticles and a process for preparing the same.
  • Solid lipid nanoparticles are generally spherical in shape with an average diameter between 10 to 1000 nanometers.
  • Solid lipid nanoparticles possess a solid lipid core matrix that can solubilize lipophilic molecules.
  • the lipid core is stabilized by surfactants (emulsifiers).
  • the lipid typically includes triglycerides (e.g. tristearin), diglycerides (e.g. glycerol behenate), monoglycerides (e.g. glycerol monostearate), fatty acids (e.g. stearic acid), steroids (e.g. cholesterol) and waxes (e.g. cetyl palmitate).
  • emulsifiers with respect to charge and molecular weight have been used to stabilize the lipid dispersion. It has been found that the combination of emulsifiers might prevent particle agglomeration more efficiently (Mehnert et al, 2001 and Small, 1986).
  • Biological membrane lipids such as phospholipids, sphingomyelins, bile salts (sodium taurocholate) and sterols (cholesterol) are utilized as stabilizers.
  • Biological lipids having minimum carrier cytotoxicity and the solid state of the lipid permit better controlled drug release due to increased mass transfer resistance (Manjunath et al., 2005).
  • lipid nanoparticles offer the possibility to develop new therapeutics.
  • the ability to incorporate drugs into nanocarriers offers a new prototype in drug delivery that could hold great promise for attaining the bioavailability enhancement along with controlled and site specific drug delivery.
  • Solid lipid nanoparticles have recently materialized as a novel approach to oral and parenteral drug delivery systems.
  • SLNs combine " the advantages of lipid emulsion and polymeric nanoparticle systems while overcoming the temporal and in- vivo stability iiSues that troubles the conventional as well as polymeric nanoparticle drug delivery approaches (Mehnert et al, 2001).
  • SLNs are the use of physiological lipids, the avoidance of organic solvents, a potential wide application spectrum (oral, dermal, intravenous) and the high pressure homogenization as an established production method. Additionally, improved bioavailability, protection of sensitive drug molecules from the outer environment (water and light) and even controlled release characteristics are claimed by incorporation of poorly water soluble drugs in the solid lipid matrix.
  • SLNs combine numerous advantages over the other colloidal carriers i.e. feasibility of incorporating lipophilic and hydrophilic drugs, no biotoxicity of the carrier, avoidance of organic solvents, possibility of controlled drug release and drug targeting, increased drug stability and no problems with respect to large scale production.
  • hydrophilic drug is meant to refer to a drug that is readily soluble in water. Hydrophilic drugs generally have an aqueous solubility greater than about 10 g/liter.
  • Hydrophobic/lipophilic drug means a drug which lacks affinity for water or which do not readily dissolve in water.
  • Amphiphilic drugs are the drugs which tend to include hydrophobic and/or lipophilic regions.
  • the hydrophilic drugs or amphiphilic drugs have limited permeability across biological barriers such as blood brain barrier (BBB), as biological barriers are highly lipo-philic in nature. Further, as a result of limited permeability across biological barriers these drugs need to be administered in high dose on a daily basis which may lead to several side effects of varying severity.
  • BBB blood brain barrier
  • the drug candidate For the delivery of drug across biological barriers including the blood brain barrier and gut mucosal barrier, the drug candidate must be in a lipid soluble form. Contrary to this hydrophilic drugs or amphiphilic drugs face permeability related problems across biological barriers. Drug delivery systems which can in some way tailor the entry of the hydrophilic or amphiphilic drug candidates across the biological membranes like brain can form a more effective therapy especially for conditions where prolonged therapy is desired. ST>me representative patent documents which disclose lipid nanoparticles are discussed herein below.
  • US Patent No. 5250236 discloses a process for preparing solid lipid microspheres in which a) a molten lipid, which may contain a drug, is contacted with a mixture consisting of water, a surfactant and possibly a co-surfactant heated to a temperature at least equal to the melting temperature of the lipid; b) the obtained micro-emulsion is dispersed in water of 2°C to 10 °C; c) the obtained lipid microsphere dispersion is washed with water by diafiltration and lyophilized. Later two steps make the process lengthy and complex. Furthermore it may lead to formation of aggregates which may not redisperse to give back the same particle size.
  • WO 2006/109317 discloses a process for the preparation of Poly DL-Lactide-coglycolide nanoparticles encapsulated with anti-tubercular drugs.
  • the process includes (i) preparation of an aqueous solution of stable water soluble drugs in DW/NS/PBS, (ii) preparation of unstable drugs in DW/NS/PBS, (iii) preparation of a polymer and hydrophobic drug solution in an organic solvent, (iv) mixing separately the solutions of steps (i) and (ii) with that of step (iii) and sonicating under cold conditions, (v) adding the above emulsion to aqueous poly vinyl alcohol and resonicating under cold conditions, (vi) stirring the emulsion and centrifuging the same and (vii) washing the said particles, reconstituting the same and lyophilizing.
  • the process disclosed in WO 2006/109317 uses an organic solvent (dichloromethane) for the preparation of nanoparticles. The presence of organic solvent
  • US Patent No. 761 1733 discloses solid lipid nanoparticles of antitumor platinum complexes and process for preparing the same.
  • the process includes the following steps: a) first preparing a micro-emulsion by mixing a molten lipid, a surfactant, and optionally a co-surfactant and the platinum compound aqueous solution; b) preparing a solution by mixing a surfactant and optionally a co-surfactant in water, heating to complete solution, preferably at the same melting temperature as of the lipid used in step a) and adding a co- surfactant; c) dispersing the micro-emulsion obtained in step a) into the solution obtained in step b) obtaining a multiple micro-emulsion; d) dispersing the micro-emulsion obtained in step c) in aqueous medium at a temperature ranging from 0.5 °C to 4.0 °C obtaining a dispersion of solid lipid microspheres; e) washing with
  • Pment No. 761 1733 involves preparation of multiple micro-emulsions which renders the method complex.
  • a process for preparing solid lipid nanoparticles comprising the following steps: i) melting at least one lipid selected from the group consisting of glycerides and fatty acids; ii) preparing an aqueous emulsifier mix by admixing at least one emulsifier and water followed by heating at a temperature atleast equal to the melting point of the selected lipid; iii) adding at least one drug to the aqueous emulsifier mix to obtain a solution; iv) adding the solution containing at least one drug to the lipid melt to obtain a microemulsion; and v) preparing solid lipid nanoparticles having a particle size in the range of 10 nm to 1000 nm, preferably 20 nm to 150 nm from the microemulsion.
  • the method step of preparing solid lipid nanoparticles comprises dispersing the microemulsion in water maintained at a temperature ranging between 0 °C and 5 °C under continuous stirring/homogenizing to obtain solid lipid nanoparticles, wherein the ratio of microemulsion and water ranges between 1 : 1 and 1 :4.9.
  • the method step of preparing the solid lipid nanoparticles comprises purging nitrogen through the microemulsion to obtain solid lipid nanoparticles.
  • the glyceride is at least one selected from the group consisting of mono- glycerides, di-glycerides and tri-glycerides.
  • the glyceride is at least one selected from the group consisting of glyceryl behenate tricaprin, trilaurin, trimyristin, tripalmitin, tristearin, 1 ,2-dioctanoyl-s «-glycerol, 1 ,2-didecanoyl- ⁇ «-glycerol, 1 ,2-dilauroyl-5 «-glycerol, 1 ,2-dimyristoyl-,s7?-glycerol, 1 ⁇ -dipalmitoyl-sn-glycerol, 1- palmitoyl-2-oleoyl-.s77-glycerol, 1 -stearoyl-2-linoleoyl-sn-glycerol, 1 -stea
  • the glyceride is glyceryl behenate.
  • the fatty acid is selected from the group consisting of saturated C 4 -C 2 8 fatty acids and unsaturated C 4 -C 28 fatty acids.
  • the fatty acid is stearic acid.
  • the proportion of the glyceride and the fatty acid is in the range of 1 :0 to 1 :20.
  • the proportion of the fatty acid and the glyceride is in the range of 1 :0 to 1 :20.
  • the emulsifier is at least one selected from the group consisting of anionic emulsifiers, cationic emulsifiers, non ionic emulsifiers or zwitterionic emulsifiers.
  • the emulsifier is at least one selected from the group consisting of soy lecithin, egg lecithin, phosphatidylcholine; ethylene oxide copolymers, propylene oxide copolymers, poloxamers, sorbitan ethylene oxide/propylene oxide copolymers, polysorbate 20, polysorbate 60, polysorbate 80, sorbitan esters, span 20, span 40, span 60, span 80, afkyllaryl polyether alcohol polymers, tyloxapol, bile salts, cholate, glycocholate, taurocholate, taurodeoxycholate, gemini surfactants and alcohols.
  • the drug is at least one selected from the group consisting of hydrophilic drugs and amphiphilic drugs.
  • the hydrophilic drug is atleast one selected from the group consisting of isoniazid, rifampicin, pyrazinamide, ethambutol, streptomycin, acyclovir, acetyl cysteine, acetylcholine chloride, alatrofloxacin, alendronate, amantadine hydrochloride, ambenomium, amifostine, amiloride hydrochloride, aminocaproic acid, amphiphilicin B, atenolol, atracurium besylate, atropine, azithromycin, aztreonam, bacitracin, becalermin, belladona, bepridil hydrochloride, bleomycin sulfate, calcitonin, calcitonin salmon, carboplatin, capecitabine, capreomycin sulfate, cefamandole nafate, cefazolin sodium, cefepime hydroch
  • the amphiphilic drug is at least one selected from the group consisting of amphiphilicin B, bupivacaine, ropivacaine, prilocaine, mepivacaine, tetracaine, etidocaine, morphine, fentanyl, alfentanil and sulfentanil.
  • the drug is a compound containing at least one hydroxyl group or at least one hydrophilic linkage.
  • the method step of dispersing the micro emulsion in water is carried out by continuous stirring/homogenizing at 4,000 to 12,000 rpm, preferably 5,000 to 7,000 rpm for a time period ranging between 20 minutes and 2 hours.
  • solid lipid nanoparticles prepared by the process of the present disclosure, said solid lipid nanoparticles comprising: i) at least one lipid selected from the group consisting of glycerides and fatty acids; ii) at least one drug selected from the group consisting of hydrophilic drugs and amphiphilic drugs; and iii) at least one emulsifier.
  • the amount of drug is in the range of about 0.001% to about 99% with respect to the mass of the lipid.
  • the particle size of the solid lipid nanoparticles ranges between 10 to 1000 nm, preferably between 20- 150 nm.
  • Figure 1 illustrates comparative In-vitro drug release.
  • the present disclosure provides solid lipid nanopaiticles (SLNs) containing drug selected from the group consisting of hydrophilic drugs and amphiphilic drugs.
  • SSNs solid lipid nanopaiticles
  • the solid lipid nanoparticles with a particle size in the range of 10 to 1000 nm, preferably between 20-150 nm prepared in accordance with the present disclosure can bypass reticuloendothelial system (RES) detection and limit metabolic elimination of the entrapped drug.
  • the active ingredient (hydrophilic drug or amphiphilic drug) entrapped in solid lipid nanoparticles (having a particle size less than 200 nm) effectively cross the biological barriers such as cerebral barrier and gut mucosal barrier.
  • the enhanced targetability, permeability and bioavailability of hydrophilic drug or amphiphilic drug across the biological barriers including blood brain barrier and gut mucosal barrier are achieved as a result of solid lipid nanoparticles having a particle size below 200 nm.
  • the solid lipid nanoparticles prepared in accordance with the present disclosure in turn reduce the dose and the dosing frequency of hydrophilic drugs or amphiphilic drugs. Furthermore, the solid lipid nanoparticles prepared in accordance with the present disclosure provides sustained / controlled therapeutic effect with reduced side effects.
  • the selected lipid or lipid mixture is heated to melt.
  • the lipid is selected from the group consisting glycerides and fatty acids.
  • the glyceride used to prepare the lipid melt in accordance with present disclosure is selected from the group consisting of mono-glycerides, di-glycerides and tri-glycerides.
  • the glyceride used include but is not limited to glyceryl behenate, tricaprin, trilaurin, trimyristin, tripalmitin, tristearin, 1,2-dioctanoyl- s «-glycerol, l,2-didecanoyl-s «-glycerol, 1 ,2-dilauroyl-5 «-glycerol, 1 ,2-dimyristoyl-s «- glycerol, l,2-dipalmitoyl-i «-glycerol, l-palmitoyl-2-oleoyl-s «-glycerol, 1 -stearoyl-2- linoleoyl-s «-glycerol, 1 -stearoyl-2-arachidonoyl-s «-glycerol, 1 -stearoyl-2- docosahexaenoyl-sn-glycerol, 1 -
  • glyceryl behenate is preferred in accordance with present disclosure.
  • the fatty acid used to prepare a lipid melt is selected from the group consisting of saturated C 4 -C 28 fatty acids and unsaturated C 4 -C 2 8 fatty acids.
  • one of the preferred fatty acids is stearic acid.
  • the proportion of the glyceride and the fatty acid is : maintained in between 1 :0 to 1 :20 and vice versa.
  • an aqueous emulsifier mix is prepared separately by admixing an emulsifier and water followed by heating at a temperature at least equal to the melting point of the selected lipid.
  • the emulsifier used for the preparation of emulsifier mix in accordance with the present disclosure include anionic emulsifiers, cationic emulsifiers, non ionic emulsifiers or zwitterionic emulsifiers which include but is not limited to soy lecithin, egg lecithin, phosphatidylcholine; ethylene oxide copolymers, propylene oxide copolymers, poloxamers, sorbitan ethylene oxide/propylene oxide copolymers, polysorbate 20, polysorbate 60, polysorbate 80, sorbitan esters, span 20, span 40, span 60, span 80, alkyllaryl polyether alcohol polymers, tyloxapol, bile salts, cholate, glycocholate, taurocholate, taurodeoxycholate, gemini surfactants, alcohols and the like.
  • a dispersion containing at least one drug is obtained by adding at least one drug to the aqueous emulsifier mix obtained in the second step.
  • the drug is at least one selected from the group consisting of hydrophilic drugs and amphiphilic drugs.
  • Trie hydrophilic drug includes but is not limited to isoniazid, rifampicin, pyrazinamide, ethambutol, streptomycin, acyclovir; acetyl cysteine, acetylcholine chloride, alatrofloxacin, alendronate, amantadine hydrochloride, ambenomium, amifostine, amiloride hydrochloride, aminocaproic acid, amphiphilicin B, atenolol, atracurium besylate, atropine, azithromycin, aztreonam, bacitracin, becalermin, belladona, bepridil hydrochloride, bleomycin sulfate, calcitonin, calcitonin salmon, carboplatin, capecitabine, capreomycin sulfate, cefamandole
  • amphiphilic drug includes but is not limited to amphiphilicin B, bupivacaine, ropivacaine, prilocaine, mepivacaine, tetrocaine, etidocaine, morphine, fentanyl, alfentanil, sulfentanil and the like.
  • ⁇ ie solid lipid nanoparticles of the present disclosure contain a drug which contains at least one hydroxy 1 group or at least one hydrophilic linkage.
  • a micro emulsion is obtained by adding the solution containing at least one drug obtained in the third step to the lipid melt obtained in the first.
  • solid lipid nanoparticles are prepared from the microemulsion.
  • the microemulsion obtained in the fourth step is dispersed in water maintained at a temperature ranging between 0 °C and 5 °C under continuous stirring/homogenizing.
  • the stirring is carried out at 4,000 to 12,000 rpm for a time period between 20 minutes to 2 hours.
  • the stirring is preferably carried out at 5,000 to 7,000 rpm.
  • the ratio of microemulsion and water ranges between 1 : 1 and 1 :4.9.
  • the alternative way of obtaining solid lipid nanoparticles from the microemulsion is by purging nitrogen through the microemulsion.
  • the solid lipid nanoparticles prepared in accordance with the present disclosure have a particle size in the range of 10 nm to lOOOnm.
  • the preferred particle size of the solid lipid nanoparticles is in the range of 20 nm to 150 nm.
  • solid lipid nanoparticles prepared by the process of present disclosure; said solid lipid nanoparticles comprising: i) at least lipid selected from the group consisting of glycerides and fatty acids; ii) at least one drug selected from the group consisting of hydrophilic drugs and amphiphilic drugs; and iii) at least one emulsifier.
  • the amount of drug is in the range of about 0.001% to about 99%.
  • the particle size of the solid lipid nanoparticles prepared by the process of present disclosure is in the range of 10 to 1000 nm, preferably between 20-150 nm. Tfie present disclosure is further described in light of the following examples which are set forth for illustration purpose only and not to be construed for limiting the scope of the disclosure.
  • Example 1 Procedure for preparing the solid lipid nanoparticles containing hydrophilic drug
  • glyceryl behenate was heated at 90 °C to obtain a lipid melt.
  • 7.9 gm of Isoniazid was added as hydrophilic drug to the emulsifier mix at 90 °C to obtain an aqueous solution containing isoniazid.
  • the solution containing isoniazid was added to lipid melt, under continuous magnetic stirring at 90 °C to obtain a clear microemulsion.
  • the 1000.0 gm of microemulsion thus formed was transferred into 1000.0 gm of cold water maintained at 2°C under continuous mechanical stirring/homogenizing at 5,000 rpm for a time period of about 30 minutes using a Wise Tis Homogenizer.
  • Example 2 Procedure for preparing the solid lipid nanoparticles containing hydrophilic drug
  • Example 3 Procedure for preparing the solid lipid nanoparticles containing hydrophilic drug
  • glyceryl behenate 63.36 gm of glyceryl behenate and 15.84 gm of stearic acid were heated at 90 °C to obtain a lipid melt. 7.9 gm of Isoniazid was added as hydrophilic drug to the emulsifier mix at 90 °C to obtain an aqueous solution containing isoniazid. Then, the solution containing isoniazid was added to lipid melt, under continuous magnetic stirring at 90 °C to obtain a clear microemulsion.
  • TTie 1000.0 gm of microemulsion thus formed was transferred into 1000.0 gm of cold water maintained at 2°C under continuous mechanical stirring/homogenizing at 5,000 rpm for a time period of about 30 minutes using a Wise Tis Homogenizer.
  • Example 4 Procedure for preparing the solid lipid nanoparticles containing hydrophilic drug
  • the solid lipid nanoparticles prepared in accordance with the present disclosure were evaluated for various parameters such as total drug content, entrapment efficiency, particle size, zeta potential, poly dispersity index, in-vitro release (figure 1). The results are provided in Table 1.
  • Solid lipid nanoparticles of Example 1 were evaluated for particle size, poly dispersity index and zeta potential using Beckman Coulter Delsa(TM) Nano Zeta Potential and Submicron Particle Size Analyzer wherein triple distilled water (TDW) was used as a diluting medium.
  • TDC total drug content
  • EE entrapment efficiency
  • SSNs Solid lipid nanoparticles
  • Table No. 1 Determination of particle size (D), poly dispersity index (PDI), total drug content (TDC), entrapment efficiency (EE) and zeta potential of the solid lipid nano particles containing isoniazid drug.
  • Solid lipid nanoparticles prepared in accordance with the present disclosure exhibited a small particle size below 120 nm.
  • the total drug content was found to be greater than 92% w/v.
  • the poly dispersity index for all the developed solid lipid nanoparticles was found to be below 0.3 which indicates a narrow particle size distribution.
  • the entrapment efficiency for solid lipid nanoparticles of combination of glyceryl behenate and stearic acid was found to be 84.032 ⁇ 1.103% in comparison to 77.600 ⁇ 1.961% and 69.013 ⁇ 0.675% for solid lipid nanoparticles of glyceryl behenate and solid lipid nanoparticles of stearic acid, respectively.
  • the solid lipid nanoparticles prepared using a combination of lipids or a lipid and a fatty acid prepared in accordance with the present disclosure possesses enhanced entrapment efficiency for hydrophilic and/or amphiphilic drugs.
  • T3 ⁇ 4ble No. 2 In-vitro drug release of isoniazid from Solid Lipid Nanoparticles of the present disclosure.
  • solid lipid nanoparticles solid lipid nanoparticles of glyceryl behenate, solid lipid nanoparticles of stearic acid and solid lipid nanoparticles of combination of glyceryl behenate and stearic acid.
  • Group A and Group B Two groups each containing forty two female Wistar rats were formed and named as Group A and Group B. Both the groups of rats were kept at uniform atmospheric conditions.
  • the Wistar rats of Group A were given isoniazid (free drug) by an oral route while rats of Group B were given solid lipid nanoparticles of combination of glyceryl behenate and stearic acid containing isoniazid drug by an oral route.
  • Drug concentration in the samples was determined using HPLC. The plasma samples were collected before sacrificing the rats. Six Wistar rats were sacrificed per time point. The details are mentioned below.
  • 20 % aqueous tissue homogenates were prepared in cold 150 mM KC1. The homogenates were then centrifuged at 15000 rpm for 10 minutes at 4 °C to obtain clear tissue homogenates. To 150- ⁇ aliquot of the clear tissue homogenates, a 300 ⁇ . of the deproteinizing agent (methanol) was added to obtain a dispersion which was vortexed for 2 minutes. The samples were then centrifuged at 15000 rpm for 10 minutes at 4 °C. The supernatant was then collected and an equal volume of water was added to the clear supernatant. The samples were then filtered (0.20 ⁇ nylon filters) and were injected into the HPLC system.
  • the deproteinizing agent methanol
  • the HPLC system consisted of Waters Alliance (Waters Corporation, MA, USA) equipped with a Waters e-2695 ALLIANCE separation module comprising of a solvent (quaternary gradient mode), auto injector, column oven and a 4 channel in line degasser, a sample management system (sample heater cooler) and a 2998 PDA detector. Chromatographic separation was performed using symmetry CI 8 column at 254 nm. Data acquisition was performed by the Empower 2® software.
  • the mobile phase consisted of a mixture phosphate buffer of pH 6.8 and methanol (85: 15). The mobile phase was delivered at a flow rate of 0.9 ml/min and the detection of isoniazid (INH) was carried out at 254 nm.
  • the injection volume was 20 ⁇ , and the analysis was performed at 30°C (sample 10°C).
  • Table No. 3 Comparative analysis of concentration of isoniazid in plasma.
  • the solid lipid nanoparticles with a particle size in the range of 10 to 1000 nm, preferably between 20-150 nm prepared in accordance with the present disclosure provide enhanced concentration of hydrophilic drug or amphiphilic drug in plasma.
  • the solid lipid nanoparticles prepared in accordance with the present disclosure improves cerebral permeability of hydrophilic drug or amphiphilic drug entrapped therein.
  • the solid lipid nanoparticles prepared in accordance with the present disclosure enhance the bioavailability of the hydrophilic drug or amphiphilic drug as a result of improved permeability across the biological barriers.
  • the improved bioavailability reduces dose, dosing frequency and dose dependent side effects of the drug.
  • the applicant craves leave to submit further clinical data.

Abstract

Solid lipid nanoparticles and the process thereof are disclosed. The solid lipid nanoparticles comprise: at least one lipid selected from the group consisting of glycerides and fatty acids, at least one hydrophilic drug or amphiphilic drug and at least one emulsifier.

Description

sfeuD LIPID NANOPARTICLES ENTRAPPING HYDROPHILIC/ AMPHIPHILIC DRUG AND A PROCESS FOR PREPARING THE SAME
FIELD OF THE DISCLOSURE
The present disclosure relates to entrapping hydrophilic or an amphiphilic drug within nanoparticles and a process for preparing the same.
Particularly, the present disclosure relates to solid lipid nanoparticles and a process for preparing the same.
BACKGROUND
Solid lipid nanoparticles (SLN) are generally spherical in shape with an average diameter between 10 to 1000 nanometers. Solid lipid nanoparticles possess a solid lipid core matrix that can solubilize lipophilic molecules. The lipid core is stabilized by surfactants (emulsifiers). The lipid typically includes triglycerides (e.g. tristearin), diglycerides (e.g. glycerol behenate), monoglycerides (e.g. glycerol monostearate), fatty acids (e.g. stearic acid), steroids (e.g. cholesterol) and waxes (e.g. cetyl palmitate). Various classes of emulsifiers (with respect to charge and molecular weight) have been used to stabilize the lipid dispersion. It has been found that the combination of emulsifiers might prevent particle agglomeration more efficiently (Mehnert et al, 2001 and Small, 1986).
Biological membrane lipids such as phospholipids, sphingomyelins, bile salts (sodium taurocholate) and sterols (cholesterol) are utilized as stabilizers. Biological lipids having minimum carrier cytotoxicity and the solid state of the lipid permit better controlled drug release due to increased mass transfer resistance (Manjunath et al., 2005).
Development of solid lipid nanoparticles is one of the emerging fields of nanotechnology with several potential applications in drug delivery, clinical medicine and research, as well as in other discipline. Due to their unique size-dependent properties, lipid nanoparticles offer the possibility to develop new therapeutics. The ability to incorporate drugs into nanocarriers offers a new prototype in drug delivery that could hold great promise for attaining the bioavailability enhancement along with controlled and site specific drug delivery.
Solid lipid nanoparticles have recently materialized as a novel approach to oral and parenteral drug delivery systems. SLNs combine "the advantages of lipid emulsion and polymeric nanoparticle systems while overcoming the temporal and in- vivo stability iiSues that troubles the conventional as well as polymeric nanoparticle drug delivery approaches (Mehnert et al, 2001).
Advantages of SLNs are the use of physiological lipids, the avoidance of organic solvents, a potential wide application spectrum (oral, dermal, intravenous) and the high pressure homogenization as an established production method. Additionally, improved bioavailability, protection of sensitive drug molecules from the outer environment (water and light) and even controlled release characteristics are claimed by incorporation of poorly water soluble drugs in the solid lipid matrix.
It has been proposed that SLNs combine numerous advantages over the other colloidal carriers i.e. feasibility of incorporating lipophilic and hydrophilic drugs, no biotoxicity of the carrier, avoidance of organic solvents, possibility of controlled drug release and drug targeting, increased drug stability and no problems with respect to large scale production.
The term "hydrophilic drug" is meant to refer to a drug that is readily soluble in water. Hydrophilic drugs generally have an aqueous solubility greater than about 10 g/liter.
Hydrophobic/lipophilic drug means a drug which lacks affinity for water or which do not readily dissolve in water.
Amphiphilic drugs are the drugs which tend to include hydrophobic and/or lipophilic regions.
The hydrophilic drugs or amphiphilic drugs have limited permeability across biological barriers such as blood brain barrier (BBB), as biological barriers are highly lipo-philic in nature. Further, as a result of limited permeability across biological barriers these drugs need to be administered in high dose on a daily basis which may lead to several side effects of varying severity. For the delivery of drug across biological barriers including the blood brain barrier and gut mucosal barrier, the drug candidate must be in a lipid soluble form. Contrary to this hydrophilic drugs or amphiphilic drugs face permeability related problems across biological barriers. Drug delivery systems which can in some way tailor the entry of the hydrophilic or amphiphilic drug candidates across the biological membranes like brain can form a more effective therapy especially for conditions where prolonged therapy is desired. ST>me representative patent documents which disclose lipid nanoparticles are discussed herein below.
US Patent No. 5250236 discloses a process for preparing solid lipid microspheres in which a) a molten lipid, which may contain a drug, is contacted with a mixture consisting of water, a surfactant and possibly a co-surfactant heated to a temperature at least equal to the melting temperature of the lipid; b) the obtained micro-emulsion is dispersed in water of 2°C to 10 °C; c) the obtained lipid microsphere dispersion is washed with water by diafiltration and lyophilized. Later two steps make the process lengthy and complex. Furthermore it may lead to formation of aggregates which may not redisperse to give back the same particle size.
WO 2006/109317 discloses a process for the preparation of Poly DL-Lactide-coglycolide nanoparticles encapsulated with anti-tubercular drugs. The process includes (i) preparation of an aqueous solution of stable water soluble drugs in DW/NS/PBS, (ii) preparation of unstable drugs in DW/NS/PBS, (iii) preparation of a polymer and hydrophobic drug solution in an organic solvent, (iv) mixing separately the solutions of steps (i) and (ii) with that of step (iii) and sonicating under cold conditions, (v) adding the above emulsion to aqueous poly vinyl alcohol and resonicating under cold conditions, (vi) stirring the emulsion and centrifuging the same and (vii) washing the said particles, reconstituting the same and lyophilizing. The process disclosed in WO 2006/109317 uses an organic solvent (dichloromethane) for the preparation of nanoparticles. The presence of organic solvent may cause toxic effect in case of prolonged treatment, as is required for the anti-tubercular therapy.
US Patent No. 761 1733 discloses solid lipid nanoparticles of antitumor platinum complexes and process for preparing the same. The process includes the following steps: a) first preparing a micro-emulsion by mixing a molten lipid, a surfactant, and optionally a co-surfactant and the platinum compound aqueous solution; b) preparing a solution by mixing a surfactant and optionally a co-surfactant in water, heating to complete solution, preferably at the same melting temperature as of the lipid used in step a) and adding a co- surfactant; c) dispersing the micro-emulsion obtained in step a) into the solution obtained in step b) obtaining a multiple micro-emulsion; d) dispersing the micro-emulsion obtained in step c) in aqueous medium at a temperature ranging from 0.5 °C to 4.0 °C obtaining a dispersion of solid lipid microspheres; e) washing with aqueous medium through ultra filtration to obtain lipid microspheres and lyophilizing, optionally in the presence of a bulking agent and of a cryoprotecting agent. The method disclosed in US ■
Pment No. 761 1733 involves preparation of multiple micro-emulsions which renders the method complex.
The representative patent documents disclose comple methods of preparation of solid lipid nanoparticles. Further, the methods disclosed are not suitable for entrapment of hydrophilic or amphiphilic drugs and effective delivery of drug across biological barriers. In view of the above, there is envisaged in accordance with the present disclosure solid lipid nanoparticles having enhanced hydrophilic or amphiphilic drug entrapment efficiency and improved delivery through biological barriers.
OBJECTS:
Some of the objects of the present disclosure are as follows:
It is an object of the present disclosure to provide solid lipid nanoparitcles.
It is another object of the present disclosure to provide a simple process for preparing solid lipid nanoparticles.
It is yet another object of the present disclosure to provide a process which is capable of producing concentrated solid lipid nanoparticles without using lyophilization technique.
It is yet another object of the present disclosure to provide a simple process for preparing solid lipid nanoparticles using at least one lipid.
It is still another object of the present disclosure to provide solid lipid nanoparticles having improved drug entrapment efficiency.
It is still another object of the present disclosure to provide solid lipid nanoparticles with improved entrapment efficiency of hydrophilic / amphiphilic drugs.
It is a further object of the present disclosure to provide solid lipid nanoparticles which can effectively deliver the drug across biological barriers such as cerebral barrier, gut mucosal barrier and the like. It is still another object of the present disclosure to provide solid lipid nanoparticles which are able to improve the pharmacokinetic behavior (viz Cmax, AUC, clearance and t1/2) of the drug entrapped therein.
It is still further object of the present disclosure to provide solid lipid nanoparticles which reduce the dosing frequency and dose dependent side effects.
SUMMARY
These and other objects of the present disclosure are to a great extent dealt in the disclosure.
In one aspect of the present disclosure there is provided a process for preparing solid lipid nanoparticles, said process comprising the following steps: i) melting at least one lipid selected from the group consisting of glycerides and fatty acids; ii) preparing an aqueous emulsifier mix by admixing at least one emulsifier and water followed by heating at a temperature atleast equal to the melting point of the selected lipid; iii) adding at least one drug to the aqueous emulsifier mix to obtain a solution; iv) adding the solution containing at least one drug to the lipid melt to obtain a microemulsion; and v) preparing solid lipid nanoparticles having a particle size in the range of 10 nm to 1000 nm, preferably 20 nm to 150 nm from the microemulsion.
Typically, the method step of preparing solid lipid nanoparticles comprises dispersing the microemulsion in water maintained at a temperature ranging between 0 °C and 5 °C under continuous stirring/homogenizing to obtain solid lipid nanoparticles, wherein the ratio of microemulsion and water ranges between 1 : 1 and 1 :4.9.
Alternatively, the method step of preparing the solid lipid nanoparticles comprises purging nitrogen through the microemulsion to obtain solid lipid nanoparticles.
Typically, the glyceride is at least one selected from the group consisting of mono- glycerides, di-glycerides and tri-glycerides. in accordance with one of the embodiments of the present disclosure the glyceride is at least one selected from the group consisting of glyceryl behenate tricaprin, trilaurin, trimyristin, tripalmitin, tristearin, 1 ,2-dioctanoyl-s«-glycerol, 1 ,2-didecanoyl-^«-glycerol, 1 ,2-dilauroyl-5«-glycerol, 1 ,2-dimyristoyl-,s7?-glycerol, 1 ^-dipalmitoyl-sn-glycerol, 1- palmitoyl-2-oleoyl-.s77-glycerol, 1 -stearoyl-2-linoleoyl-sn-glycerol, 1 -stearoyl-2- arachidonoy l-sn-gly cerol, 1 -stearoy 1-2-docosahexaenoy 1-^n-glycerol, 1 -oleoy 1-2-acety 1- .sn-glycerol, 1,2-di-O-phytanyl-^-glycerol, 1 ,2-dipalmitoyl ethylene glycol, 1-2-dioleoyl ethylene glycol, glyceryl monostearate, behenoyl polyoxyl-8 glycerides, glyceryl palmitostearate, 1 -O-hexadecyl-src-glycerol, l-O-hexadecyl-2-acetyl-j,«-glycerol, l-O- hexadecyl-2-O-methyl-5«-gly cerol, l,2-diacyl-3-O-(a-D-glucopyranosyl)-s«-gly cerol, stearoyl macrogol-32 glycerides, stearoyl polyoxyl-32 glycerides, lauroyl macrogol-32 glycerides, lauroyl polyoxyl-32 glycerides, lauroyl macrogol-6 glycerides, lauroyl polyoxyl-6 glycerides, oleoyl macrogol-6 glycerides, oleoyl polyoxyl-6 glycerides, linoleoyl macrogol-6 glycerides, polyglyceryl-3 dioleate, glycerol monolinoleate, glyceryl monolinoleate, glycerol monooleates, diethylene glycol monoethyl ether, glyceryl dibehenate, glycerol distearate, glyceryl distearate, glyceryl dipalmitostearate and linoleoyl polyoxyl-6 glyceride.
Preferably, the glyceride is glyceryl behenate.
Typically, the fatty acid is selected from the group consisting of saturated C4-C28 fatty acids and unsaturated C4-C28 fatty acids.
Preferably, the fatty acid is stearic acid.
Typically, the proportion of the glyceride and the fatty acid is in the range of 1 :0 to 1 :20.
Alternatively, the proportion of the fatty acid and the glyceride is in the range of 1 :0 to 1 :20.
Typically, the emulsifier is at least one selected from the group consisting of anionic emulsifiers, cationic emulsifiers, non ionic emulsifiers or zwitterionic emulsifiers.
In accordance with one of the embodiment of the present disclosure the emulsifier is at least one selected from the group consisting of soy lecithin, egg lecithin, phosphatidylcholine; ethylene oxide copolymers, propylene oxide copolymers, poloxamers, sorbitan ethylene oxide/propylene oxide copolymers, polysorbate 20, polysorbate 60, polysorbate 80, sorbitan esters, span 20, span 40, span 60, span 80, afkyllaryl polyether alcohol polymers, tyloxapol, bile salts, cholate, glycocholate, taurocholate, taurodeoxycholate, gemini surfactants and alcohols.
Typically, the drug is at least one selected from the group consisting of hydrophilic drugs and amphiphilic drugs.
Typically, the hydrophilic drug is atleast one selected from the group consisting of isoniazid, rifampicin, pyrazinamide, ethambutol, streptomycin, acyclovir, acetyl cysteine, acetylcholine chloride, alatrofloxacin, alendronate, amantadine hydrochloride, ambenomium, amifostine, amiloride hydrochloride, aminocaproic acid, amphiphilicin B, atenolol, atracurium besylate, atropine, azithromycin, aztreonam, bacitracin, becalermin, belladona, bepridil hydrochloride, bleomycin sulfate, calcitonin, calcitonin salmon, carboplatin, capecitabine, capreomycin sulfate, cefamandole nafate, cefazolin sodium, cefepime hydrochloride, cefixime, cefonicid sodium, cefoperazone, cefotetan disodium, cefotoxime, cefoxitin sodium, ceftizoxime, ceftriaxone, cefuroxime axetil, cephalexin, cephapirin sodium, chrionic gonadotropin, cidofovir, cisplatin, cladribine, clidinium bromide, clindamycin, ciprofloxacin, clondronate, colistimethate sodium, deferoxamine, denileukin diftitox, desmopressin, diatrizoate megluamine, dicyclomine, didanosine, dirithromycin, dopamine hydrochloride, dornase alpha doxacurium chloride, doxorubicin, editronate disodium, elanaprilat, enkephalin, enoxacin, enoxaprin sodium, ephedrine, epinephrine, erythromycin, esmol hydrochloride, famiciclovir, fludarabine, fluoxetine, ganciclovir, gentamycin, glucagon, glycopyrolate, heparin sodium, indinavir sulfate, insulin, lamivudine, leucovorin calcium, leuprolide acetate, levofloxacin, lincomycin, lobucavir, lomefloxacin, loracarbef, mannitol, methotrexate, methscopolamine, metformin hydrochloride, metroprolol, mezocillin sodium, mivacurium chloride, nedocromil sodium, neostigmine bromide, neostigmine methyl sulfate, neutontin, norfloxacin, octreotide acetate, ofloxacin, olpadronate, oxytocin, pamidronate disodium, pancuronium bromide, paroxetine, pefloxacin, pentamindine isethionate, pentostatin, pentoxifylline, periciclovir, pentagastrin, phentolamine mesylate, phenylalanine, physostigmine salicylate, piperacillin sodium, polymixin B sulfate, pralidoxine chloride, pramlintide, pregabalin, propofenone, propenthaline bromide, pyridostigmine bromide, residronate, ribavarin, rimantadine hydrochloride, salmetrol xinafoate, sinca!ide, solatol, somatostatin, sparfloxacin, spectinomycin, stavudine, streptozocin, suxamethonium chloride, tacrine hydrochloride, terbutaline sulfate, thiopbeta ticarcillin, tiludronate, timolol, trandolapril, trimetrexate gluconate, trospectinomycin, trovafloxacin, tubocurarine chloride, valaciclovir, valsartan, vasopressin, vecoronium bromide, vfhblastin, vincristine, vinorelbine, warfarin sodium, zalcitabine, zanamavir, zolandronate and zidovudine.
Typically, the amphiphilic drug is at least one selected from the group consisting of amphiphilicin B, bupivacaine, ropivacaine, prilocaine, mepivacaine, tetracaine, etidocaine, morphine, fentanyl, alfentanil and sulfentanil.
Typically, the drug is a compound containing at least one hydroxyl group or at least one hydrophilic linkage.
Typically, the method step of dispersing the micro emulsion in water is carried out by continuous stirring/homogenizing at 4,000 to 12,000 rpm, preferably 5,000 to 7,000 rpm for a time period ranging between 20 minutes and 2 hours.
In another aspect of the present disclosure there is provided solid lipid nanoparticles prepared by the process of the present disclosure, said solid lipid nanoparticles comprising: i) at least one lipid selected from the group consisting of glycerides and fatty acids; ii) at least one drug selected from the group consisting of hydrophilic drugs and amphiphilic drugs; and iii) at least one emulsifier.
Typically, the amount of drug is in the range of about 0.001% to about 99% with respect to the mass of the lipid.
Typically, the particle size of the solid lipid nanoparticles ranges between 10 to 1000 nm, preferably between 20- 150 nm.
BRIEF DESCRIPTION OF ACCOMPANYING DRAWING:
The disclosure will now be described with reference to accompanying drawing.
Figure 1 illustrates comparative In-vitro drug release. tfETAIL DESCRIPTION OF THE DISCLOSURE
The present disclosure provides solid lipid nanopaiticles (SLNs) containing drug selected from the group consisting of hydrophilic drugs and amphiphilic drugs.
The solid lipid nanoparticles with a particle size in the range of 10 to 1000 nm, preferably between 20-150 nm prepared in accordance with the present disclosure can bypass reticuloendothelial system (RES) detection and limit metabolic elimination of the entrapped drug. The active ingredient (hydrophilic drug or amphiphilic drug) entrapped in solid lipid nanoparticles (having a particle size less than 200 nm) effectively cross the biological barriers such as cerebral barrier and gut mucosal barrier.
The enhanced targetability, permeability and bioavailability of hydrophilic drug or amphiphilic drug across the biological barriers including blood brain barrier and gut mucosal barrier are achieved as a result of solid lipid nanoparticles having a particle size below 200 nm.
The solid lipid nanoparticles prepared in accordance with the present disclosure in turn reduce the dose and the dosing frequency of hydrophilic drugs or amphiphilic drugs. Furthermore, the solid lipid nanoparticles prepared in accordance with the present disclosure provides sustained / controlled therapeutic effect with reduced side effects.
In one aspect of the present disclosure there is provided a process for preparing solid lipid nanoparticles.
In the first step, the selected lipid or lipid mixture is heated to melt. The lipid is selected from the group consisting glycerides and fatty acids.
The glyceride used to prepare the lipid melt in accordance with present disclosure is selected from the group consisting of mono-glycerides, di-glycerides and tri-glycerides.
In accordance with the present disclosure the glyceride used include but is not limited to glyceryl behenate, tricaprin, trilaurin, trimyristin, tripalmitin, tristearin, 1,2-dioctanoyl- s«-glycerol, l,2-didecanoyl-s«-glycerol, 1 ,2-dilauroyl-5«-glycerol, 1 ,2-dimyristoyl-s«- glycerol, l,2-dipalmitoyl-i«-glycerol, l-palmitoyl-2-oleoyl-s«-glycerol, 1 -stearoyl-2- linoleoyl-s«-glycerol, 1 -stearoyl-2-arachidonoyl-s«-glycerol, 1 -stearoyl-2- docosahexaenoyl-sn-glycerol, 1 -oleoyl-2-acetyl-^-glycerol, 1 ,2-di-O-phytanyl-.s77- glycerol, 1,2-dipalmitoyl ethylene glycol, 1-2-dioleoyl ethylene glycol, glyceryl monostearate, behenoyl polyoxyl-8 glycerides, glyceryl palmitostearate, 1-O-hexadecyl- sn-glycerol, l-O-hexadecyl-2-acetyl-.w-glycerol, 1 -O-hexadecyl-2-O-methyl-^«-glycerol, l,2-diacyl-3-O-(a-D-glucopyranosyl)-1y«-glycerol, stearoyl macrogol-32 glycerides, stearoyl polyoxyl-32 glycerides, lauroyl macrogol-32 glycerides, lauroyl polyoxyl-32 glycerides, lauroyl macrogol-6 glycerides, lauroyl polyoxyl-6 glycerides, oleoyl macrogol-6 glycerides, oleoyl polyoxyl-6 glycerides, linoleoyl macrogol-6 glycerides, polyglyceryl-3 dioleate, glycerol monolinoleate, glyceryl monolinoleate, glycerol monooleates, diethylene glycol monoethyl ether, glyceryl dibehenate, glycerol distearate, glyceryl distearate, glyceryl dipalmitostearate and linoleoyl polyoxyl-6 glyceride.
Amongst the various glycerides, glyceryl behenate is preferred in accordance with present disclosure.
In accordance with the present disclosure the fatty acid used to prepare a lipid melt is selected from the group consisting of saturated C4-C28 fatty acids and unsaturated C4-C28 fatty acids. In accordance with the present disclosure, one of the preferred fatty acids is stearic acid.
During the preparation of a lipid melt, the proportion of the glyceride and the fatty acid is : maintained in between 1 :0 to 1 :20 and vice versa.
In the second step, an aqueous emulsifier mix is prepared separately by admixing an emulsifier and water followed by heating at a temperature at least equal to the melting point of the selected lipid.
The emulsifier used for the preparation of emulsifier mix in accordance with the present disclosure include anionic emulsifiers, cationic emulsifiers, non ionic emulsifiers or zwitterionic emulsifiers which include but is not limited to soy lecithin, egg lecithin, phosphatidylcholine; ethylene oxide copolymers, propylene oxide copolymers, poloxamers, sorbitan ethylene oxide/propylene oxide copolymers, polysorbate 20, polysorbate 60, polysorbate 80, sorbitan esters, span 20, span 40, span 60, span 80, alkyllaryl polyether alcohol polymers, tyloxapol, bile salts, cholate, glycocholate, taurocholate, taurodeoxycholate, gemini surfactants, alcohols and the like.
In the third step, a dispersion containing at least one drug is obtained by adding at least one drug to the aqueous emulsifier mix obtained in the second step.
The drug is at least one selected from the group consisting of hydrophilic drugs and amphiphilic drugs. Trie hydrophilic drug includes but is not limited to isoniazid, rifampicin, pyrazinamide, ethambutol, streptomycin, acyclovir; acetyl cysteine, acetylcholine chloride, alatrofloxacin, alendronate, amantadine hydrochloride, ambenomium, amifostine, amiloride hydrochloride, aminocaproic acid, amphiphilicin B, atenolol, atracurium besylate, atropine, azithromycin, aztreonam, bacitracin, becalermin, belladona, bepridil hydrochloride, bleomycin sulfate, calcitonin, calcitonin salmon, carboplatin, capecitabine, capreomycin sulfate, cefamandole nafate, cefazolin sodium, cefepime hydrochloride, cefixime, cefonicid sodium, cefoperazone, cefotetan disodium, cefotoxime, cefoxitin sodium, ceftizoxime, ceftriaxone, cefuroxime axetil, cephalexin, cephapirin sodium, chrionic gonadotropin, cidofovir, cisplatin, cladribine, clidinium bromide, clindamycin, ciprofloxacin, clondronate, colistimethate sodium, deforoxamine, denileukin diftitox, desmopressin, diatrizoate megluamine, dicyclomine, didanosine, dirithromycin, dopamine hydrochloride, dornase alpha doxacurium chloride, doxorubicin, editronate disodium, elanaprilat, enkephalin, enoxacin, enoxaprin sodium, ephedrine, epinephrine, erythromycin, esmol hydrochloride, famiciclovir, fludarabine, fluoxetine, ganciclovir, gentamycin, glucagon, glycopyrolate, heparin sodium, indinavir sulfate, insulin, lamivudine, leucovorin calcium, leuprolide acetate, levofloxacin, lincomycin, lobucavir, lomefloxacin, loracarbef, mannitol, methotrexate, methscopolamine, metformin hydrochloride, metroprolol, mezocillin sodium, mivacurium chloride, nedocromil sodium, neostigmine bromide, neostigmine methyl sulfate, neutontin, norfloxacin, octreotide acetate, ofloxacin, olpadronate, oxytocin, pamidronate disodium, pancuronium bromide, paroxetine, pefloxacin, pentamindine isethionate, pentostatin, pentoxifylline, periciclovir, pentagastrin, phentolamine mesylate, phenylalanine, physostigmine salicylate, piperacillin sodium, polymixin B sulfate, pralidoxine chloride, pramlintide, pregabalin, propofenone, propenthaline bromide, pyridostigmine bromide, residronate, ribavarin, rimantadine hydrochloride, salmetrol xinafoate, sincalide, solatol, somatostatin, sparfloxacin, spectinomycin, stavudine, streptozocin, suxamethonium chloride, tacrine hydrochloride, terbutaline sulfate, thiopbeta ticarcillin, tiludronate, timolol, trandolapril, trimetrexate gluconate, trospectinomycin, trovafloxacin, tubocurarine chloride, valaciclovir, valsartan, vasopressin, vecoronium bromide, vinblastin, vincristine, vinorelbine, warfarin sodium, zalcitabine, zanamavir, zolandronate, zidovudine and the like.
The amphiphilic drug includes but is not limited to amphiphilicin B, bupivacaine, ropivacaine, prilocaine, mepivacaine, tetrocaine, etidocaine, morphine, fentanyl, alfentanil, sulfentanil and the like. †ie solid lipid nanoparticles of the present disclosure contain a drug which contains at least one hydroxy 1 group or at least one hydrophilic linkage.
In the fourth step, a micro emulsion is obtained by adding the solution containing at least one drug obtained in the third step to the lipid melt obtained in the first.
Finally, solid lipid nanoparticles are prepared from the microemulsion.
The microemulsion obtained in the fourth step is dispersed in water maintained at a temperature ranging between 0 °C and 5 °C under continuous stirring/homogenizing. The stirring is carried out at 4,000 to 12,000 rpm for a time period between 20 minutes to 2 hours. The stirring is preferably carried out at 5,000 to 7,000 rpm. The ratio of microemulsion and water ranges between 1 : 1 and 1 :4.9.
The alternative way of obtaining solid lipid nanoparticles from the microemulsion is by purging nitrogen through the microemulsion.
The solid lipid nanoparticles prepared in accordance with the present disclosure have a particle size in the range of 10 nm to lOOOnm.
The preferred particle size of the solid lipid nanoparticles is in the range of 20 nm to 150 nm.
In another aspect of the present disclosure there is provided solid lipid nanoparticles prepared by the process of present disclosure; said solid lipid nanoparticles comprising: i) at least lipid selected from the group consisting of glycerides and fatty acids; ii) at least one drug selected from the group consisting of hydrophilic drugs and amphiphilic drugs; and iii) at least one emulsifier.
The amount of drug is in the range of about 0.001% to about 99%.
The particle size of the solid lipid nanoparticles prepared by the process of present disclosure is in the range of 10 to 1000 nm, preferably between 20-150 nm. Tfie present disclosure is further described in light of the following examples which are set forth for illustration purpose only and not to be construed for limiting the scope of the disclosure.
Example 1: Procedure for preparing the solid lipid nanoparticles containing hydrophilic drug
531.0 gm of polysorbate 80 and 7.9 gm of soy lecithin (phospholipon 90H) 374.0 ml of water were taken in a beaker and heated to 90 °C to obtain an emulsifier mix.
Separately, 79.2 gm of glyceryl behenate was heated at 90 °C to obtain a lipid melt. 7.9 gm of Isoniazid was added as hydrophilic drug to the emulsifier mix at 90 °C to obtain an aqueous solution containing isoniazid. Then, the solution containing isoniazid was added to lipid melt, under continuous magnetic stirring at 90 °C to obtain a clear microemulsion. The 1000.0 gm of microemulsion thus formed, was transferred into 1000.0 gm of cold water maintained at 2°C under continuous mechanical stirring/homogenizing at 5,000 rpm for a time period of about 30 minutes using a Wise Tis Homogenizer.
Example 2: Procedure for preparing the solid lipid nanoparticles containing hydrophilic drug
531.0 gm of polysorbate 80 and 7.9 gm of soy lecithin (phospholipon 90H) 374.0 ml of water were taken in a beaker and heated to 90 °C to obtain an emulsifier mix.
Separately, 79.2 gm of stearic acid was heated at 90 °C to obtain a lipid melt. 7.9 gm of Isoniazid was added as hydrophilic drug to the emulsifier mix at 90 °C to obtain an aqueous solution containing isoniazid. Then, the solution containing isoniazid was added to lipid melt, under continuous magnetic stirring at 90 °C to obtain a clear microemulsion. The 1000.0 gm of microemulsion thus formed, was transferred into 1000.0 gm of cold water maintained at 2°C under continuous mechanical stirring/homogenizing at 5,000 rpm for a time period of about 30 minutes using a Wise Tis Homogenizer.
Example 3: Procedure for preparing the solid lipid nanoparticles containing hydrophilic drug
531.0 gm of polysorbate 80 and 7.9 gm of soy lecithin (phospholipon 90H) 374.0 ml of water were taken in a beaker and heated to 90 °C to obtain an emulsifier mix.
63.36 gm of glyceryl behenate and 15.84 gm of stearic acid were heated at 90 °C to obtain a lipid melt. 7.9 gm of Isoniazid was added as hydrophilic drug to the emulsifier mix at 90 °C to obtain an aqueous solution containing isoniazid. Then, the solution containing isoniazid was added to lipid melt, under continuous magnetic stirring at 90 °C to obtain a clear microemulsion. TTie 1000.0 gm of microemulsion thus formed, was transferred into 1000.0 gm of cold water maintained at 2°C under continuous mechanical stirring/homogenizing at 5,000 rpm for a time period of about 30 minutes using a Wise Tis Homogenizer.
Example 4: Procedure for preparing the solid lipid nanoparticles containing hydrophilic drug
531.0 gm of polysorbate 80 and 7.9 gm of soy lecithin (phospholipon 90H) 374.0 ml of water were taken in a beaker and heated to 90 °C to obtain an emulsifier mix.
Separately,63.36 gm of glyceryl behenate and 15.84 gm of stearic acid was heated at 90 °C to obtain a lipid melt. 7.9 gm of Isoniazid was added as hydrophilic drug to the emulsifier mix at 90 °C to obtain an aqueous solution containing isoniazid. Then, the solution containing isoniazid was added to lipid melt, under continuous magnetic stirring at 90 °C to obtain a clear microemulsion. The dry nitrogen gas was purged through the microemulsion for 2 hours to obtain the solid lipid nanoparticles.
The solid lipid nanoparticles prepared in accordance with the present disclosure were evaluated for various parameters such as total drug content, entrapment efficiency, particle size, zeta potential, poly dispersity index, in-vitro release (figure 1). The results are provided in Table 1.
Determination of particle size, poly dispersity index and zeta potential:
Solid lipid nanoparticles of Example 1 were evaluated for particle size, poly dispersity index and zeta potential using Beckman Coulter Delsa(TM) Nano Zeta Potential and Submicron Particle Size Analyzer wherein triple distilled water (TDW) was used as a diluting medium.
Determination of total drug content (TDC) and entrapment efficiency (EE):
Total amount of drug per unit volume present in the solid lipid nanoparticles (SLNs) was determined by disrupting 0.1 ml of the solid lipid nano-particle dispersion with 5 ml of chloroform and methanol (1 : 1). The amount of isoniazid was determined by HPLC. Each experiment was performed in triplicate. A similarly processed blank solid lipid nano- particle sample was taken as control. The total drug content was determined by using the equation given below.
^ _ Calculated amount of drug / ml of SLN dispersion 1 ΛΛ
TDC = - x 100
Total amount of drug added / ml of SLN dispersion Entrapment efficiency was determined by analyzing the clear supernatant obtained by centrifuging the developed solid lipid nanoparticle dispersions at 8.02 Lac g for 2 h at 4°C using Beckman Coulter Ultracentrifuge (LlOO K and 100 Ti rotor). The entrapment efficiency was calculated as follows:
TDC - Df
EE = ^ xl OO
TDC
Where Df = amount of drug in clear supernatant fluid
The results are provided herein below.
Table No. 1: Determination of particle size (D), poly dispersity index (PDI), total drug content (TDC), entrapment efficiency (EE) and zeta potential of the solid lipid nano particles containing isoniazid drug.
Figure imgf000016_0001
Conclusion:
Solid lipid nanoparticles prepared in accordance with the present disclosure exhibited a small particle size below 120 nm. The total drug content was found to be greater than 92% w/v. Further, the poly dispersity index for all the developed solid lipid nanoparticles was found to be below 0.3 which indicates a narrow particle size distribution.
The entrapment efficiency for solid lipid nanoparticles of combination of glyceryl behenate and stearic acid was found to be 84.032±1.103% in comparison to 77.600± 1.961% and 69.013±0.675% for solid lipid nanoparticles of glyceryl behenate and solid lipid nanoparticles of stearic acid, respectively.
From these results it is clear that the solid lipid nanoparticles prepared using a combination of lipids or a lipid and a fatty acid prepared in accordance with the present disclosure possesses enhanced entrapment efficiency for hydrophilic and/or amphiphilic drugs.
In vitro drug release of isoniazid
In vitro release of isoniazid was determined in phosphate buffer of pH 6.8 by dialysis bag method using dialysis membrane with a molecular weight cut off of 12000-14000 Da. An accurate volume (1 ml) of solid lipid nanoparticles dispersion containing 2.6 mg of isoniazid (as per the calculated Total Drug Content) was placed inside the dialysis bag, tied at both the ends and dipped in the dissolution medium (80 ml). Stirring was maintained at 100 rpm, using magnetic bead, at 37±0.2 °C. Two milliliters aliquots were withdrawn at pre-set time intervals (0.25, 0.5, 1, 2, 3, 4, 6, 8, 12, 24 h) and replaced by an equal volume of fresh dissolution medium. After suitable dilution, the samples were analyzed by HPLC. Corrected concentration of isoniazid was calculated in the test samples using the regression equation of the calibration curve. The results are show in Table No. 2 and Figure l(Free drug *, SLN of stearic acid**, SLN of glyceryl behenate*** and SLN of combination of glyceryl behenate and stearic acid****)
T¾ble No. 2: In-vitro drug release of isoniazid from Solid Lipid Nanoparticles of the present disclosure.
Figure imgf000018_0001
Conclusion:
The drug release study was carried out for all the three developed solid lipid nanoparticles (solid lipid nanoparticles of glyceryl behenate, solid lipid nanoparticles of stearic acid and solid lipid nanoparticles of combination of glyceryl behenate and stearic acid). In vitro drug release showed tri-phasic behavior comprising an initial release of free drug (the free drug was not removed from the prepared solid lipid nanoparticle dispersions which were loaded into the dialysis bag), this phase could be due to the very small solid lipid nanoparticles (SLNs) present in the dispersion, across the dialysis membrane followed by a hump and a fast release probably corresponding to the release of drug present in the outer surfactant coat of the SLNs, considering that isoniazid is a hydrophilic drug. This was followed by a delayed release phase starting approximately at 5 h showing almost a controlled release. The release kinetics followed peppas model (r > 0.9) and 65.086 %, 79.967% and 86.756% of the drug was released in a period of 24 h ίόι solid lipid nanoparticles of glyceryl behenate, , solid lipid nanoparticles of stearic acid and solid lipid nanoparticles of combination of glyceryl behenate and stearic acid respectively.
Determination of drug concentration in plasma, brain, liver and kidney:
Two groups each containing forty two female Wistar rats were formed and named as Group A and Group B. Both the groups of rats were kept at uniform atmospheric conditions. The Wistar rats of Group A were given isoniazid (free drug) by an oral route while rats of Group B were given solid lipid nanoparticles of combination of glyceryl behenate and stearic acid containing isoniazid drug by an oral route. Drug concentration in the samples was determined using HPLC. The plasma samples were collected before sacrificing the rats. Six Wistar rats were sacrificed per time point. The details are mentioned below.
Preparation of plasma samples:
To 150-μΤ aliquot of plasma, a 300 μΐ, of the deproteinizing agent (methanol) was added to precipitate the proteins which was then vortexed for 2 minutes. The samples were centrifuged at 15000 rpm for 10 minutes at 4 °C. The supernatant was then collected and an equal volume of water was added to the clear supernatant. The samples were filtered (0.20 μπι nylon filters) and injected into the HPLC system.
Preparation of tissue samples (Brain, Liver and Kidney)
20 % aqueous tissue homogenates were prepared in cold 150 mM KC1. The homogenates were then centrifuged at 15000 rpm for 10 minutes at 4 °C to obtain clear tissue homogenates. To 150-μΤ aliquot of the clear tissue homogenates, a 300 μΐ. of the deproteinizing agent (methanol) was added to obtain a dispersion which was vortexed for 2 minutes. The samples were then centrifuged at 15000 rpm for 10 minutes at 4 °C. The supernatant was then collected and an equal volume of water was added to the clear supernatant. The samples were then filtered (0.20 μπι nylon filters) and were injected into the HPLC system.
Chromatographic conditions
The HPLC system consisted of Waters Alliance (Waters Corporation, MA, USA) equipped with a Waters e-2695 ALLIANCE separation module comprising of a solvent (quaternary gradient mode), auto injector, column oven and a 4 channel in line degasser, a sample management system (sample heater cooler) and a 2998 PDA detector. Chromatographic separation was performed using symmetry CI 8 column at 254 nm. Data acquisition was performed by the Empower 2® software. The mobile phase consisted of a mixture phosphate buffer of pH 6.8 and methanol (85: 15). The mobile phase was delivered at a flow rate of 0.9 ml/min and the detection of isoniazid (INH) was carried out at 254 nm. The injection volume was 20 μΐ, and the analysis was performed at 30°C (sample 10°C).
The results are tabulated herein Table No. 3 to Table No. 6.
Table No. 3: Comparative analysis of concentration of isoniazid in plasma.
Figure imgf000020_0001
Table No. 4: Comparative analysis of concentration of isoniazid in brain (cerebral permeability)
Figure imgf000021_0001
I able No. 5: Comparative analysis of concentration isoniazid in liver
Figure imgf000022_0001
liable No. 6: Comparative analysis of concentration of isoniazid in kidney
Figure imgf000023_0001
Conclusion:
From the results as shown in Table No. 3 and Table No. 4 it is found that:
- The solid lipid nanoparticles with a particle size in the range of 10 to 1000 nm, preferably between 20-150 nm prepared in accordance with the present disclosure provide enhanced concentration of hydrophilic drug or amphiphilic drug in plasma. - The solid lipid nanoparticles prepared in accordance with the present disclosure improves cerebral permeability of hydrophilic drug or amphiphilic drug entrapped therein.
TECHNICAL ADVANCEMENT AND ECONOMIC SIGNIFICANCE
The solid lipid nanoparticles prepared in accordance with the present disclosure enhance the bioavailability of the hydrophilic drug or amphiphilic drug as a result of improved permeability across the biological barriers. The improved bioavailability reduces dose, dosing frequency and dose dependent side effects of the drug.
Throughout this specification the word "comprise", or variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated element, integer or step, or group of elements, integers or steps, but not the exclusion of any other element, integer or step, or group of elements, integers or steps.
The use of the expression "at least" or "at least one" suggests the use of one or more elements or ingredients or quantities, as the use may be in the embodiment of ; the invention to achieve one or more of the desired objects or results.
The numerical values mentioned for the various physical parameters, dimensions or quantities are only approximations and it is envisaged that the values higher/lower than the numerical values assigned to the parameters, dimensions or quantities fall within the scope of the invention, unless there is a statement in the specification specific to the contrary.
The applicant craves leave to submit further clinical data.
While considerable emphasis has been placed herein on the particular features of this disclosure, it will be appreciated that various modifications can be made, and that many changes can be made in the preferred embodiments without departing from the principle of the disclosure. These and other modifications in the nature of the disclosure or the preferred embodiments will be apparent to those skilled in the art from the disclosure herein, whereby it is to be distinctly understood that the foregoing descriptive matter is to be interpreted merely as illustrative of the disclosure and not as a limitation.

Claims

A process for preparing solid lipid nanoparticles, said process comprising the following steps: i) melting at least one lipid selected from the group consisting of glycerides and fatty acids; ii) preparing an aqueous emulsifler mix by admixing at least one emulsifier and water followed by heating at a temperature atleast equal to the melting point of the selected lipid; iii) adding at least one drug to the aqueous emulsifier mix to obtain a dispersion; iv) adding the dispersion containing at least one drug to the lipid melt to obtain a microemulsion; and v) preparing solid lipid nanoparticles having a particle size in the range of 10 nm to 1000 nm, preferably 20 nm to 150 nm from the microemulsion.
The process claimed in claim 1, wherein the method step of preparing solid lipid nanoparticles comprises dispersing the microemulsion in water maintained at a temperature ranging between 0 °C and 5 °C under continuous stirring/homogenizing to obtain solid lipid nanoparticles, wherein the ratio of microemulsion and water ranges between 1 : 1 and 1 :4.9.
The process claimed in claim 1 , wherein the method step of preparing the solid lipid nanoparticles comprises purging nitrogen through the microemulsion to obtain solid lipid nanoparticles.
The process claimed in claim 1, wherein the glyceride is at least one selected from the group consisting of mono-glycerides, di-glycerides and tri-glycerides.
The process claimed in claim 1 , wherein the glyceride is at least one selected from the group consisting of glyceryl behenate tricaprin, trilaurin, trimyristin, tripalmitin, tristearin, l,2-dioctanoyl-^«-glycerol, l,2-didecanoyl-.y«-glycerol, 1 ,2-dilauroyl-^- glycerol, 1 ^-dimyristoyl-s -glycerol, 1 ,2-dipalmitoyl-.s«-glycerol, l-palmitoyl-2- oleoyl-src-glycerol, 1 -stearoyl-2-linoleoyl-.s«-glycerol, 1 -stearoyl-2-arachidonoyl-5«- glycerol, 1 -stearoyl-2-docosahexaenoyl-sft-glycerol, 1 -oleoyl-2-acetyl-5,«-glycerol, l,2-di-O-phytanyl-,s«-glycerol, 1 ,2-dipalmitoyl ethylene glycol, 1-2-dioleoyl ethylene glycol, glyceryl monostearate, behenoyl polyoxyl-8 glycerides, glyceryl palmitostearate, 1-O-hexadecyl-^-glycerol, l-O-hexadecyl-2-acetyl-sn-glycerol, 1- O-hexadecyl-2-O-methyl-5,n-glycerol, l,2-diacyl-3-O-(a-D-glucopyranosyl)-s«- glycerol, stearoyl macrogol-32 glycerides, stearoyi polyoxyl-32 glycerides, lauroyl macrogol-32 glycerides, lauroyl polyoxyl-32 glycerides, lauroyl macrogol-6 glycerides, lauroyl polyoxyl-6 glycerides, oleoyl macrogol-6 glycerides, oleoyl polyoxyl-6 glycerides, linoleoyl macrogol-6 glycerides, polyglyceryl-3 dioleate, glycerol monolinoleate, glyceryl monolinoleate, glycerol monooleates, diethylene glycol monoethyl ether, glyceryl dibehenate, glycerol distearate, glyceryl distearate, glyceryl dipalmitostearate and linoleoyl polyoxyl-6 glyceride, preferably, glyceryl behenate.
6. The process claimed in claim 1, wherein the fatty acid is selected from the group consisting of saturated C4-C28 fatty acids and unsaturated C4-C28 fatty acids.
7. The process claimed in claim 1, wherein the fatty acid is stearic acid.
8. The process claimed in claim 1, wherein the proportion of the glyceride and the fatty acid is in the range of 1 :0 to 1 :20.
9. The process claimed in claim 1, wherein the proportion of the fatty acid and the glyceride is in the range of 1 :0 to 1 :20.
10. The process claimed in claim 1, wherein the emulsifier is at least one selected from the group consisting of anionic emulsifiers, cationic emulsifiers, non ionic
emulsifiers or zwitterionic emulsifiers.
1 1. The process claimed in claim 1 , wherein the emulsifier is at least one selected from the group consisting of soy lecithin, egg lecithin, phosphatidylcholine; ethylene oxide copolymers, propylene oxide copolymers, poloxamers, sorbitan ethylene oxide/propylene oxide copolymers, polysorbate 20, polysorbate 60, polysorbate 80, sorbitan esters, span 20, span 40, span 60, span 80, alkyllaryl polyether alcohol polymers, tyloxapol, bile salts, cholate, glycocholate, taurocholate, taurodeoxycholate, gemini surfactants and alcohols.
12. The process claimed in claim 1, wherein the drug is at least one selected from the group consisting of hydrophilic drugs and amphiphilic drugs. if. The process claimed in claim 1, wherein the drug is at least one hydrophilic drug selected from the group consisting of isoniazid, rifampicin, pyrazinamide, ethambutol, streptomycin, acyclovir, acetyl cysteine, acetylcholine chloride, alatrofloxacin, alendronate, amantadine hydrochloride, ambenomium, amifostine, amiloride hydrochloride, aminocaproic acid, amphiphilicin B, atenolol, atracurium besylate, atropine, azithromycin, aztreonam, bacitracin, becalermin, belladona, bepridil hydrochloride, bleomycin sulfate, calcitonin, calcitonin salmon, carboplatin, capecitabine, capreomycin sulfate, cefamandole nafate, cefazolin sodium, cefepime hydrochloride, cefixime, cefonicid sodium, cefoperazone, cefotetan disodium, cefotoxime, cefoxitin sodium, ceftizoxime, ceftriaxone, cefuroxime axetil, cephalexin, cephapirin sodium, chrionic gonadotropin, cidofovir, cisplatin, cladribine, clidinium bromide, clindamycin, ciprofloxacin, clondronate, colistimethate sodium, deforoxamine, denileukin diftitox, desmopressin, diatrizoate megluamine, dicyclomine, didanosine, dirithromycin, dopamine hydrochloride, dornase alpha doxacurium chloride, doxorubicin, editronate disodium, elanaprilat, enkephalin, enoxacin, enoxaprin sodium, ephedrine, epinephrine, erythromycin, esmol hydrochloride, famiciclovir, fludarabine, fluoxetine, ganciclovir, gentamycin, glucagon, glycopyrolate, heparin sodium, indinavir sulfate, insulin, lamivudine, leucovorin calcium, leuprolide acetate, levofloxacin, lincomycin, lobucavir, lomefloxacin, loracarbef, mannitol, methotrexate, methscopolamine, metformin hydrochloride, metroprolol, mezocillin sodium, mivacurium chloride, nedocromil sodium, neostigmine bromide, neostigmine methyl sulfate, neutontin, norfloxacin, octreotide acetate, ofloxacin, olpadronate, oxytocin, pamidronate disodium, pancuronium bromide, paroxetine, pefloxacin, pentamindine isethionate, pentostatin, pentoxifylline, periciclovir, pentagastrin, phentolamine mesylate, phenylalanine, physostigmine salicylate, piperacillin sodium, polymixin B sulfate, pralidoxine chloride, pramlintide, pregabalin, propofenone, propenthaline bromide, pyridostigmine bromide, residronate, ribavarin, rimantadine hydrochloride, salmetrol xinafoate, sincalide, solatol, somatostatin, sparfloxacin, spectinomycin, stavudine, streptozocin, suxamethonium chloride, tacrine hydrochloride, terbutaline sulfate, thiopbeta ticarcillin, tiludronate, timolol, trandolapril, trimetrexate gluconate, trospectinomycin, trovafloxacin, tubocurarine chloride, valaciclovir, valsartan, vasopressin, vecoronium bromide, vinblastin, vincristine, vinorelbine, warfarin sodium, zalcitabine, zanamavir, zolandronate and zidovudine. l The process claimed in claim 1, wherein the drug is at least one amphiphilic drug selected from the group consisting of amphiphilicin B, bupivacaine, ropivacaine, prilocaine, mepivacaine, tetrocaine, etidocaine, morphine, fentanyl, alfentanil and sulfentanil.
15. The process claimed in claim 1, wherein the drug is at least one drug containing at least one hydroxyl group or at least one hydrophilic linkage.
16. The process claimed in claim 2, wherein the method step of dispersing the micro emulsion in water is carried out by continuous stirring/homogenizing at 4,000 to 12,000 rpm, preferably 5,000 to 7,000 rpm for a time period ranging between 20 minutes and 2 hours.
17. Solid lipid nanoparticles prepared by the process as claimed in any of the preceding claims, said solid lipid nanoparticles comprising: i) at least one lipid selected from the group consisting of glycerides and fatty acids; ii) at least one drug selected from the group consisting of hydrophilic drugs and amphiphilic drugs; and iii) at least one emulsifier.
18. The solid lipid nanoparticles claimed in claim 17, wherein the glyceride is at least one selected from the group consisting of mono-glycerides, di-glycerides and triglycerides.
19. The solid lipid nanoparticles claimed in claim 17, wherein the glyceride is at least one selected from the group consisting of glyceryl behenate, tricaprin, trilaurin, trimyristin, tripalmitin, tristearin, l,2-dioctanoyl-5«-glycerol, 1 ,2-didecanoyl-.s«- glycerol, 1 ,2-dilauroyl-5«-glycerol, l,2-dimyristoyl-5«-glycerol, 1,2-dipalmitoyl-^- glycerol, l-palmitoyl-2-oleoyl-5«-glycerol, l-stearoyl-2-linoleoyl-s«-glycerol, 1- stearoyl-2-arachidonoyl-5,«-glycerol, 1 -stearoyl-2-docosahexaenoyl-5«-glycerol, 1 - oleoyl-2-acetyl-5«-glycerol, l,2-di-O-phytanyl->s«-glycerol, 1,2-dipalmitoyl ethylene glycol, 1-2-dioleoyl ethylene glycol, glyceryl monostearate, behenoyl polyoxyl-8 glycerides, glyceryl palmitostearate, l-O-hexadecyl-s -glycerol, 1 -O-hexadecyl-2- acetyl-572-glycerol, 1 -O-hexadecyl-2-O-methyl-in-glycerol, 1 ,2-diacyl-3-O-(a-D- glucopyranosyl)-sft-glycerol, stearoyl macrogol-32 glycerides, stearoyl polyoxyl-32 glycerides, lauroyl macrogol-32 glycerides, lauroyl polyoxyl-32 glycerides, lauroyl macrogol-6 glycerides, lauroyl polyoxyl-6 glycerides, oleoyl macrogol-6 glycerides, oleoyl polyoxyl-6 glycerides, linoleoyl macrogol-6 glycerides, polyglyceryl-3 dioleate, glycerol monolinoleate, glyceryl monolinoleate, glycerol monooleates, diethylene glycol monoethyl ether, glyceryl dibehenate, glycerol distearate, glyceryl distearate, glyceryl dipalmitostearate and linoleoyl polyoxyl-6 glyceride, preferably, glyceryl behenate.
20. The solid lipid nanoparticles claimed in claim 17, wherein the fatty acid is at least one selected from the group consisting of saturated C4-C28 fatty acids and unsaturated C4-C28 fatty acids.
21. The solid lipid nanoparticles claimed in claim 17, wherein the fatty acid is stearic acid.
22. The process claimed in claim 17, wherein the proportion of the glyceride and the fatty acid is in the range of 1 :0 to 1 :20.
23. The process claimed in claim 17, wherein the proportion of the fatty acid and the glyceride is in the range of 1 :0 to 1 :20.
24. The solid lipid nanoparticles claimed in claim 17, wherein the drug is at least one hydrophilic drug selected from the group consisting of isoniazid, rifampicin, pyrazinamide, ethambutol, streptomycin, acyclovir; acetyl cysteine, acetylcholine chloride, alatrofloxacin, alendronate, amantadine hydrochloride, ambenomium, amifostine, amiloride hydrochloride, aminocaproic acid, amphiphilicin B, atenolol, atracurium besylate, atropine, azithromycin, aztreonam, bacitracin, becalermin, belladona, bepridil hydrochloride, bleomycin sulfate, calcitonin, calcitonin salmon, carboplatin, capecitabine, capreomycin sulfate, cefamandole nafate, cefazolin sodium, cefepime hydrochloride, cefixime, cefonicid sodium, cefoperazone, cefotetan disodium, cefotoxime, cefoxitin sodium, ceftizoxime, ceftriaxone, cefuroxime axetil, cephalexin, cephapirin sodium, chrionic gonadotropin, cidofovir, cisplatin, cladribine, clidinium bromide, clindamycin, ciprofloxacin, clondronate, colistimethate sodium, deforoxamine, denileukin diftitox, desmopressin, diatrizoate megluamine, dicyclomine, didanosine, dirithromycin, dopamine hydrochloride, dornase alpha doxacurium chloride, doxorubicin, editronate disodium, elanaprilat, enkephalin, enoxacin, enoxaprin sodium, ephedrine, epinephrine, erythromycin, esmol hydrochloride, famiciclovir, fludarabine, fluoxetine, ganciclovir, gentamycin, glucagon, glycopyrolate, heparin sodium, indinavir sulfate, insulin, lamivudine, leucovorin calcium, leuprolide acetate, levofloxacin, lincomycin, lobucavir, lomefloxacin, loracarbef, mannitol, methotrexate, methscopolamine, metformin hydrochloride, metroprolol, mezocillin sodium, mivacurium chloride, nedocromil sodium, neostigmine bromide, neostigmine methyl sulfate, neutontin, norfloxacin, octreotide acetate, ofloxacin, olpadronate, oxytocin, pamidronate disodium, pancuronium bromide, paroxetine, pefloxacin, pentamindine isethionate, pentostatin, pentoxifylline, periciclovir, pentagastrin, phentolamine mesylate, phenylalanine, physostigmine salicylate, piperacillin sodium, polymixin B sulfate, pralidoxine chloride, pramlintide, pregabalin, propofenone, propenthaline bromide, pyridostigmine bromide, residronate, ribavarin, rimantadine hydrochloride, salmetrol xinafoate, sincalide, solatol, somatostatin, sparfloxacin, spectinomycin, stavudine, streptozocin, suxamethonium chloride, tacrine hydrochloride, terbutaline sulfate, thiopbeta ticarcillin, tiludronate, timolol, trandolapril, trimetrexate gluconate, trospectinomycin, trovafloxacin, tubocurarine chloride, valaciclovir, valsartan, vasopressin, vecoronium bromide, vinblastin, vincristine, vinorelbine, warfarin sodium, zalcitabine, zanamavir, zolandronate and zidovudine.
25. The solid lipid nanoparticles claimed in claim 17, wherein the drug is at least one amphiphilic drug selected from the group consisting of amphiphilicin B, bupivacaine, ropivacaine, prilocaine, mepivacaine, tetrocaine, etidocaine, morphine, fentanyl, alfentanil and sulfentanil.
26. The solid" lipid nanoparticles claimed in claim 17, wherein the amount of drug is in the range of about 0.001% to about 99% with respect to the mass of the lipid.
27. The solid lipid nanoparticles claimed in claim 17, wherein the drug is at least one drug containing at least one hydroxyl group or at least one hydrophilic linkage.
28. The solid lipid nanoparticles claimed in any of the claims 17 to 27, wherein the particle size of the solid lipid nanoparticles ranges between 10 to 1000 nm, preferably between 20- 150 nm.
29. The solid lipid nanoparticles claimed in claim 17, wherein the emulsifier is at least one selected from the group consisting of anionic emulsiflers, cationic emulsifiers, non ionic emulsifiers or zwitterionic emulsifiers. The solid lipid nanoparticles claimed in claim 17, wherein the emulsifier is at least one selected from the group consisting of soy lecithin, egg lecithin, phosphatidylcholine; ethylene oxide copolymers, propylene oxide copolymers, poloxamers, sorbitan ethylene oxide/propylene oxide copolymers, polysorbate 20, polysorbate 60, polysorbate 80, sorbitan esters, span 20, span 40, span 60, span 80, alkyllaryl polyether alcohol polymers, tyloxapol, bile salts, cholate, glycocholate, taurocholate, taurodeoxy cholate, gemini surfactants and alcohols.
PCT/IN2012/000154 2012-01-13 2012-03-05 Solid lipid nanoparticles entrapping hydrophilic/ amphiphilic drug and a process for preparing the same WO2013105101A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
IN127DE2012 2012-01-13
IN127/DEL/2012 2012-01-13

Publications (2)

Publication Number Publication Date
WO2013105101A1 true WO2013105101A1 (en) 2013-07-18
WO2013105101A8 WO2013105101A8 (en) 2014-01-30

Family

ID=48781104

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IN2012/000154 WO2013105101A1 (en) 2012-01-13 2012-03-05 Solid lipid nanoparticles entrapping hydrophilic/ amphiphilic drug and a process for preparing the same

Country Status (1)

Country Link
WO (1) WO2013105101A1 (en)

Cited By (60)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103816111A (en) * 2014-02-26 2014-05-28 中国人民解放军第二军医大学 Ropivacaine nanometer lipid carrier temperature-sensitive in-situ gel and preparation method thereof
WO2014152211A1 (en) 2013-03-14 2014-09-25 Moderna Therapeutics, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
WO2015034925A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Circular polynucleotides
WO2015034928A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Chimeric polynucleotides
WO2015051214A1 (en) 2013-10-03 2015-04-09 Moderna Therapeutics, Inc. Polynucleotides encoding low density lipoprotein receptor
WO2015057751A1 (en) * 2013-10-14 2015-04-23 Nanosphere Health Sciences, Llc Nanoparticle compositions and methods as carriers of nutraceutical factors across cell membranes and biological barriers
WO2016014846A1 (en) 2014-07-23 2016-01-28 Moderna Therapeutics, Inc. Modified polynucleotides for the production of intrabodies
WO2017112943A1 (en) 2015-12-23 2017-06-29 Modernatx, Inc. Methods of using ox40 ligand encoding polynucleotides
WO2017120612A1 (en) 2016-01-10 2017-07-13 Modernatx, Inc. Therapeutic mrnas encoding anti ctla-4 antibodies
US10028919B2 (en) 2015-03-10 2018-07-24 Nanosphere Health Sciences, Llc Lipid nanoparticle compositions and methods as carriers of cannabinoids in standardized precision-metered dosage forms
CN108484898A (en) * 2018-04-20 2018-09-04 江苏保易制药有限公司 The preparation method of polysorbate without polyoxyethylene isobide aliphatic ester
WO2018213789A1 (en) 2017-05-18 2018-11-22 Modernatx, Inc. Modified messenger rna comprising functional rna elements
WO2018213731A1 (en) 2017-05-18 2018-11-22 Modernatx, Inc. Polynucleotides encoding tethered interleukin-12 (il12) polypeptides and uses thereof
WO2018232006A1 (en) 2017-06-14 2018-12-20 Modernatx, Inc. Polynucleotides encoding coagulation factor viii
CN109260181A (en) * 2017-07-17 2019-01-25 北京盈科瑞创新药物研究有限公司 Heparin nebulization sucking pharmaceutical solutions and preparation method thereof
WO2019104160A2 (en) 2017-11-22 2019-05-31 Modernatx, Inc. Polynucleotides encoding phenylalanine hydroxylase for the treatment of phenylketonuria
WO2019104195A1 (en) 2017-11-22 2019-05-31 Modernatx, Inc. Polynucleotides encoding propionyl-coa carboxylase alpha and beta subunits for the treatment of propionic acidemia
WO2019104152A1 (en) 2017-11-22 2019-05-31 Modernatx, Inc. Polynucleotides encoding ornithine transcarbamylase for the treatment of urea cycle disorders
WO2019116062A1 (en) 2017-12-12 2019-06-20 Lead Biotherapeutics Ltd. Solid lipid nanoparticle for intracellular release of active substances and method for production the same
WO2019136241A1 (en) 2018-01-05 2019-07-11 Modernatx, Inc. Polynucleotides encoding anti-chikungunya virus antibodies
WO2019200171A1 (en) 2018-04-11 2019-10-17 Modernatx, Inc. Messenger rna comprising functional rna elements
WO2019226650A1 (en) 2018-05-23 2019-11-28 Modernatx, Inc. Delivery of dna
WO2020023390A1 (en) 2018-07-25 2020-01-30 Modernatx, Inc. Mrna based enzyme replacement therapy combined with a pharmacological chaperone for the treatment of lysosomal storage disorders
WO2020047201A1 (en) 2018-09-02 2020-03-05 Modernatx, Inc. Polynucleotides encoding very long-chain acyl-coa dehydrogenase for the treatment of very long-chain acyl-coa dehydrogenase deficiency
WO2020056161A1 (en) 2018-09-12 2020-03-19 University Of Florida Research Foundation, Inc. Slow-cycling cell-rna based nanoparticle vaccine to treat cancer
WO2020056155A2 (en) 2018-09-13 2020-03-19 Modernatx, Inc. Polynucleotides encoding branched-chain alpha-ketoacid dehydrogenase complex e1-alpha, e1-beta, and e2 subunits for the treatment of maple syrup urine disease
WO2020056147A2 (en) 2018-09-13 2020-03-19 Modernatx, Inc. Polynucleotides encoding glucose-6-phosphatase for the treatment of glycogen storage disease
WO2020056239A1 (en) 2018-09-14 2020-03-19 Modernatx, Inc. Polynucleotides encoding uridine diphosphate glycosyltransferase 1 family, polypeptide a1 for the treatment of crigler-najjar syndrome
WO2020069169A1 (en) 2018-09-27 2020-04-02 Modernatx, Inc. Polynucleotides encoding arginase 1 for the treatment of arginase deficiency
WO2020097409A2 (en) 2018-11-08 2020-05-14 Modernatx, Inc. Use of mrna encoding ox40l to treat cancer in human patients
WO2020227642A1 (en) 2019-05-08 2020-11-12 Modernatx, Inc. Compositions for skin and wounds and methods of use thereof
WO2020227615A1 (en) 2019-05-08 2020-11-12 Modernatx, Inc. Polynucleotides encoding methylmalonyl-coa mutase for the treatment of methylmalonic acidemia
CN112120228A (en) * 2020-08-28 2020-12-25 盐城工学院 Solid lipid particle embedded with feruloyl oligosaccharide, preparation method thereof and application of solid lipid particle as food additive
WO2020263883A1 (en) 2019-06-24 2020-12-30 Modernatx, Inc. Endonuclease-resistant messenger rna and uses thereof
WO2020263985A1 (en) 2019-06-24 2020-12-30 Modernatx, Inc. Messenger rna comprising functional rna elements and uses thereof
WO2021061707A1 (en) 2019-09-23 2021-04-01 Omega Therapeutics, Inc. Compositions and methods for modulating apolipoprotein b (apob) gene expression
WO2021061815A1 (en) 2019-09-23 2021-04-01 Omega Therapeutics, Inc. COMPOSITIONS AND METHODS FOR MODULATING HEPATOCYTE NUCLEAR FACTOR 4-ALPHA (HNF4α) GENE EXPRESSION
WO2021183720A1 (en) 2020-03-11 2021-09-16 Omega Therapeutics, Inc. Compositions and methods for modulating forkhead box p3 (foxp3) gene expression
WO2021247507A1 (en) 2020-06-01 2021-12-09 Modernatx, Inc. Phenylalanine hydroxylase variants and uses thereof
US11260033B2 (en) 2018-12-11 2022-03-01 Disruption Labs Inc. Compositions for the delivery of therapeutic agents and methods of use and making thereof
WO2022104131A1 (en) 2020-11-13 2022-05-19 Modernatx, Inc. Polynucleotides encoding cystic fibrosis transmembrane conductance regulator for the treatment of cystic fibrosis
EP4011451A1 (en) 2015-10-22 2022-06-15 ModernaTX, Inc. Metapneumovirus mrna vaccines
CN114931551A (en) * 2022-05-11 2022-08-23 四川科瑞德制药股份有限公司 Milakammonium chloride injection with storage stability at 25 ℃, preparation method and application thereof
WO2022204390A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding phenylalanine hydroxylase and uses thereof
WO2022204380A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding propionyl-coa carboxylase alpha and beta subunits and uses thereof
WO2022204371A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding glucose-6-phosphatase and uses thereof
WO2022204370A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles and polynucleotides encoding ornithine transcarbamylase for the treatment of ornithine transcarbamylase deficiency
WO2022204369A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Polynucleotides encoding methylmalonyl-coa mutase for the treatment of methylmalonic acidemia
WO2022266083A2 (en) 2021-06-15 2022-12-22 Modernatx, Inc. Engineered polynucleotides for cell-type or microenvironment-specific expression
WO2022271776A1 (en) 2021-06-22 2022-12-29 Modernatx, Inc. Polynucleotides encoding uridine diphosphate glycosyltransferase 1 family, polypeptide a1 for the treatment of crigler-najjar syndrome
WO2023283359A2 (en) 2021-07-07 2023-01-12 Omega Therapeutics, Inc. Compositions and methods for modulating secreted frizzled receptor protein 1 (sfrp1) gene expression
EP4159741A1 (en) 2014-07-16 2023-04-05 ModernaTX, Inc. Method for producing a chimeric polynucleotide encoding a polypeptide having a triazole-containing internucleotide linkage
WO2023056044A1 (en) 2021-10-01 2023-04-06 Modernatx, Inc. Polynucleotides encoding relaxin for the treatment of fibrosis and/or cardiovascular disease
CZ309587B6 (en) * 2021-01-22 2023-05-03 Oncora S.R.O. Microemulsion preconcentrate containing cladribine and preparing it
US11707436B2 (en) 2014-12-15 2023-07-25 Nanosphere Health Sciences Inc. Methods of treating inflammatory disorders and global inflammation with compositions comprising phospholipid nanoparticle encapsulations of NSAIDS
WO2023161350A1 (en) 2022-02-24 2023-08-31 Io Biotech Aps Nucleotide delivery of cancer therapy
WO2023183909A2 (en) 2022-03-25 2023-09-28 Modernatx, Inc. Polynucleotides encoding fanconi anemia, complementation group proteins for the treatment of fanconi anemia
WO2023196399A1 (en) 2022-04-06 2023-10-12 Modernatx, Inc. Lipid nanoparticles and polynucleotides encoding argininosuccinate lyase for the treatment of argininosuccinic aciduria
WO2023215498A2 (en) 2022-05-05 2023-11-09 Modernatx, Inc. Compositions and methods for cd28 antagonism
WO2024026254A1 (en) 2022-07-26 2024-02-01 Modernatx, Inc. Engineered polynucleotides for temporal control of expression

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104306389A (en) * 2014-10-20 2015-01-28 河南牧翔动物药业有限公司 Lincomycin-spectinomycin compound nano-emulsion

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1490055A (en) * 2003-08-28 2004-04-21 东南大学 Method for preparing nanometre structure medicine
CN1621029A (en) * 2004-10-14 2005-06-01 东南大学 Method for preparing ten nanometers level solid lipid nanometer particle
WO2007000531A2 (en) * 2005-05-16 2007-01-04 Universite Claude Bernard Lyon I Method for preparing solid lipidic particles using a membrane reactor

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1490055A (en) * 2003-08-28 2004-04-21 东南大学 Method for preparing nanometre structure medicine
CN1621029A (en) * 2004-10-14 2005-06-01 东南大学 Method for preparing ten nanometers level solid lipid nanometer particle
WO2007000531A2 (en) * 2005-05-16 2007-01-04 Universite Claude Bernard Lyon I Method for preparing solid lipidic particles using a membrane reactor

Cited By (68)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014152211A1 (en) 2013-03-14 2014-09-25 Moderna Therapeutics, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
WO2015034925A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Circular polynucleotides
WO2015034928A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Chimeric polynucleotides
WO2015051214A1 (en) 2013-10-03 2015-04-09 Moderna Therapeutics, Inc. Polynucleotides encoding low density lipoprotein receptor
WO2015057751A1 (en) * 2013-10-14 2015-04-23 Nanosphere Health Sciences, Llc Nanoparticle compositions and methods as carriers of nutraceutical factors across cell membranes and biological barriers
US9925149B2 (en) 2013-10-14 2018-03-27 Nanosphere Health Sciences, Llc Nanoparticle compositions and methods as carriers of nutraceutical factors across cell membranes and biological barriers
CN103816111A (en) * 2014-02-26 2014-05-28 中国人民解放军第二军医大学 Ropivacaine nanometer lipid carrier temperature-sensitive in-situ gel and preparation method thereof
EP4159741A1 (en) 2014-07-16 2023-04-05 ModernaTX, Inc. Method for producing a chimeric polynucleotide encoding a polypeptide having a triazole-containing internucleotide linkage
WO2016014846A1 (en) 2014-07-23 2016-01-28 Moderna Therapeutics, Inc. Modified polynucleotides for the production of intrabodies
US11707436B2 (en) 2014-12-15 2023-07-25 Nanosphere Health Sciences Inc. Methods of treating inflammatory disorders and global inflammation with compositions comprising phospholipid nanoparticle encapsulations of NSAIDS
US10596124B2 (en) 2015-03-10 2020-03-24 Nanosphere Health Sciences, Llc Lipid nanoparticle compositions and methods as carriers of cannabinoids in standardized precision-metered dosage forms
US10028919B2 (en) 2015-03-10 2018-07-24 Nanosphere Health Sciences, Llc Lipid nanoparticle compositions and methods as carriers of cannabinoids in standardized precision-metered dosage forms
EP4011451A1 (en) 2015-10-22 2022-06-15 ModernaTX, Inc. Metapneumovirus mrna vaccines
EP4349404A2 (en) 2015-10-22 2024-04-10 ModernaTX, Inc. Respiratory virus vaccines
EP4349405A2 (en) 2015-10-22 2024-04-10 ModernaTX, Inc. Respiratory virus vaccines
EP4039699A1 (en) 2015-12-23 2022-08-10 ModernaTX, Inc. Methods of using ox40 ligand encoding polynucleotides
WO2017112943A1 (en) 2015-12-23 2017-06-29 Modernatx, Inc. Methods of using ox40 ligand encoding polynucleotides
WO2017120612A1 (en) 2016-01-10 2017-07-13 Modernatx, Inc. Therapeutic mrnas encoding anti ctla-4 antibodies
WO2018213789A1 (en) 2017-05-18 2018-11-22 Modernatx, Inc. Modified messenger rna comprising functional rna elements
EP4253544A2 (en) 2017-05-18 2023-10-04 ModernaTX, Inc. Modified messenger rna comprising functional rna elements
WO2018213731A1 (en) 2017-05-18 2018-11-22 Modernatx, Inc. Polynucleotides encoding tethered interleukin-12 (il12) polypeptides and uses thereof
WO2018232006A1 (en) 2017-06-14 2018-12-20 Modernatx, Inc. Polynucleotides encoding coagulation factor viii
CN109260181A (en) * 2017-07-17 2019-01-25 北京盈科瑞创新药物研究有限公司 Heparin nebulization sucking pharmaceutical solutions and preparation method thereof
WO2019104152A1 (en) 2017-11-22 2019-05-31 Modernatx, Inc. Polynucleotides encoding ornithine transcarbamylase for the treatment of urea cycle disorders
WO2019104195A1 (en) 2017-11-22 2019-05-31 Modernatx, Inc. Polynucleotides encoding propionyl-coa carboxylase alpha and beta subunits for the treatment of propionic acidemia
WO2019104160A2 (en) 2017-11-22 2019-05-31 Modernatx, Inc. Polynucleotides encoding phenylalanine hydroxylase for the treatment of phenylketonuria
WO2019116062A1 (en) 2017-12-12 2019-06-20 Lead Biotherapeutics Ltd. Solid lipid nanoparticle for intracellular release of active substances and method for production the same
WO2019136241A1 (en) 2018-01-05 2019-07-11 Modernatx, Inc. Polynucleotides encoding anti-chikungunya virus antibodies
WO2019200171A1 (en) 2018-04-11 2019-10-17 Modernatx, Inc. Messenger rna comprising functional rna elements
CN108484898A (en) * 2018-04-20 2018-09-04 江苏保易制药有限公司 The preparation method of polysorbate without polyoxyethylene isobide aliphatic ester
WO2019226650A1 (en) 2018-05-23 2019-11-28 Modernatx, Inc. Delivery of dna
WO2020023390A1 (en) 2018-07-25 2020-01-30 Modernatx, Inc. Mrna based enzyme replacement therapy combined with a pharmacological chaperone for the treatment of lysosomal storage disorders
WO2020047201A1 (en) 2018-09-02 2020-03-05 Modernatx, Inc. Polynucleotides encoding very long-chain acyl-coa dehydrogenase for the treatment of very long-chain acyl-coa dehydrogenase deficiency
WO2020056161A1 (en) 2018-09-12 2020-03-19 University Of Florida Research Foundation, Inc. Slow-cycling cell-rna based nanoparticle vaccine to treat cancer
WO2020056155A2 (en) 2018-09-13 2020-03-19 Modernatx, Inc. Polynucleotides encoding branched-chain alpha-ketoacid dehydrogenase complex e1-alpha, e1-beta, and e2 subunits for the treatment of maple syrup urine disease
WO2020056147A2 (en) 2018-09-13 2020-03-19 Modernatx, Inc. Polynucleotides encoding glucose-6-phosphatase for the treatment of glycogen storage disease
WO2020056239A1 (en) 2018-09-14 2020-03-19 Modernatx, Inc. Polynucleotides encoding uridine diphosphate glycosyltransferase 1 family, polypeptide a1 for the treatment of crigler-najjar syndrome
WO2020069169A1 (en) 2018-09-27 2020-04-02 Modernatx, Inc. Polynucleotides encoding arginase 1 for the treatment of arginase deficiency
WO2020097409A2 (en) 2018-11-08 2020-05-14 Modernatx, Inc. Use of mrna encoding ox40l to treat cancer in human patients
US11260033B2 (en) 2018-12-11 2022-03-01 Disruption Labs Inc. Compositions for the delivery of therapeutic agents and methods of use and making thereof
WO2020227615A1 (en) 2019-05-08 2020-11-12 Modernatx, Inc. Polynucleotides encoding methylmalonyl-coa mutase for the treatment of methylmalonic acidemia
WO2020227642A1 (en) 2019-05-08 2020-11-12 Modernatx, Inc. Compositions for skin and wounds and methods of use thereof
WO2020263883A1 (en) 2019-06-24 2020-12-30 Modernatx, Inc. Endonuclease-resistant messenger rna and uses thereof
WO2020263985A1 (en) 2019-06-24 2020-12-30 Modernatx, Inc. Messenger rna comprising functional rna elements and uses thereof
WO2021061707A1 (en) 2019-09-23 2021-04-01 Omega Therapeutics, Inc. Compositions and methods for modulating apolipoprotein b (apob) gene expression
WO2021061815A1 (en) 2019-09-23 2021-04-01 Omega Therapeutics, Inc. COMPOSITIONS AND METHODS FOR MODULATING HEPATOCYTE NUCLEAR FACTOR 4-ALPHA (HNF4α) GENE EXPRESSION
WO2021183720A1 (en) 2020-03-11 2021-09-16 Omega Therapeutics, Inc. Compositions and methods for modulating forkhead box p3 (foxp3) gene expression
WO2021247507A1 (en) 2020-06-01 2021-12-09 Modernatx, Inc. Phenylalanine hydroxylase variants and uses thereof
CN112120228A (en) * 2020-08-28 2020-12-25 盐城工学院 Solid lipid particle embedded with feruloyl oligosaccharide, preparation method thereof and application of solid lipid particle as food additive
CN112120228B (en) * 2020-08-28 2024-03-26 盐城工学院 Solid lipid particles embedded with feruloyl oligosaccharides, preparation method thereof and application of solid lipid particles as food additive
WO2022104131A1 (en) 2020-11-13 2022-05-19 Modernatx, Inc. Polynucleotides encoding cystic fibrosis transmembrane conductance regulator for the treatment of cystic fibrosis
CZ309587B6 (en) * 2021-01-22 2023-05-03 Oncora S.R.O. Microemulsion preconcentrate containing cladribine and preparing it
WO2022204370A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles and polynucleotides encoding ornithine transcarbamylase for the treatment of ornithine transcarbamylase deficiency
WO2022204380A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding propionyl-coa carboxylase alpha and beta subunits and uses thereof
WO2022204390A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding phenylalanine hydroxylase and uses thereof
WO2022204369A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Polynucleotides encoding methylmalonyl-coa mutase for the treatment of methylmalonic acidemia
WO2022204371A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding glucose-6-phosphatase and uses thereof
WO2022266083A2 (en) 2021-06-15 2022-12-22 Modernatx, Inc. Engineered polynucleotides for cell-type or microenvironment-specific expression
WO2022271776A1 (en) 2021-06-22 2022-12-29 Modernatx, Inc. Polynucleotides encoding uridine diphosphate glycosyltransferase 1 family, polypeptide a1 for the treatment of crigler-najjar syndrome
WO2023283359A2 (en) 2021-07-07 2023-01-12 Omega Therapeutics, Inc. Compositions and methods for modulating secreted frizzled receptor protein 1 (sfrp1) gene expression
WO2023056044A1 (en) 2021-10-01 2023-04-06 Modernatx, Inc. Polynucleotides encoding relaxin for the treatment of fibrosis and/or cardiovascular disease
WO2023161350A1 (en) 2022-02-24 2023-08-31 Io Biotech Aps Nucleotide delivery of cancer therapy
WO2023183909A2 (en) 2022-03-25 2023-09-28 Modernatx, Inc. Polynucleotides encoding fanconi anemia, complementation group proteins for the treatment of fanconi anemia
WO2023196399A1 (en) 2022-04-06 2023-10-12 Modernatx, Inc. Lipid nanoparticles and polynucleotides encoding argininosuccinate lyase for the treatment of argininosuccinic aciduria
WO2023215498A2 (en) 2022-05-05 2023-11-09 Modernatx, Inc. Compositions and methods for cd28 antagonism
CN114931551B (en) * 2022-05-11 2023-04-25 四川科瑞德制药股份有限公司 Miku ammonium chloride injection with storage stability at 25 ℃ and preparation method and application thereof
CN114931551A (en) * 2022-05-11 2022-08-23 四川科瑞德制药股份有限公司 Milakammonium chloride injection with storage stability at 25 ℃, preparation method and application thereof
WO2024026254A1 (en) 2022-07-26 2024-02-01 Modernatx, Inc. Engineered polynucleotides for temporal control of expression

Also Published As

Publication number Publication date
WO2013105101A8 (en) 2014-01-30

Similar Documents

Publication Publication Date Title
WO2013105101A1 (en) Solid lipid nanoparticles entrapping hydrophilic/ amphiphilic drug and a process for preparing the same
El-Zaafarany et al. Identifying lipidic emulsomes for improved oxcarbazepine brain targeting: In vitro and rat in vivo studies
Mu et al. Solid lipid nanocarriers in drug delivery: characterization and design
Wavikar et al. Nose to brain delivery of rivastigmine by in situ gelling cationic nanostructured lipid carriers: enhanced brain distribution and pharmacodynamics
Yasir et al. Solid lipid nanoparticles for nose to brain delivery of haloperidol: in vitro drug release and pharmacokinetics evaluation
Nasr et al. In vitro and in vivo evaluation of cubosomes containing 5-fluorouracil for liver targeting
Luo et al. Solid lipid nanoparticles for enhancing vinpocetine's oral bioavailability
Qi et al. Absorption, disposition and pharmacokinetics of solid lipid nanoparticles
Narang et al. Pharmaceutical development and regulatory considerations for nanoparticles and nanoparticulate drug delivery systems
Nagpal et al. Optimization of brain targeted chitosan nanoparticles of Rivastigmine for improved efficacy and safety
Simovic et al. An oral delivery system for indomethicin engineered from cationic lipid emulsions and silica nanoparticles
Wang et al. Submicron lipid emulsion as a drug delivery system for nalbuphine and its prodrugs
Mishra et al. Intestinal lymphatic delivery of praziquantel by solid lipid nanoparticles: formulation design, in vitro and in vivo studies
Pathak et al. Lipid nanocarriers: influence of lipids on product development and pharmacokinetics
JPS63122620A (en) Polylactic acid microsphere and production thereof
Vuddanda et al. Investigations on agglomeration and haemocompatibility of vitamin E TPGS surface modified berberine chloride nanoparticles
Liu et al. Combined strategies of apomorphine diester prodrugs and nanostructured lipid carriers for efficient brain targeting
Deng et al. The studies of N-Octyl-N-Arginine-Chitosan coated liposome as an oral delivery system of Cyclosporine A
Shah et al. Ciprofloxacin hydrochloride-loaded glyceryl monostearate nanoparticle: Factorial design of Lutrol F68 and Phospholipon 90G
Ghasemian et al. Brain delivery of baclofen as a hydrophilic drug by nanolipid carriers: Characteristics and pharmacokinetics evaluation
Hassan et al. Antiviral nanodelivery systems: current trends in acyclovir administration
US9186326B2 (en) Method and system for synthesizing nanocarrier based long acting drug delivery system for dexamethasone
Slabbert et al. Evaluation of the physical properties and stability of two lipid drug delivery systems containing mefloquine
Yuwanda et al. Study of valproic acid liposomes for delivery into the brain through an intranasal route
Hsu et al. PEGylated liposomes incorporated with nonionic surfactants as an apomorphine delivery system targeting the brain: in vitro release and in vivo real-time imaging

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: 12864771

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: 12864771

Country of ref document: EP

Kind code of ref document: A1