WO2021259673A1 - Hydroxychloroquine liquid compositions - Google Patents

Hydroxychloroquine liquid compositions Download PDF

Info

Publication number
WO2021259673A1
WO2021259673A1 PCT/EP2021/065801 EP2021065801W WO2021259673A1 WO 2021259673 A1 WO2021259673 A1 WO 2021259673A1 EP 2021065801 W EP2021065801 W EP 2021065801W WO 2021259673 A1 WO2021259673 A1 WO 2021259673A1
Authority
WO
WIPO (PCT)
Prior art keywords
pharmaceutical composition
liquid pharmaceutical
hcq
hydroxychloroquine
composition according
Prior art date
Application number
PCT/EP2021/065801
Other languages
French (fr)
Inventor
Joel SCHLATTER
Salvatore CISTERNINO
Philippe-Henri SECRETAN
Pascale DE LONLAY
Caroline Tuchmann-Durand
Perrine RENARD
Original Assignee
INSERM (Institut National de la Santé et de la Recherche Médicale)
Centre National De La Recherche Scientifique
Universite De Paris
Assistance Publique - Hopitaux De Paris (Ap-Hp)
Fondation Imagine
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 INSERM (Institut National de la Santé et de la Recherche Médicale), Centre National De La Recherche Scientifique, Universite De Paris, Assistance Publique - Hopitaux De Paris (Ap-Hp), Fondation Imagine filed Critical INSERM (Institut National de la Santé et de la Recherche Médicale)
Publication of WO2021259673A1 publication Critical patent/WO2021259673A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the invention relates to the pharmaceutical field.
  • the invention relates to pharmaceutical compositions including hydroxychloroquine or any one of its pharmaceutically acceptable salts as an active agent.
  • Hydroxychloroquine has been known since the early 1950s.
  • Hydroxychloroquine as an active agent, is an antimalarial drug indicated for the treatment or prevention of a plurality of acute and chronic conditions, including in particular the acute attacks of malaria due to Plasmodium vivax, P. malariae, P. ovale, and susceptible strains of P. falciparum. It is also particularly indicated for the treatment of discoid and systemic lupus erythematosus, and rheumatoid arthritis.
  • HCQ ulcerative colitis .
  • systemic autoimmune disease HCQ decreases pro -inflammatory cytokine secretion and impairs immune cell function.
  • HCQ reverses platelet activation induced by human antiphospholipid antibodies and protects the annexin A5 anticoagulant shield from disruption by antiphospholipid antibodies.
  • HCQ markedly suppresses the TLR9-mediated human B cell functions during inflammatory processes.
  • HCQ sulfate HCQ sulfate
  • the dosage is not relevant for pediatric daily posology.
  • children under 6 year’s old or geriatric population have difficulties in swallowing tablets. In this situation, the tablets are crushed to a powder that is then dissolved in some beverage such as fruit juice or water, and then the appropriate volume is administered to the patient. This practice leads to major problem concerning dosage accuracy and thus the drug efficacy.
  • HCQ sulfate 25-mg/mL suspensions reports the preparation of a HCQ sulfate 25-mg/mL suspensions in plastic bottles and syringes.
  • HCQ suspensions were prepared by crushing commercially available 200-mg tablets and re-suspending the powder in oral suspension media.
  • the preparations were reported to be chemically stable throughout the 90 days of the study period when stored at 25°C and 4°C.
  • these oral suspensions are not satisfactory, because they are very bitter to the point that may affect the patient’s acceptance and consequently the achievement of the therapeutic outcome. Also, their stability beyond 90 days had not been tested.
  • the global acceptability of the product must combine the effect of multiple contributing elements of the drug design including swallowability, palatability, appearance, ease of administration, and packaging, as well as the characteristics of the patient such as age.
  • the palatability defined as the organoleptic properties which include smell, taste, dose volume and texture is essential to consider for the formulation strategy of drug preparations.
  • the invention has for purpose to meet the above-mentioned needs.
  • the invention relates to a liquid pharmaceutical composition
  • a liquid pharmaceutical composition comprising: hydroxychloroquine (HCQ) or any one of its pharmaceutically acceptable salts, in a concentration equal or superior to 5 mg/mL; at least one flavouring and/or sweetening agent; sodium chloride; and an aqueous solvent.
  • HCQ hydroxychloroquine
  • the invention relates to its use as a medicament.
  • the invention relates to a container comprising a liquid pharmaceutical composition comprising hydroxychloroquine as defined above.
  • the invention further relates to a container comprising a liquid pharmaceutical composition, comprising: hydroxychloroquine (HCQ) or any one of its pharmaceutically acceptable salts, in a concentration equal or superior to 5 mg/mL; at least one flavouring and/or sweetening agent; sodium chloride; and an aqueous solvent.
  • HCQ hydroxychloroquine
  • the invention relates to a method for preparing a liquid pharmaceutical composition of hydroxychloroquine (HCQ), as previously defined.
  • the invention relates to a method for preparing a liquid pharmaceutical composition of hydroxychloroquine (HCQ), comprising the steps of: a) providing hydroxychloroquine (HCQ) or any one of its pharmaceutically acceptable salts in powder form; and b) bringing into contact said HCQ or pharmaceutically acceptable salt thereof in powder form, with at least one flavouring and/or sweetening agent, sodium chloride and an aqueous solvent; thereby preparing the liquid pharmaceutical composition.
  • Figure 1 Five-face study scale presented to subjects.
  • Figure 2 Results of palability comparing the selected formulation with an oral liquid formulation prepared from tablets as previously published in McHenry. From left to right: Very Bad - Bad - Not nice Not bad - Nice - Very nice. Results corresponding to the selected formulation are marked by an asterisk.
  • the inventors now provide herein a liquid pharmaceutical composition comprising hydroxychloroquine in substantial amounts, which is easy to prepare, easy to administer, palatable, and includes minimum potential adverse ingredients. This study further shows that the provided compositions possess excellent physical, chemical and microbiological stability of the selected oral suspensions over a 150-day study period.
  • liquid pharmaceutical compositions of hydroxychloroquine, as an active agent are also particularly convenient for paediatric use, in spite of the know bitter taste induced by the presence of hydroxychloroquine or is pharmaceutically acceptable salts.
  • the invention relates to a liquid pharmaceutical composition
  • a liquid pharmaceutical composition comprising: hydroxychloroquine (HCQ) or any one of its pharmaceutically acceptable salts, in a concentration equal or superior to 5 mg/mL, in particular equal or superior to 7 mg/mL; at least one flavouring and/or sweetening agent; sodium chloride; and an aqueous solvent.
  • HCQ hydroxychloroquine
  • the invention relates to a container comprising a liquid pharmaceutical composition comprising hydroxychloroquine as defined above.
  • the invention further relates to a container comprising a liquid pharmaceutical composition, comprising: hydroxychloroquine (HCQ) or any one of its pharmaceutically acceptable salts, in a concentration equal or superior to 5 mg/mL, in particular equal or superior to 7 mg/mL; at least one flavouring and/or sweetening agent; sodium chloride; and an aqueous solvent.
  • HCQ hydroxychloroquine
  • the invention relates to a method for preparing a liquid pharmaceutical composition of hydroxychloroquine (HCQ), as previously defined.
  • the invention relates to a method for preparing a liquid pharmaceutical composition of hydroxychloroquine (HCQ), comprising the steps of: c) providing hydroxychloroquine (HCQ) or any one of its pharmaceutically acceptable salts in powder form; and d) bringing into contact said HCQ or pharmaceutically acceptable salt thereof in powder form, with at least one flavouring and/or sweetening agent, sodium chloride and an aqueous solvent; thereby preparing the liquid pharmaceutical composition.
  • hydroxychloroquine or “ Oxychloroquine ” is intented to refer to the compound (2-[4-[(7-chloroquinolin-4-yl)amino]pentyl-ethylamino]ethanol, which is of formula:
  • this term is meant to include all the enantiomeric forms of the compound, including (R)-(-)-2-[ ⁇ 4-[(7-chloroquinolin-4- yl)amino]pentyl ⁇ (ethyl)amino]ethanol) [hereinafter (R)-(-)-hydroxychloroquine] and (S)-(+)- 2- [ ⁇ 4- [(7 -chloroquinolin-4-yl)amino]pentyl ⁇ (ethyl)amino] ethanol) [hereinafter (S )-(+)- hydroxychloroquine], and their racemates.
  • commercially available hydroxychloroquine is generally a racemic of the two previously mentioned enantiomers.
  • hydroxychloroquine and pharmaceutically acceptable salts thereof are also considered, including racemic and non-racemic compositions, comprising (S)-(+)-hydroxychloroquine and/or (R)-( — )-hydroxychloroquine
  • salts which are pharmaceutically acceptable, as defined herein, and which possess the desired pharmacological activity of the parent compound.
  • such salts may include:
  • hydrates and solvates (2) pharmaceutically acceptable acid addition salts formed with pharmaceutically acceptable inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like; or formed with pharmaceutically acceptable organic acids such as acetic acid, benzenesulfonic acid, benzoic acid, camphorsulfonic acid, citric acid, ethanesulfonic acid, fumaric acid, glucoheptonic acid, gluconic acid, acid glutamic acid, glycolic acid, hydroxynaphthoic acid, 2-hydroxyethanesulfonic acid, lactic acid, maleic acid, malic acid, mandelic acid, methanesulfonic acid, muconic acid, 2-naphthalenesulfonic acid, propionic acid Salicylic acid, succinic acid, dibenzoyl-L-tartaric acid, tartaric acid, p- toluenesulfonic acid, trimethylacetic acid,
  • compositions formed when an acidic proton present in the parent compound is either replaced by a metal ion, for example an alkali metal ion, an alkaline earth metal ion, or an aluminum ion ; is coordinated with a pharmaceutically acceptable organic or inorganic base.
  • Acceptable organic bases include diethanolamine, ethanolamine, N-methylglucamine, triethanolamine, tromethamine and the like.
  • Acceptable inorganic bases include aluminum hydroxide, calcium hydroxide, potassium hydroxide, sodium carbonate and sodium hydroxide.
  • This term is thus meant to include all pharmaceutically acceptable salts of hydroxychloroquine, although the sulfate form is preferred.
  • pharmaceutical acceptable salts of hydroxychloroquine which are considered herein, and throughout the specifically, include those selected from hydroxychloroquine sulfate and hydroxychloroquine phosphate and hydroxychloroquine diphosphonate; and preferably hydroxychloroquine sulfate.
  • suspension refers to a heterogeneous composition that contains solid particles sufficiently large for sedimentation. Generally, suspensions can be characterized by the presence of suspended particles or aggregates with an hydrodynamic radius superior to 1000 nm.
  • colloid refers to a heterogeneous composition that generally contains solid particles with an hydrodynamic radius from 1 to 1000 nm.
  • Particle size may be determined by any known method in the Art; in particular turbidimetry.
  • solution refers to a homogeneous composition that generally does not contain solid particles, or that contains solid particles with an hydrodynamic radius inferior to 1 nm.
  • colloids and solutions can be distinguished from suspensions in that they cannot be separated by filtration.
  • sweetener(s ) or “ sweetening agent(s)” refers to substances used to impart a sweet taste to foods or in table-top sweeteners. Such substances/compounds are generally used as an additive in compositions to increase palatability.
  • Sweeteners can be natural sweeteners or synthetic sweeteners.
  • sweeteners may include the common saccharide sweeteners, e.g., sucrose, fructose, glucose, and sweetener compositions comprising natural sugars, such as corn syrup (including high fructose corm syrup) or other syrups or sweetener concentrates derived from natural fruit and vegetable sources, semi-synthetic “ sugar alcohol” sweeteners such as erythritol, isomalt, lactitol, mannitol, sorbitol, xylitol, maltodextrin, and the like, and artificial sweeteners such as aspartame, saccharin, acesulfame-K, advantame, cyclamate, sucralose, Neohesperidin dihydrochalchone, neotame, saccharin and alitame.
  • natural sugars such as corn syrup (including high fructose corm syrup) or other syrups or sweetener concentrates derived from natural fruit and vegetable sources
  • Sweeteners also include cyclamic acid, mogroside, tagatose, maltose, galactose, mannose, sucrose, fructose, lactose, neotame and other aspartame derivatives, glucose, D-tryptophan, glycine, maltitol, lactitol, isomalt, hydrogenated glucose syrup (HGS), hydrogenated starch hydrolyzate (HSH), steviol glycosides such as stevioside, rebaudioside A and other sweet Stevia-based glycosides, carrelame and other guanidine-based sweeteners, etc.
  • Alternative sweeteners include sucralose and fructose.
  • Sucralose is a ‘high potency’ or ‘high intensity’ sweetener that is approximately 600 times as sweet as sucrose.
  • Fructose also known as “fruit sugar” is a 6- carbon polyhydroxyketone monosaccharide sugar that is often found in plants and in honey. The monosaccharide is found in crystalline form, often referred to as D-fmctose. Fructose can also be found as a component of other sweeteners such as high-fructose corn syrup (HFCS), which is a mixture of glucose and fructose.
  • HFCS high-fructose corn syrup
  • sweeteners include brazzein, curculin, erythritol, glycyrrhizic acid, mabilin/miraculin, mogrol glycosides, Luo Han Guo sweeteners, mogroside V, monatin, monellin, pentadin, and thaumatin.
  • sweeteners or also includes combinations of sweeteners as disclosed herein.
  • flavour or “flavouring agent(s)” or “flavoring agent(s)” encompasses to the definition of flavor enhancer as laid down in point 14 of Annex I of Regulation (EC) No 1333/2008 on food additives; which includes compounds/substances which enhance the existing taste and/or odor of a foodstuff. In particular, this term includes substances that enhance the sweet flavor of otherwise present sugars or sweeteners.
  • the term “pharmaceutical composition” refers to a formulation of a pharmaceutical active which renders the biological activity of the active ingredient (hydroxychloroquine or anyone of its pharmaceutically acceptable salts) therapeutically effective, but which does not include other ingredients which are obviously toxic to a subject to which the formulation are intended to be administered.
  • liquid composition further includes liquid compositions stored and/or packaged in any recipient or compartment, sealed or not, which is suitable for pharmaceutical compositions, which may thus include any liquid composition stored in vials, bottles, intraveinous (IV) bags, ampoules, cartridges and prefilled syringes.
  • liquid compositions which are particulatly considered in the context of the present invention are those which are compatible with oral administration.
  • the term “ container ” refers to any primary or secondary packaging material which is compatible with the storage of a liquid pharmaceutical composition.
  • such container may include single-dose containers, multi-dose containers, well-closed containers, airtight containers, light-resistant containers.
  • Such containers may be formed, completely or in-part, in glass, plastics, rubbers, paper/card boards and metals.
  • glass containers may include or consist of Type-I glass, Type-II glass, Type-III glass or any other non parental usage glass.
  • Plastic containers may include or consist of Urea formaldehyde (UF), Phenol formaldehyde, Melamine formaldehyde (MF), Epoxy resins (epoxides), Polyurethanes (PURs), Polyethylene, Polyvinylchloride, Polyethylene terepthalate (PET), Polyvinylidene chloride (PVdC), Polycarbonate Acrylonitrile butadiene styrene (ABS).
  • Urea formaldehyde UF
  • Phenol formaldehyde Phenol formaldehyde
  • MF Melamine formaldehyde
  • Epoxy resins epoxides
  • PURs Polyurethanes
  • Polyethylene Polyvinylchloride
  • PET Polyethylene terepthalate
  • PVdC Polyvinylidene chloride
  • ABS Polycarbonate Acrylonitrile butadiene styrene
  • Such containers may comprise or consist of vials, bottles, intravenous (
  • sterile composition refers to any form of administration which is substantially free, or even devoid of viable or revivable germs, potentially infectious, microbial known to those skilled in the art.
  • the term “substanti free”, when used in relation to a given component of a composition refers to a composition to which essentially none of said component has been added.
  • a composition is “ substantially free” of a given component, said composition suitably comprises no more than 0.001 wt % of said component, suitably no more than 0.0001 wt % of said component, suitably no more than 0.00001 wt %, more suitably no more than 0.000001 wt.
  • the term “buffering agent” or « buffer » refers to an acid or base component (usually a weak acid or weak base) of a buffer or buffer solution.
  • a buffering agent helps maintain the pH of a given solution (i.e. the liquid pharmaceutical composition) at or near to a pre-determined value, and the buffering agents are generally chosen to complement the pre-determined value.
  • a buffering agent is suitably a single compound which gives rise to a desired buffering effect, especially when said buffering agent is mixed with (and suitably capable of proton exchange with) an appropriate amount (depending on the pre determined pH desired) of its corresponding acid/base conjugate, or if the required amount of its corresponding acid/base conjugate is formed in situ — this may be achieved by adding strong acid or base until the required pH is reached.
  • palatability » refers to the organoleptic properties which include smell, taste, dose, volume and texture.
  • hydroxychloroquine (HCQ) or its pharmaceutically acceptable salts are selected from the group consisting of hydroxychloroquine sulfate, or hydroxychloroquine phosphate; preferably hydroxychloroquine sulfate.
  • the liquid pharmaceutical composition according to the invention comprises a substantial amount of hydroxychloroquine or its its pharmaceutically acceptable salts.
  • the liquid pharmaceutical composition comprises an amount of HCQ or its pharmaceutically acceptable salts equal or superior to 5 mg/mL of the liquid pharmaceutical composition.
  • the liquid pharmaceutical composition comprises an amount of HCQ or its pharmaceutically acceptable salts equal or superior to 7 mg/mL of the liquid pharmaceutical composition.
  • the liquid pharmaceutical composition comprises an amount of HCQ or its pharmaceutically acceptable salts (i.e. hydroxychloroquine sulfate) superior to 5 mg/mL, or even superior to 7 mg/mL.
  • the liquid pharmaceutical composition comprises an amount of HCQ or its pharmaceutically acceptable salts (i.e. hydroxychloroquine sulfate) equal or superior to 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49 mg/mL; which also includes equal or superior to 50 mg/mL.
  • HCQ or its salts i.e. hydroxychloroquine sulfate
  • HCQ or its salts i.e. hydroxychloroquine sulfate
  • HCQ or its salts is present in a concentration equal or superior to 50 mg/mL.
  • the at least one flavouring and/or sweetening agent is/are selected from the group consisting of: caramel aroma, orange aroma, strawberry aroma, raspberry aroma, maltitol, or mixtures thereof.
  • the at least one flavouring and/or sweetening agent is/are present in the liquid pharmaceutical composition as a total volume fraction ranging from 10% to 80% (w/v).
  • the caramel aroma is a composition comprising glucose, fructose and maltose.
  • the liquid pharmaceutical composition comprises from 10% (w/v) to 40% (w/v) of glucose.
  • the liquid pharmaceutical composition comprises from 10% (w/v) to 30% (w/v) of fructose.
  • the liquid pharmaceutical composition comprises from 1% (w/v) to 10% (w/v) of maltose.
  • the liquid pharmaceutical composition comprises:
  • the liquid pharmaceutical composition comprises less than 1% (w/v) of sucrose; or is even substantially devoid of sucrose.
  • the liquid pharmaceutical composition may comprise maltitol.
  • the at least one flavouring and/or sweetening agent is/are selected from a caramel aroma and an orange aroma; and preferably is a mixture of caramel and orange aroma.
  • the flavouring and/or sweetening agent i.e. orange aroma
  • the flavouring and/or sweetening agent is a natural aroma.
  • the liquid pharmaceutical composition is further characterized in that it comprises sodium chloride.
  • the concentration of sodium chloride is optimised for reducing the bitterness of HCQ, yet also maintained below the daily dose required.
  • sodium chloride is present in the liquid pharmaceutical composition in a concentration equal or superior to 10 mg/mL.
  • sodium chloride is present in the liquid pharmaceutical composition in a concentration equal or lower than 30 mg/mL.
  • sodium chloride is present in the liquid pharmaceutical composition in a concentration ranging from 10 mg/mL to 50 mg/mL, in particular from 10 mg/mL to 30 mg/mL; which may thus include 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49 and 50 mg/mL of liquid pharmaceutical composition.
  • the liquid pharmaceutical composition may further comprise a preservative agent; preferably sodium methylparaben.
  • the liquid pharmaceutical composition may further comprise an excipient, in particular one or more thickeners, such as a derivative of cellulose.
  • the liquid pharmaceutical composition comprises hydroxypropylcellulose.
  • the liquid pharmaceutical composition may comprise hydroxypropylcellulose in an amount comprised between 1% and 3%, (w/v), for instance of 2% (w/v).
  • the pH of the liquid pharmaceutical composition ranges from 3 to 8 (including 3, 4, 5, 6, 7, and 8) at 4°C or 25°C.
  • the pH of the liquid pharmaceutical composition ranges from 3 to 5 at 4°C or 25 °C; for instance of about 3.4 at 4°C or 25°C.
  • the liquid pharmaceutical composition comprises one or more buffering agents.
  • Buffering agents may be selected from : ascorbic acid, acetic acid, tartaric acid, citric acid monohydrate, trisodium citrate dehydrate, sodium citrate, potassium citrate, sodium phosphate, tricalcium phosphate, calcium carbonate, sodium bicarbonate, calcium phosphate, carbonated calcium phosphate, magnesium hydroxide, hydrochloric acid, sodium hydroxide or their mixtures thereof.
  • the liquid pharmaceutical composition comprises a macrocyclic carbohydrate, such as a cyclodextrine (i.e. hydroxypropyl-B-cyclodextrin (HPBCD)).
  • HPBCD hydroxypropyl-B-cyclodextrin
  • the liquid pharmaceutical composition does not comprise a macrocyclic carbohydrate, such as a cyclodextrine (i.e. hydroxypropyl-B-cyclodextrin (HPBCD)).
  • a macrocyclic carbohydrate such as a cyclodextrine (i.e. hydroxypropyl-B-cyclodextrin (HPBCD)).
  • HPBCD hydroxypropyl-B-cyclodextrin
  • the liquid pharmaceutical composition a sterile and/or apyretic composition.
  • the liquid pharmaceutical composition is a colloid or a solution.
  • the liquid pharmaceutical composition is not a suspension.
  • the liquid pharmaceutical composition comprises an aqueous solvent selected from water and aqueous mixtures of a polar organic solvent, for example an alcohol such as methanol, ethanol or the like, or apolar ether such as dioxane, tetrahydrofuran or similar.
  • a polar organic solvent for example an alcohol
  • apolar ether such as dioxane, tetrahydrofuran or similar.
  • the aqueous solvent is water (i.e. purified water).
  • the liquid pharmaceutical composition is suitable for use as a medicament.
  • the liquid pharmaceutical composition is suitable for use for the treatment and/or prevention and/or reduction of the likelihood of occurrence of acute and chronic conditions, including in particular the acute attacks of malaria due to Plasmodium vivax, P. malariae, P. ovale, and susceptible strains of P. falciparum.
  • the liquid pharmaceutical composition is suitable for use for the treatment and/or prevention and/or reduction of the likelihood of occurrence of discoid and systemic lupus erythematosus, and rheumatoid arthritis.
  • the liquid pharmaceutical composition is particularly suitable as a medicament for paediatric and geriatric populations.
  • the paediatric population is defined herein as that group of the population between birth and 18 years of age.
  • the geriatric population is defined herein as that group of the population aged 65 and over.
  • the invention relates to a method for preparing a liquid pharmaceutical composition of hydroxychloroquine (HCQ), as previously defined.
  • the invention relates to a method for preparing a liquid pharmaceutical composition of hydroxychloroquine (HCQ), comprising the steps of: a) providing hydroxychloroquine (HCQ) or any one of its pharmaceutically acceptable salts in powder form; and b) bringing into contact said HCQ or pharmaceutically acceptable salt thereof in powder form, with at least one flavouring and/or sweetening agent, sodium chloride and an aqueous solvent; thereby preparing the liquid pharmaceutical composition.
  • the method may further include a step c) of mixing the composition obtained at step b).
  • the invention relates to a method for preparing a liquid pharmaceutical composition of hydroxychloroquine (HCQ), comprising the steps of: a) providing hydroxychloroquine (HCQ) or any one of its pharmaceutically acceptable salts in powder form; and b) bringing into contact said HCQ or pharmaceutically acceptable salt thereof in powder form, with at least one flavouring and/or sweetening agent, sodium chloride and an aqueous solvent; thereby preparing the liquid pharmaceutical composition; c) mixing the composition obtained at step b), preferably in a manner suitable to obtain a colloid or a solution.
  • HCQ hydroxychloroquine
  • the above-mentioned methods further include introducing the obtained pharmaceutical composition at step b) or c) into a container; preferably a container suitable for oral administration.
  • ICH International Council on Harmonisation
  • the physical appearance properties were examined using visual observation of the samples stored at each condition. Color and pH were evaluated at 0, 7, 14, 21, 30, 45, 60, 90, and 150 days. The pH value of the samples was determined on each study day using a SevenEasy model pH meter (Mettler-Toledo, Viroflay, France). The pH meter was calibrated daily with standard pH 4 and pH 7 buffer solutions purchased from VWR International (Leuven, Belgium). Three replicates of the samples were performed, and the results were shown as mean ⁇ standard deviation (SD).
  • SD standard deviation
  • the test involved the artificial contamination of the sample formulation, using a graded inoculum of prescribed microorganisms according the European pharmacopeia v. 8.0 (“Efficacy of antimicrobial preservation”; 01/2011:50103 EDQM; Council of Europe 8 th edition, France, 2013:600-601).
  • the inoculated product was kept at room temperature and away from light for 28 days.
  • the number of microorganisms was monitored by sampling at defined time intervals by counts of the microorganisms in the samples taken.
  • the product preservation properties were suitable if, under the test conditions, a significant reduction of microorganisms in the inoculated product occurred over the defined time intervals.
  • a neutralizing solution was used to ensure that any preservative effect of the formulation was neutralized at the moment of microbial enumeration, allowing the existent microorganisms to be recovered and counted in agar medium.
  • a mixture of compounds was prepared using 30 g polysorbate 80, 3 g soy lecithin, 1 g histidine, 1 g peptone for casein, 4.3g sodium chloride, 3.6 g potassium phosphate monobasic, 7.2 g phosphate disodium in 1 L purified water. Tryptone agar medium (TSA) and Sabouraud dextrose agar (SDA) were used as culture media.
  • TSA Tryptone agar medium
  • SDA Sabouraud dextrose agar
  • the method was considered validated when the number of colony-forming units (CFUs) counted in 1 mL of inoculated sample was at least 50% of that obtained in the control (neutralizing solution inoculated with each microorganism). At each time point, the log reduction in the number of viable microorganisms against the value obtained for the inoculum was calculated. This log reduction is then compared with the recommended values of the European Pharmacopeia described in the following table:
  • the HPFC method was validated for specificity, limit of detection (FOD), limit of quantification (FOQ), linearity, precision, accuracy, according to the European Medicines Agency guidelines (ICH Topic Q1A(R2): stability testing of new drug substances and products; August 2003).
  • the specificity was assessed by subjecting HCQ suspensions to various forced degradation conditions. Suspensions of HCQ 25 mg/mL were mixed with 0.1M HC1, 0.1M NaOH, and 3% H202 prior to being maintained at 60°C away from light. The UV spectral purity (200-365 nm) of the HCG peak in chromatograms of degraded sample was retained to evaluate the final chromatographic system. For linearity determination, calibration curve was determined using triplicate injections at six concentration levels.
  • the calibration curve was used to confirm the linear relationship between the analyte peak areas and the analyte concentration.
  • the slope, intercept, and regression coefficient (r) were calculated as regression parameters by the least square method.
  • the accuracy for the active compound was determined by analyzing three replicates of samples prepared at 80 %, 100 % and 120 % of the target concentration. Accuracy was expressed as percentage of recovery determined by experimental concentration/theoretical concentration x 100. The acceptance criterion was ⁇ 2% deviation from the normal value for the recovery of HCQ.
  • the precision was determined by analyzing six replicates samples and expressed as relative standard deviation (RSD) which was expected to be lower than 2%.
  • the LOD and LOQ for HCQ assay were determined by calibration curve method by using the following equations:
  • Table 3 here below provides one example of a composition of HCQ preparations and EMA data on the limits of the excipients known to have an adverse effect according to the European Medicines Agency (EMA) (Annex to the European Commission guideline on ‘Excipients in the labelling and package leaflet of medicinal products for human use’ (SANTE-2017- 11668). As published in November 2019).
  • EMA European Medicines Agency
  • the thresholds in oral liquids were zero for glucose and fructose, and 391-mg sodium representing approximatively 20% of the WHO adult recommended maximum daily dietary intake of 2 g sodium.
  • Table X summarizes the intra- and inter-day precision for the determination of HCQ.
  • the relative standard deviation (RSD) values were less than 1.5% for all concentrations tested and confirmed the very good intra- and inter-day precision of the method (expressed as CV: coefficient of variation).
  • HCQ 25 mg/mL and 50 mg/mL oral suspension stored in amber plastic bottles at 2-8°C and 22-25°C temperatures demonstrated chemical stability for up to 150 days (Table 8). HCQ retained at least 95% of its initial concentration for all conditions at 150 days.
  • microbiological effectiveness data with bacteria and fungi are summarized in Table 9.
  • Table 9 The microbiological effectiveness data with bacteria and fungi are summarized in Table 9.
  • the HCQ suspensions presented a suitable preservative efficacy against the 6 strains tested to bring the product in the line with criteria of the European Pharmacopeia oral preparation.
  • An extemporaneously prepared oral suspension of HCQ sulfate 25 mg/mL and 50 mg/mL was physically, chemically, and microbiologically stable for 150 days when stored at room or refrigerated temperature in plastic bottles.
  • the suspensions were optimized to ensure a satisfactory palatability for adult and pediatric patients.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Dispersion Chemistry (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention relates to liquid pharmaceutical compositions including hydroxychloroquine or any one of its pharmaceutically acceptable salts as an active agent. The inventors have identified a pharmaceutical composition for the administration of hydroxychloroquine, with better acceptance and high stability. Containers comprising the liquid pharmaceutical compositions and methods of preparation are further disclosed.

Description

HYDROXYCHLOROQUINE LIQUID COMPOSITIONS
FIELD OF THE DISCLOSURE
The invention relates to the pharmaceutical field.
In particular, the invention relates to pharmaceutical compositions including hydroxychloroquine or any one of its pharmaceutically acceptable salts as an active agent.
BACKGROUND OF THE DISCLOSURE
Hydroxychloroquine (HCQ) has been known since the early 1950s.
Hydroxychloroquine, as an active agent, is an antimalarial drug indicated for the treatment or prevention of a plurality of acute and chronic conditions, including in particular the acute attacks of malaria due to Plasmodium vivax, P. malariae, P. ovale, and susceptible strains of P. falciparum. It is also particularly indicated for the treatment of discoid and systemic lupus erythematosus, and rheumatoid arthritis.
The mechanism of action of HCQ is complex. In systemic autoimmune disease, HCQ decreases pro -inflammatory cytokine secretion and impairs immune cell function. Moreover, HCQ reverses platelet activation induced by human antiphospholipid antibodies and protects the annexin A5 anticoagulant shield from disruption by antiphospholipid antibodies. HCQ markedly suppresses the TLR9-mediated human B cell functions during inflammatory processes.
Currently, its most wide-spread commercial form, HCQ sulfate, is only available in 200-mg tablets (i.e. Plaquenil®), which are not easy to administer for certain populations, and especially pediatric patients.
First, the dosage is not relevant for pediatric daily posology. Second, children under 6 year’s old or geriatric population have difficulties in swallowing tablets. In this situation, the tablets are crushed to a powder that is then dissolved in some beverage such as fruit juice or water, and then the appropriate volume is administered to the patient. This practice leads to major problem concerning dosage accuracy and thus the drug efficacy.
To overcome these difficulties, the development of liquid presentations is needed to ensure that the patients and medical staff have access to safe and accurate dosage forms of drugs. The European Paediatric Formulations Initiatives (EuPFI) set out criteria for the choice of an oral form in children, including child acceptability, doses suitable for pediatric use, convenience of use with minimal impact on pediatric life-style, and efficacy and safety (excipients, stability, errors in administration). McHenry et al. (“ Stability of Extemporaneously Prepared Hydroxychloroquine Sulfate 25-mg/mL Suspensions in Plastic Bottles and Syringes”. Int J. Pharm. Compd; 2017) reports the preparation of a HCQ sulfate 25-mg/mL suspensions in plastic bottles and syringes. According to this work, HCQ suspensions were prepared by crushing commercially available 200-mg tablets and re-suspending the powder in oral suspension media. The preparations were reported to be chemically stable throughout the 90 days of the study period when stored at 25°C and 4°C. Unfortunately, these oral suspensions are not satisfactory, because they are very bitter to the point that may affect the patient’s acceptance and consequently the achievement of the therapeutic outcome. Also, their stability beyond 90 days had not been tested.
Pauli et al. (“ Evaluation of an Immediate-Release Formulation of Hydroxychloroquine Sulfate With an Interwoven Pediatric Taste-Masking System”. Pharmaceutics, Drug Delivery; 2020) teaches an immediate-release formulation of hydroxychloroquine sulfate including a taste-masking system. This document includes a preliminary study on the effect of ion pairing and pH increase in overall taste intensity for compositions of hydroxychloroquine in amounts of 4 mg/mL and 6.5 mg/mL. However those formulations are not meant to be ready-for-use. Their stability is not assessed and the studied concentrations are lower, which limits their use and posology.
The global acceptability of the product must combine the effect of multiple contributing elements of the drug design including swallowability, palatability, appearance, ease of administration, and packaging, as well as the characteristics of the patient such as age. The palatability defined as the organoleptic properties which include smell, taste, dose volume and texture is essential to consider for the formulation strategy of drug preparations.
There is thus a need to identify novel means for the administration of hydroxychloroquine. In particular, there is a need to identify novel formulations comprising hydroxychloroquine in substantial amounts, and yet which remain suitable for administration, to pediatric and geriatric populations.
There is also a need for formulations which remain easy to prepare, easy to administer, customizable, palatable, and with minimum potential adverse ingredients.
There is also a need for formulations which remain stable over time.
The invention has for purpose to meet the above-mentioned needs.
SUMMARY According to a first main embodiment, the invention relates to a liquid pharmaceutical composition comprising: hydroxychloroquine (HCQ) or any one of its pharmaceutically acceptable salts, in a concentration equal or superior to 5 mg/mL; at least one flavouring and/or sweetening agent; sodium chloride; and an aqueous solvent.
In particular, the invention relates to its use as a medicament.
According to a second main embodiment, the invention relates to a container comprising a liquid pharmaceutical composition comprising hydroxychloroquine as defined above. Hence, according this second main embodiment, the invention further relates to a container comprising a liquid pharmaceutical composition, comprising: hydroxychloroquine (HCQ) or any one of its pharmaceutically acceptable salts, in a concentration equal or superior to 5 mg/mL; at least one flavouring and/or sweetening agent; sodium chloride; and an aqueous solvent.
According to a third main embodiment, the invention relates to a method for preparing a liquid pharmaceutical composition of hydroxychloroquine (HCQ), as previously defined. Hence, the invention relates to a method for preparing a liquid pharmaceutical composition of hydroxychloroquine (HCQ), comprising the steps of: a) providing hydroxychloroquine (HCQ) or any one of its pharmaceutically acceptable salts in powder form; and b) bringing into contact said HCQ or pharmaceutically acceptable salt thereof in powder form, with at least one flavouring and/or sweetening agent, sodium chloride and an aqueous solvent; thereby preparing the liquid pharmaceutical composition.
DESCRIPTION OF THE FIGURES
Figure 1: Five-face study scale presented to subjects.
Figure 2: Results of palability comparing the selected formulation with an oral liquid formulation prepared from tablets as previously published in McHenry. From left to right: Very Bad - Bad - Not nice Not bad - Nice - Very nice. Results corresponding to the selected formulation are marked by an asterisk.
DETAILED DESCRIPTION OF THE DISCLOSURE
The inventors now provide herein a liquid pharmaceutical composition comprising hydroxychloroquine in substantial amounts, which is easy to prepare, easy to administer, palatable, and includes minimum potential adverse ingredients. This study further shows that the provided compositions possess excellent physical, chemical and microbiological stability of the selected oral suspensions over a 150-day study period.
The inventors propose that such liquid pharmaceutical compositions of hydroxychloroquine, as an active agent, are also particularly convenient for paediatric use, in spite of the know bitter taste induced by the presence of hydroxychloroquine or is pharmaceutically acceptable salts.
Thus, according to a first main embodiment, the invention relates to a liquid pharmaceutical composition comprising: hydroxychloroquine (HCQ) or any one of its pharmaceutically acceptable salts, in a concentration equal or superior to 5 mg/mL, in particular equal or superior to 7 mg/mL; at least one flavouring and/or sweetening agent; sodium chloride; and an aqueous solvent.
According to a second main embodiment, the invention relates to a container comprising a liquid pharmaceutical composition comprising hydroxychloroquine as defined above. Hence, according this second main embodiment, the invention further relates to a container comprising a liquid pharmaceutical composition, comprising: hydroxychloroquine (HCQ) or any one of its pharmaceutically acceptable salts, in a concentration equal or superior to 5 mg/mL, in particular equal or superior to 7 mg/mL; at least one flavouring and/or sweetening agent; sodium chloride; and an aqueous solvent.
According to a third main embodiment, the invention relates to a method for preparing a liquid pharmaceutical composition of hydroxychloroquine (HCQ), as previously defined. Hence, the invention relates to a method for preparing a liquid pharmaceutical composition of hydroxychloroquine (HCQ), comprising the steps of: c) providing hydroxychloroquine (HCQ) or any one of its pharmaceutically acceptable salts in powder form; and d) bringing into contact said HCQ or pharmaceutically acceptable salt thereof in powder form, with at least one flavouring and/or sweetening agent, sodium chloride and an aqueous solvent; thereby preparing the liquid pharmaceutical composition.
Definitions
As used herein, the term “hydroxychloroquine” , or “ Oxychloroquine ” is intented to refer to the compound (2-[4-[(7-chloroquinolin-4-yl)amino]pentyl-ethylamino]ethanol, which is of formula:
Figure imgf000006_0001
In the absence of contrary indications, this term is meant to include all the enantiomeric forms of the compound, including (R)-(-)-2-[{4-[(7-chloroquinolin-4- yl)amino]pentyl}(ethyl)amino]ethanol) [hereinafter (R)-(-)-hydroxychloroquine] and (S)-(+)- 2- [ { 4- [(7 -chloroquinolin-4-yl)amino]pentyl } (ethyl)amino] ethanol) [hereinafter (S )-(+)- hydroxychloroquine], and their racemates. For example, commercially available hydroxychloroquine is generally a racemic of the two previously mentioned enantiomers.
Other particular forms of hydroxychloroquine, and pharmaceutically acceptable salts thereof are also considered, including racemic and non-racemic compositions, comprising (S)-(+)-hydroxychloroquine and/or (R)-( — )-hydroxychloroquine
As used herein, the term "pharmaceutically acceptable salts" of a compound thus means salts which are pharmaceutically acceptable, as defined herein, and which possess the desired pharmacological activity of the parent compound. In a non-exhaustive manner, such salts may include:
(1) hydrates and solvates, (2) pharmaceutically acceptable acid addition salts formed with pharmaceutically acceptable inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like; or formed with pharmaceutically acceptable organic acids such as acetic acid, benzenesulfonic acid, benzoic acid, camphorsulfonic acid, citric acid, ethanesulfonic acid, fumaric acid, glucoheptonic acid, gluconic acid, acid glutamic acid, glycolic acid, hydroxynaphthoic acid, 2-hydroxyethanesulfonic acid, lactic acid, maleic acid, malic acid, mandelic acid, methanesulfonic acid, muconic acid, 2-naphthalenesulfonic acid, propionic acid Salicylic acid, succinic acid, dibenzoyl-L-tartaric acid, tartaric acid, p- toluenesulfonic acid, trimethylacetic acid, trifluoroacetic acid and the like, or
(3) pharmaceutically acceptable base addition salts formed when an acidic proton present in the parent compound is either replaced by a metal ion, for example an alkali metal ion, an alkaline earth metal ion, or an aluminum ion ; is coordinated with a pharmaceutically acceptable organic or inorganic base. Acceptable organic bases include diethanolamine, ethanolamine, N-methylglucamine, triethanolamine, tromethamine and the like. Acceptable inorganic bases include aluminum hydroxide, calcium hydroxide, potassium hydroxide, sodium carbonate and sodium hydroxide.
This term is thus meant to include all pharmaceutically acceptable salts of hydroxychloroquine, although the sulfate form is preferred.
In particular, pharmaceutical acceptable salts of hydroxychloroquine which are considered herein, and throughout the specifically, include those selected from hydroxychloroquine sulfate and hydroxychloroquine phosphate and hydroxychloroquine diphosphonate; and preferably hydroxychloroquine sulfate.
As used herein, the term “ suspension ” refers to a heterogeneous composition that contains solid particles sufficiently large for sedimentation. Generally, suspensions can be characterized by the presence of suspended particles or aggregates with an hydrodynamic radius superior to 1000 nm.
As used herein, the term “ colloid ’ refers to a heterogeneous composition that generally contains solid particles with an hydrodynamic radius from 1 to 1000 nm.
Particle size may be determined by any known method in the Art; in particular turbidimetry.
As used herein, the term “ solution ” refers to a homogeneous composition that generally does not contain solid particles, or that contains solid particles with an hydrodynamic radius inferior to 1 nm. In general, colloids and solutions can be distinguished from suspensions in that they cannot be separated by filtration.
As used herein, the term “ sweetener(s )” or “ sweetening agent(s)” refers to substances used to impart a sweet taste to foods or in table-top sweeteners. Such substances/compounds are generally used as an additive in compositions to increase palatability. Sweeteners can be natural sweeteners or synthetic sweeteners. In a non- exhaustive manner, such sweeteners may include the common saccharide sweeteners, e.g., sucrose, fructose, glucose, and sweetener compositions comprising natural sugars, such as corn syrup (including high fructose corm syrup) or other syrups or sweetener concentrates derived from natural fruit and vegetable sources, semi-synthetic “ sugar alcohol” sweeteners such as erythritol, isomalt, lactitol, mannitol, sorbitol, xylitol, maltodextrin, and the like, and artificial sweeteners such as aspartame, saccharin, acesulfame-K, advantame, cyclamate, sucralose, Neohesperidin dihydrochalchone, neotame, saccharin and alitame. Sweeteners also include cyclamic acid, mogroside, tagatose, maltose, galactose, mannose, sucrose, fructose, lactose, neotame and other aspartame derivatives, glucose, D-tryptophan, glycine, maltitol, lactitol, isomalt, hydrogenated glucose syrup (HGS), hydrogenated starch hydrolyzate (HSH), steviol glycosides such as stevioside, rebaudioside A and other sweet Stevia-based glycosides, carrelame and other guanidine-based sweeteners, etc. Alternative sweeteners include sucralose and fructose. Sucralose is a ‘high potency’ or ‘high intensity’ sweetener that is approximately 600 times as sweet as sucrose. Fructose (also known as “fruit sugar”) is a 6- carbon polyhydroxyketone monosaccharide sugar that is often found in plants and in honey. The monosaccharide is found in crystalline form, often referred to as D-fmctose. Fructose can also be found as a component of other sweeteners such as high-fructose corn syrup (HFCS), which is a mixture of glucose and fructose. Other types of natural sweeteners, include brazzein, curculin, erythritol, glycyrrhizic acid, mabilin/miraculin, mogrol glycosides, Luo Han Guo sweeteners, mogroside V, monatin, monellin, pentadin, and thaumatin. The term “ sweeteners” or also includes combinations of sweeteners as disclosed herein.
As used herein, the term “ flavour ” or “flavouring agent(s)” or “flavoring agent(s)” encompasses to the definition of flavor enhancer as laid down in point 14 of Annex I of Regulation (EC) No 1333/2008 on food additives; which includes compounds/substances which enhance the existing taste and/or odor of a foodstuff. In particular, this term includes substances that enhance the sweet flavor of otherwise present sugars or sweeteners. As used herein, the term “pharmaceutical composition” refers to a formulation of a pharmaceutical active which renders the biological activity of the active ingredient (hydroxychloroquine or anyone of its pharmaceutically acceptable salts) therapeutically effective, but which does not include other ingredients which are obviously toxic to a subject to which the formulation are intended to be administered.
As used herein, the term “ liquid composition” further includes liquid compositions stored and/or packaged in any recipient or compartment, sealed or not, which is suitable for pharmaceutical compositions, which may thus include any liquid composition stored in vials, bottles, intraveinous (IV) bags, ampoules, cartridges and prefilled syringes. The liquid compositions which are particulatly considered in the context of the present invention are those which are compatible with oral administration.
As used herein, the term “ container ” refers to any primary or secondary packaging material which is compatible with the storage of a liquid pharmaceutical composition. In a non-exhaustive manner, such container may include single-dose containers, multi-dose containers, well-closed containers, airtight containers, light-resistant containers. Such containers may be formed, completely or in-part, in glass, plastics, rubbers, paper/card boards and metals. For example, glass containers may include or consist of Type-I glass, Type-II glass, Type-III glass or any other non parental usage glass. Plastic containers may include or consist of Urea formaldehyde (UF), Phenol formaldehyde, Melamine formaldehyde (MF), Epoxy resins (epoxides), Polyurethanes (PURs), Polyethylene, Polyvinylchloride, Polyethylene terepthalate (PET), Polyvinylidene chloride (PVdC), Polycarbonate Acrylonitrile butadiene styrene (ABS). Such containers may comprise or consist of vials, bottles, intravenous (IV) bags, ampoules, cartridges and prefilled syringes.
As used herein, the term “ sterile composition” refers to any form of administration which is substantially free, or even devoid of viable or revivable germs, potentially infectious, microbial known to those skilled in the art.
As used herein, the term “ substantially free”, when used in relation to a given component of a composition (e.g. “ a liquid pharmaceutical composition substantially free of germs”), refers to a composition to which essentially none of said component has been added. When a composition is “ substantially free” of a given component, said composition suitably comprises no more than 0.001 wt % of said component, suitably no more than 0.0001 wt % of said component, suitably no more than 0.00001 wt %, more suitably no more than 0.000001 wt. As used herein, the term “buffering agent” or « buffer » refers to an acid or base component (usually a weak acid or weak base) of a buffer or buffer solution. A buffering agent helps maintain the pH of a given solution (i.e. the liquid pharmaceutical composition) at or near to a pre-determined value, and the buffering agents are generally chosen to complement the pre-determined value. A buffering agent is suitably a single compound which gives rise to a desired buffering effect, especially when said buffering agent is mixed with (and suitably capable of proton exchange with) an appropriate amount (depending on the pre determined pH desired) of its corresponding acid/base conjugate, or if the required amount of its corresponding acid/base conjugate is formed in situ — this may be achieved by adding strong acid or base until the required pH is reached.
As used herein, the term « palatability » refers to the organoleptic properties which include smell, taste, dose, volume and texture.
Liquid pharmaceutical compositions & containers
According to one particular embodiment of the liquid pharmaceutical composition, hydroxychloroquine (HCQ) or its pharmaceutically acceptable salts are selected from the group consisting of hydroxychloroquine sulfate, or hydroxychloroquine phosphate; preferably hydroxychloroquine sulfate.
Advantageously, the liquid pharmaceutical composition according to the invention comprises a substantial amount of hydroxychloroquine or its its pharmaceutically acceptable salts. Hence, according to one particular embodiment, the liquid pharmaceutical composition comprises an amount of HCQ or its pharmaceutically acceptable salts equal or superior to 5 mg/mL of the liquid pharmaceutical composition. According to one particular embodiment, the liquid pharmaceutical composition comprises an amount of HCQ or its pharmaceutically acceptable salts equal or superior to 7 mg/mL of the liquid pharmaceutical composition.
Hence, according to particular embodiments, the liquid pharmaceutical composition comprises an amount of HCQ or its pharmaceutically acceptable salts (i.e. hydroxychloroquine sulfate) superior to 5 mg/mL, or even superior to 7 mg/mL.
According to particular embodiments, the liquid pharmaceutical composition comprises an amount of HCQ or its pharmaceutically acceptable salts (i.e. hydroxychloroquine sulfate) equal or superior to 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49 mg/mL; which also includes equal or superior to 50 mg/mL. According to one embodiment, HCQ or its salts (i.e. hydroxychloroquine sulfate) is present in a concentration equal or superior to 25 mg/mL.
According to one embodiment, HCQ or its salts (i.e. hydroxychloroquine sulfate) is present in a concentration equal or superior to 50 mg/mL.
According to one embodiment, the at least one flavouring and/or sweetening agent is/are selected from the group consisting of: caramel aroma, orange aroma, strawberry aroma, raspberry aroma, maltitol, or mixtures thereof.
According to one embodiment, the at least one flavouring and/or sweetening agent is/are present in the liquid pharmaceutical composition as a total volume fraction ranging from 10% to 80% (w/v).
According to one embodiment, the caramel aroma is a composition comprising glucose, fructose and maltose.
According to one embodiment, the liquid pharmaceutical composition comprises from 10% (w/v) to 40% (w/v) of glucose.
According to one embodiment, the liquid pharmaceutical composition comprises from 10% (w/v) to 30% (w/v) of fructose.
According to one embodiment, the liquid pharmaceutical composition comprises from 1% (w/v) to 10% (w/v) of maltose.
According to one embodiment, the liquid pharmaceutical composition comprises:
- from 10% (w/v) to 40% (w/v) of glucose;
- from 10% (w/v) to 30% (w/v) of fructose;
- from 1% (w/v) to 10% (w/v) of maltose.
According to one embodiment, the liquid pharmaceutical composition comprises less than 1% (w/v) of sucrose; or is even substantially devoid of sucrose.
According to some particular embodiment, the liquid pharmaceutical composition may comprise maltitol.
In particular, the at least one flavouring and/or sweetening agent is/are selected from a caramel aroma and an orange aroma; and preferably is a mixture of caramel and orange aroma. Advantageously, the flavouring and/or sweetening agent (i.e. orange aroma) is a natural aroma.
The liquid pharmaceutical composition is further characterized in that it comprises sodium chloride. Advantageously, the concentration of sodium chloride is optimised for reducing the bitterness of HCQ, yet also maintained below the daily dose required. Hence, according to some embodiments, sodium chloride is present in the liquid pharmaceutical composition in a concentration equal or superior to 10 mg/mL.
According to some embodiments, sodium chloride is present in the liquid pharmaceutical composition in a concentration equal or lower than 30 mg/mL.
Preferably, sodium chloride is present in the liquid pharmaceutical composition in a concentration ranging from 10 mg/mL to 50 mg/mL, in particular from 10 mg/mL to 30 mg/mL; which may thus include 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49 and 50 mg/mL of liquid pharmaceutical composition.
According to some embodiments, the liquid pharmaceutical composition may further comprise a preservative agent; preferably sodium methylparaben.
Examples of other preservative agents which are suitable for the liquid pharmaceutical composition may comprise sodium benzoate, benzoic acid, boric acid, sorbic acid and their salts thereof, benzyl alcohol, benzalkonium chloride, parahydroxybenzoic acids and their alkyl esters, methyl and propyl parabens or their mixtures thereof.
According to some embodiments, the liquid pharmaceutical composition may further comprise an excipient, in particular one or more thickeners, such as a derivative of cellulose. According to some embodiments, the liquid pharmaceutical composition comprises hydroxypropylcellulose. For example, the liquid pharmaceutical composition may comprise hydroxypropylcellulose in an amount comprised between 1% and 3%, (w/v), for instance of 2% (w/v).
According to some embodiments, the pH of the liquid pharmaceutical composition ranges from 3 to 8 (including 3, 4, 5, 6, 7, and 8) at 4°C or 25°C.
According to exemplary embodiments, the pH of the liquid pharmaceutical composition ranges from 3 to 5 at 4°C or 25 °C; for instance of about 3.4 at 4°C or 25°C.
According to some embodiments, the liquid pharmaceutical composition comprises one or more buffering agents. Buffering agents may be selected from : ascorbic acid, acetic acid, tartaric acid, citric acid monohydrate, trisodium citrate dehydrate, sodium citrate, potassium citrate, sodium phosphate, tricalcium phosphate, calcium carbonate, sodium bicarbonate, calcium phosphate, carbonated calcium phosphate, magnesium hydroxide, hydrochloric acid, sodium hydroxide or their mixtures thereof. According to some alternative embodiments, the liquid pharmaceutical composition comprises a macrocyclic carbohydrate, such as a cyclodextrine (i.e. hydroxypropyl-B-cyclodextrin (HPBCD)).
According to some alternative embodiments, the liquid pharmaceutical composition does not comprise a macrocyclic carbohydrate, such as a cyclodextrine (i.e. hydroxypropyl-B-cyclodextrin (HPBCD)).
According to a particular embodiment, the liquid pharmaceutical composition a sterile and/or apyretic composition.
According to a particular embodiment, the liquid pharmaceutical composition is a colloid or a solution.
According to a particular embodiment, the liquid pharmaceutical composition is not a suspension.
According to a particular embodiment the liquid pharmaceutical composition comprises an aqueous solvent selected from water and aqueous mixtures of a polar organic solvent, for example an alcohol Such as methanol, ethanol or the like, or apolar ether such as dioxane, tetrahydrofuran or similar. According to one exemplary embodiment, the aqueous solvent is water (i.e. purified water).
According to one embodiment, the liquid pharmaceutical composition is suitable for use as a medicament. In particular, the liquid pharmaceutical composition is suitable for use for the treatment and/or prevention and/or reduction of the likelihood of occurrence of acute and chronic conditions, including in particular the acute attacks of malaria due to Plasmodium vivax, P. malariae, P. ovale, and susceptible strains of P. falciparum.
According to one embodiment, the liquid pharmaceutical composition is suitable for use for the treatment and/or prevention and/or reduction of the likelihood of occurrence of discoid and systemic lupus erythematosus, and rheumatoid arthritis.
According to one embodiment, the liquid pharmaceutical composition is particularly suitable as a medicament for paediatric and geriatric populations. The paediatric population is defined herein as that group of the population between birth and 18 years of age. The geriatric population is defined herein as that group of the population aged 65 and over.
Process of preparation According to a third main embodiment, the invention relates to a method for preparing a liquid pharmaceutical composition of hydroxychloroquine (HCQ), as previously defined. Hence, the invention relates to a method for preparing a liquid pharmaceutical composition of hydroxychloroquine (HCQ), comprising the steps of: a) providing hydroxychloroquine (HCQ) or any one of its pharmaceutically acceptable salts in powder form; and b) bringing into contact said HCQ or pharmaceutically acceptable salt thereof in powder form, with at least one flavouring and/or sweetening agent, sodium chloride and an aqueous solvent; thereby preparing the liquid pharmaceutical composition.
In particular, the method may further include a step c) of mixing the composition obtained at step b).
Hence, the invention relates to a method for preparing a liquid pharmaceutical composition of hydroxychloroquine (HCQ), comprising the steps of: a) providing hydroxychloroquine (HCQ) or any one of its pharmaceutically acceptable salts in powder form; and b) bringing into contact said HCQ or pharmaceutically acceptable salt thereof in powder form, with at least one flavouring and/or sweetening agent, sodium chloride and an aqueous solvent; thereby preparing the liquid pharmaceutical composition; c) mixing the composition obtained at step b), preferably in a manner suitable to obtain a colloid or a solution.
Preferably, the above-mentioned methods further include introducing the obtained pharmaceutical composition at step b) or c) into a container; preferably a container suitable for oral administration.
EXAMPLES
Material & Methods
Chemicals
The pure pharmaceutical ingredients of hydroxychloroquine sulfate, sodium methyl paraben, hydroxypropylcellulose (Klucel), xanthan gum, strawberry aroma, raspberry aroma, and banana aroma were provided from Inresa-Pharma (Bartenheim, France). Sodium chloride powder and caramel liquid were purchased from COOPER (melon, France). Orange aroma was provided from La Pateliere (Condom, France). Maltitol syrup was provided from Roquette (Beinheim, France). Sterile purified water was obtained from Fresenius Kabi France (Sevres, france). Other chemicals were analytical grade. All solvents used were HPLC grade from Merck (Darmstadt, Germany). Formulation development assay
Four different formulations were used to prepare the HCQ suspensions at 25 mg/mL (Table 1):
Figure imgf000015_0001
Figure imgf000016_0001
Table 1. Formulations of suspensions of HCQ at 25 mg/mL
All formulations included as excipients a suspended agent (hydroxypropylcellulose) at different levels in order to modify the viscosity, a sweetening agent (maltitol syrup), a diluent agent (purified water), and aroma to mask the bitter taste of drug. The formulations were tested at pH 4 and pH 8 to verify the pH influence over the formulation taste. To complete the test, hedonic faces scale was used to assess the subject acceptability.
Method to assess the taste of HCQ formulations
To evaluate drug acceptability, scaling methods are the most widely applied and include direct pediatric reports as well as reports made by parents/caregivers or healthcare professionals on behalf of children. However, the best scale in terms of validity, reliability, feasibility, and preference remains unclear because the lack of appropriate standardized scale, as evidenced in Mistry et al. (“ Methodology Used to Assess Acceptability of Oral Pediatric Medicines: A Systematic Literature Search and Narrative Review”. Paediatr Drugs. 2017).
The most commonly methods used to assess the acceptability of medicines are hedonic scales and visual analog scales. A study that examined the preference of three pediatric pain measurements tools reported that children preferred the facial hedonic scale according to Luffy et al. (“Examining the validity, reliability, and preference of three pediatric pain measurement tools in African-American children”. Pediatr Nurs. 2003). Faces scales employ a number of illustrations depicting facial expressions based on children's taste experiences, thereby avoiding the need for children to quantify this experiment numerically.
To assess the palatability of the HCQ formulations, the new hedonic faces scale TASTY developed specifically for evaluating taste in children, and reported in Wagner et al. (“ Examining a New Scale for Evaluating Taste in Children (TASTY)”. J Pediatr Pharmacol Ther. 2020), was adapted to our study.
The investigator explained that the taster would be asked the question “how do you estimate the taste of the drug” and encouraged to refer to their preference by pointing to the appropriate face on the specific scale (Figure 1). Prior to the first tasting, the subject was asked to rinse his mouth out with several sips of mineral water. The investigator dispensed 0.5 mL of the first drug formulation into a 1-mL oral syringe and instructed the subject to consume full volume of medication. Subject pointed then to the appropriate face on the face scale. After completing the first measurement, the subject was instructed to rinse the mouth by taking several sips of mineral water. The test process was repeated with the second and others formulations. A period of 5 minutes was observed between each test. The acceptance of subject was rated from the faces quotation scale (Figure 1).
Stability study of the selected formulations
Stability of HCQ suspensions at 25 mg/mL and 50 mg/mL were evaluated according to the International Council on Harmonisation (ICH) guidelines on the stability testing of new drug substances and products, as described by the European Medicines Agency (ICH Topic Q1A(R2): stability testing of new drug substances and products; August 2003).
Six bottles of the preparations at each concentration were prepared, and bottles were stored at 25 ± 3°C and 5 ± 3°C. Physical and chemical examinations were performed in triplicate immediately after preparation (Day 0) and at Day 7, 14, 21, 30, 45, 60, 90, and 150 to define drug stability throughout its period of storage. The preparation was considered physically stable if appearance and pH were not changed. The chemical stability of the extemporaneous preparation was defined by the drug content that contained not less than 90% and not more 110% of the labeled amount of HCQ, according to Food and Drug Administration (FDA) guidance for industry on drug stability dated December 9, 2008.
Physical stability tests
The physical appearance properties were examined using visual observation of the samples stored at each condition. Color and pH were evaluated at 0, 7, 14, 21, 30, 45, 60, 90, and 150 days. The pH value of the samples was determined on each study day using a SevenEasy model pH meter (Mettler-Toledo, Viroflay, France). The pH meter was calibrated daily with standard pH 4 and pH 7 buffer solutions purchased from VWR International (Leuven, Belgium). Three replicates of the samples were performed, and the results were shown as mean ± standard deviation (SD).
Microbiological quality control
To evaluate the efficacy of antimicrobial preservation of the selected HCQ formulation in bottles, the test involved the artificial contamination of the sample formulation, using a graded inoculum of prescribed microorganisms according the European pharmacopeia v. 8.0 (“Efficacy of antimicrobial preservation”; 01/2011:50103 EDQM; Council of Europe 8th edition, Strasbourg, France, 2013:600-601).
The inoculated product was kept at room temperature and away from light for 28 days. The number of microorganisms was monitored by sampling at defined time intervals by counts of the microorganisms in the samples taken. The product preservation properties were suitable if, under the test conditions, a significant reduction of microorganisms in the inoculated product occurred over the defined time intervals. Six collection type strains were included corresponding to three bacteria ( Pseudomonas aeruginosa ATCC 9027, Staphylococcus aureus ATCC 6538, and Escherichia coli ATCC 8739) and three fungi (Candida albicans ATCC 10231, Aspergillus brasiliensis ATCC 16404, Zygosaccharomyces rouxii IP 2021.92).
A neutralizing solution was used to ensure that any preservative effect of the formulation was neutralized at the moment of microbial enumeration, allowing the existent microorganisms to be recovered and counted in agar medium. For this purpose, a mixture of compounds was prepared using 30 g polysorbate 80, 3 g soy lecithin, 1 g histidine, 1 g peptone for casein, 4.3g sodium chloride, 3.6 g potassium phosphate monobasic, 7.2 g phosphate disodium in 1 L purified water. Tryptone agar medium (TSA) and Sabouraud dextrose agar (SDA) were used as culture media.
For each reference strain, 1 mL of a suspension containing between 1.102 and 1.103 CFU/mL is added to 9 mL of the neutralizing agent, for the test in the absence of the product. In to two 9cm-diameter Petri dishes for each medium, 1 mL of the previous suspension was introduced separately. The same procedure was applied to the selected HCQ formulation. A count of the number of CFU per dish was performed after a maximum incubating time of 5 days at 30-38 °C for soybean-casein digest agar and a maximum incubating time of 3 days at 20-25°C for the Sabouraud dextrose agar. The method was considered validated when the number of colony-forming units (CFUs) counted in 1 mL of inoculated sample was at least 50% of that obtained in the control (neutralizing solution inoculated with each microorganism). At each time point, the log reduction in the number of viable microorganisms against the value obtained for the inoculum was calculated. This log reduction is then compared with the recommended values of the European Pharmacopeia described in the following table:
Figure imgf000019_0001
NI: No Increase in number of viable microorganisms compared to the previous reading
Table 2. European Pharmacopeia acceptance criteria for oral preparation
High-performance liquid chromatography method
The stability-indicating high-performance liquid chromatography (HPLC) method used to analyze HCQ and its degradation products was developed by modifying the method previously published by McHenry et al. (“ Stability of Extemporaneously Prepared Hydroxychloroquine Sulfate 25-mg/mL Suspensions in Plastic Bottles and Syringes”. Int J. Pharm. Compd; 2017). Briefly, the analysis was performed using a Dionex Ultimate 3000 system (Thermo-Fisher, Villebon-sur-Yvette, France) including a diode array detector (DAD) with 5 cm flow cell and with Chromeleon software (Version 8.0, Thermo-Fisher) as data processor. Separation was achieved by a reversed-phase Polaris C18 column (5-micron particle size, 250 x 4.6 mm, Agilent Technologies, Courtaboeuf, France) that was kept at 40°C with a gradient elution. The mobile phase consisted of a gradient of 0.01M ammonium acetate with 0.1% formic acid (A), acetonitrile (B), and methanol (C). The flow rate was maintained at 0.8 mF/min, and the gradient profile was as follows: t0-14.0 min: A = 95% B = 2.5% C = 2.5%; U4.0-20.0 min: A = 5% B = 47.5% C = 47.5%, t20.0-24.0 min: A = 95% B = 2.5% C = 2.5%. The injection volume was 30 pF. The ultraviolet absorption of the drug was obtained at 268 nm. Under these conditions, the retention time of HCQ was observed to be about 9.2 minutes.
Validation procedure
The HPFC method was validated for specificity, limit of detection (FOD), limit of quantification (FOQ), linearity, precision, accuracy, according to the European Medicines Agency guidelines (ICH Topic Q1A(R2): stability testing of new drug substances and products; August 2003). The specificity was assessed by subjecting HCQ suspensions to various forced degradation conditions. Suspensions of HCQ 25 mg/mL were mixed with 0.1M HC1, 0.1M NaOH, and 3% H202 prior to being maintained at 60°C away from light. The UV spectral purity (200-365 nm) of the HCG peak in chromatograms of degraded sample was retained to evaluate the final chromatographic system. For linearity determination, calibration curve was determined using triplicate injections at six concentration levels. The calibration curve was used to confirm the linear relationship between the analyte peak areas and the analyte concentration. The slope, intercept, and regression coefficient (r) were calculated as regression parameters by the least square method. The accuracy for the active compound was determined by analyzing three replicates of samples prepared at 80 %, 100 % and 120 % of the target concentration. Accuracy was expressed as percentage of recovery determined by experimental concentration/theoretical concentration x 100. The acceptance criterion was ±2% deviation from the normal value for the recovery of HCQ. The precision was determined by analyzing six replicates samples and expressed as relative standard deviation (RSD) which was expected to be lower than 2%. The LOD and LOQ for HCQ assay were determined by calibration curve method by using the following equations:
L0D =
S 3 lo3 p X e o S fD c 0 af li 7 br~ at in iot ner cC ue rP vt e
10 x SD of y — intercept
LOQ Slope of calibration curve
Statistical analysis
Data analyses were performed using Prism 6 (Version 6.01, GraphPad Software, San Diego, USA). Descriptive statistics for continuous variables were expressed as mean ±
SD.
Results
Formulation development assay
Five adult volunteers and two children (3 and 6 years old) and were randomized and evaluated to taste the formulations using the hedonic faces scale. All subjects (100%) rated the taste of each formulation including those prepared with HPBCD as very bad, the lowest possible ratings. Because the palatability analysis of all formulations demonstrated highly unsatisfactory, new formulation was proposed to ensure better palatability and well acceptance, supporting the use of HCQ in pediatric and adult patients. Finally, the selected HCQ formulation was composed of the active pharmaceutical ingredient HCQ, caramel, orange aroma, sodium chloride, sodium methylparaben, and purified water.
The Table 3 here below provides one example of a composition of HCQ preparations and EMA data on the limits of the excipients known to have an adverse effect according to the European Medicines Agency (EMA) (Annex to the European Commission guideline on ‘Excipients in the labelling and package leaflet of medicinal products for human use’ (SANTE-2017- 11668). As published in November 2019).
The thresholds in oral liquids were zero for glucose and fructose, and 391-mg sodium representing approximatively 20% of the WHO adult recommended maximum daily dietary intake of 2 g sodium.
Figure imgf000021_0001
Table 3. Selected formulations of hydroxychloroquine sulfate 25 mg/mL and 50 mg/mL and EMA data on the excipients known to potentially give adverse effects. Method validation
Linear relationship between the peak area and the concentration range for HCQ was established. The calculated regression parameters are given in Table X here below and are within the linearity acceptance criteria. Table X summarizes the intra- and inter-day precision for the determination of HCQ. The relative standard deviation (RSD) values were less than 1.5% for all concentrations tested and confirmed the very good intra- and inter-day precision of the method (expressed as CV: coefficient of variation).
Figure imgf000022_0001
Table 4. Results from study of precision
The percentage recoveries were found to be 99.3%- 101.8% with RSD ranges - 0.62-1.83% (Table 5). The results of recovery studies demonstrated accuracy of the proposed method. The determined values of Limit Of Detection (LOD) and Limit of Quantification (LOQ) were 17.2 pg/mL and 26.9 pg/mL, respectively, calculated using slope and Y- intercept.
Figure imgf000022_0002
Figure imgf000023_0001
Results from study of accuracy
Specificity
In acidic and alkaline stress conditions, 99% of the HCQ concentration was maintained with no degradation products over the 7-day study period. However, with the 3% hydrogen peroxide at 50°C, approximatively 52% of the HCQ was lost over the 7-day study period (Table 6).
Figure imgf000023_0002
Table 6. Forced degradation studies of HCQ from suspension
Palatability of the selected formulation
The results of palatability were presented in figure 2 and compared with the oral liquid formulation prepared from tablets as previously published. The new formulation prepared from HCQ powder was preferred by adults and children compared to the formulation prepared from tablets with majority rating of not nice not bad and nice. The tested pH of the final composition is of about 3.4.
Physical stability
There were no detectable changes in color and taste in any samples over the study period at two studied temperatures. The pH of HCQ suspensions 25 mg/mL and 50 mg/mL in plastic bottles were no statistically different over the time of both storage conditions (Table 7). The pH of HCQ suspensions 25 mg/mL and 50 mg/mL was no statistically different (ANOVA) over the time of refrigerated conditions (p-value >0.05). However, in ambient temperature condition, the pH of both suspensions was significantly different over the study period (p-values: 0.010 and 0.011).
Figure imgf000024_0002
Table 7. pH determination at each day assay
Chemical stability of HCQ
The HCQ 25 mg/mL and 50 mg/mL oral suspension stored in amber plastic bottles at 2-8°C and 22-25°C temperatures demonstrated chemical stability for up to 150 days (Table 8). HCQ retained at least 95% of its initial concentration for all conditions at 150 days.
Actual Mean ± SD % HCQ concentration remaining
Figure imgf000024_0001
Figure imgf000025_0001
Table 8. Chemical stability of HCQ suspensions stored at 2-8°C and 22-25°C in plastic bottles Microbiological study
The microbiological effectiveness data with bacteria and fungi are summarized in Table 9. In the context of the study, the HCQ suspensions presented a suitable preservative efficacy against the 6 strains tested to bring the product in the line with criteria of the European Pharmacopeia oral preparation.
Figure imgf000025_0002
Figure imgf000026_0001
Table 9. Results of antimicrobial effectiveness test. The inoculum is defined herein as the count of microorganisms introduced per ml of product. Counts are given as a medium of the 2 counts done for each dilution (CFU/mL). Discussion and Conclusions
An extemporaneously prepared oral suspension of HCQ sulfate 25 mg/mL and 50 mg/mL was physically, chemically, and microbiologically stable for 150 days when stored at room or refrigerated temperature in plastic bottles. The suspensions were optimized to ensure a satisfactory palatability for adult and pediatric patients.

Claims

1. A liquid pharmaceutical composition comprising: hydroxychloroquine (HCQ) or any one of its pharmaceutically acceptable salts, in a concentration equal or superior to 5 mg/mL; at least one flavouring and/or sweetening agent; sodium chloride; an aqueous solvent.
2. The liquid pharmaceutical composition according to claim 1, wherein HCQ or its salts are selected from the group consisting of hydroxychloroquine sulfate, or hydroxychloroquine phosphate; preferably hydroxychloroquine sulfate.
3. The liquid pharmaceutical composition according to claim 1 or 2, wherein HCQ or its salts is present in a concentration equal or superior to 7 mg/mL.
4. The liquid pharmaceutical composition according to claim 1 or 2, wherein HCQ or its salts is present in a concentration equal or superior to 25 mg/mL.
5. The liquid pharmaceutical composition according to any one of the preceding claims, wherein the at least one flavouring and/or sweetening agent is/are selected from the group consisting of: caramel aroma, orange aroma, strawberry aroma, raspberry aroma, maltitol, or mixtures thereof.
6. The liquid pharmaceutical composition according to any one of the preceding claims, wherein the at least one flavouring and/or sweetening agent is/are selected from a caramel aroma and an orange aroma; and preferably is a mixture of caramel and orange aroma.
7. The liquid pharmaceutical composition according to any one of the preceding claims, comprising maltitol.
8. The liquid pharmaceutical composition according to any one of the preceding claims; further comprising a preservative agent; preferably sodium methylparaben.
9. The liquid pharmaceutical composition according to any one of the preceding claims, which comprises hydroxypropylcellulose.
10. The liquid pharmaceutical composition according to any one of the preceding claims, wherein the pH ranges from 3 to 8 at 4°C or 25°C.
11. The liquid pharmaceutical composition according to any one of the preceding claims, wherein the composition is a sterile and/or apyretic composition.
12. The liquid pharmaceutical composition according to any one of the preceding claims, wherein the composition is a colloid or a solution.
13. The liquid pharmaceutical composition according to any one of the preceding claims; for use as a medicament.
14. A container comprising the liquid pharmaceutical composition according to any one of the preceding claims.
15. A method for preparing a liquid pharmaceutical composition of hydroxychloroquine (HCQ) according to any one of claims 1 to 14, comprising the steps of: a) providing hydroxychloroquine (HCQ) or any one of its pharmaceutically acceptable salts in powder form; and b) bringing into contact said HCQ or pharmaceutically acceptable salt thereof in powder form, with at least one flavouring and/or sweetening agent, sodium chloride and an aqueous solvent; thereby preparing the liquid pharmaceutical composition.
PCT/EP2021/065801 2020-06-24 2021-06-11 Hydroxychloroquine liquid compositions WO2021259673A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP20305694.0 2020-06-24
EP20305694 2020-06-24

Publications (1)

Publication Number Publication Date
WO2021259673A1 true WO2021259673A1 (en) 2021-12-30

Family

ID=71614807

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2021/065801 WO2021259673A1 (en) 2020-06-24 2021-06-11 Hydroxychloroquine liquid compositions

Country Status (1)

Country Link
WO (1) WO2021259673A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CH664284A5 (en) * 1985-07-08 1988-02-29 Arcopharma Sa Chloroquine medicaments without bitter taste - contain complex of chloroquine with copolymer of methacrylic] acid and methyl methacrylate]
WO2019075229A1 (en) * 2017-10-13 2019-04-18 Research Triangle Institute Hydroxychloroquine sulfate formulations and methods for preparation and use thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CH664284A5 (en) * 1985-07-08 1988-02-29 Arcopharma Sa Chloroquine medicaments without bitter taste - contain complex of chloroquine with copolymer of methacrylic] acid and methyl methacrylate]
WO2019075229A1 (en) * 2017-10-13 2019-04-18 Research Triangle Institute Hydroxychloroquine sulfate formulations and methods for preparation and use thereof

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
"European pharmacopeia", 2013, article "Efficacy of antimicrobial preservation", pages: 600 - 601
CHAUHAN R: "Taste Masking: A Unique Approach for Bitter Drugs", JOURNAL OF STEM CELL BIOLOGY AND TRANSPLANTATION, vol. 1, no. 2, 1 January 2017 (2017-01-01), XP055741189, DOI: 10.21767/2575-7725.100012 *
HARMIK SOHI ET AL: "Taste Masking Technologies in Oral Pharmaceuticals: Recent Developments and Approaches", JOURNAL DRUG DEVELOPMENT AND INDUSTRIAL PHARMACY, vol. 30, no. 5, 1 January 2004 (2004-01-01), US, pages 429 - 448, XP055741215, ISSN: 0363-9045, DOI: 10.1081/DDC-120037477 *
LUFFY ET AL.: "Examining the validity, reliability, and preference of three pediatric pain measurement tools in African-American children", PEDIATR NURS, 2003
MCHENRY ET AL.: "Stability of Extemporaneously Prepared Hydroxychloroquine Sulfate 25-mglmL Suspensions in Plastic Bottles and Syringes", INT J. PHARM. COMPD, 2017
MISTRY ET AL.: "Methodology Used to Assess Acceptability of Oral Pediatric Medicines: A Systematic Literature Search and Narrative Review", PAEDIATR DRUGS, 2017
PAULI ET AL.: "Evaluation of an Immediate-Release Formulation of Hydroxychloroquine Sulfate With an Interwoven Pediatric Taste-Masking System", PHARMACEUTICS, DRUG DELIVERY, 2020
WAGNER ET AL.: "Examining a New Scale for Evaluating Taste in Children (TASTY)", J PEDIATR PHARMACOL THER, 2020

Similar Documents

Publication Publication Date Title
JP5695029B2 (en) Pediatric solutions containing beta-blockers
US20120238631A1 (en) Compositions comprising cyclohexylamines and aminoadamantanes
US10959985B1 (en) Pharmaceutical compositions including carvedilol and methods of using the same
WO2021259673A1 (en) Hydroxychloroquine liquid compositions
EP3755373B1 (en) Oral solutions comprising tramadol
Fuss et al. Stability study of a compounded oral solution of nicardipine for the treatment of hypertension in children
WO2019174755A1 (en) Oral solutions comprising tramadol and acetaminophen
CN107929240A (en) Piperaquine phosphate oral liquid and preparation method thereof
WO2024132080A1 (en) Liquid pharmaceutical composition containing flecainide and process for the preparaton thereof
AU2773892A (en) Trimethoprim oral liquid
GB2614972A (en) Stable composition of chloral hydrate
CN115645357A (en) Lacosamide oral solution prescription and preparation process thereof
US20230149359A1 (en) Stable pharmaceutical compositions of clonidine
CN117122562A (en) Meifenacin ammonium oral solution
CA2828316A1 (en) Prucalopride oral solution

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: 21730630

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: 21730630

Country of ref document: EP

Kind code of ref document: A1