insoluble drug delivery strategies

Upload: psykhodelyk

Post on 24-Feb-2018

231 views

Category:

Documents


0 download

TRANSCRIPT

  • 7/25/2019 Insoluble Drug Delivery Strategies

    1/12

    H O S T E D B Y

    REVIEW

    Insoluble drug delivery strategies: review

    of recent advances and business prospects

    Sandeep Kalepua,n, Vijaykumar Nekkantib

    aDepartment of Pharmaceutical Technology, Shri Vishnu College of Pharmacy, Bhimavaram 534202, Andhra Pradesh,

    IndiabCollege of Pharmaceutical Sciences, Western University of Health Sciences, Pomona, California 91766, USA

    Received 9 January 2015; received in revised form 9 May 2015; accepted 26 May 2015

    KEY WORDS

    Bioavailability;Cocrystals;Solubility;Inclusion complexation;Nanoparticles;Self-emulsifying formula

    tions;Proliposomes

    Abstract The emerging trends in the combinatorial chemistry and drug design have led to thedevelopment of drug candidates with greater lipophilicity, high molecular weight and poor watersolubility. Majority of the failures in new drug development have been attributed to poor water solubilityof the drug. Issues associated with poor solubility can lead to low bioavailability resulting in suboptimal

    drug delivery. About 40% of drugs with market approval and nearly 90% of molecules in the discoverypipeline are poorly water-soluble. With the advent of various insoluble drug delivery technologies, thechallenge to formulate poorly water soluble drugs could be achieved. Numerous drugs associated withpoor solubility and low bioavailabilities have been formulated into successful drug products. Severalmarketed drugs were reformulated to improve efcacy, safety and patient compliance. In order to gainmarketing exclusivity and patent protection for such products, revitalization of poorly soluble drugs usinginsoluble drug delivery technologies have been successfully adopted by many pharmaceutical companies.This review covers the recent advances in the eld of insoluble drug delivery and business prospects.

    &2015 Chinese Pharmaceutical Association and Institute of Materia Medica, Chinese Academy of Medical

    Sciences. Production and hosting by Elsevier B.V. This is an open access article under the CC BY-NC-ND

    license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

    Chinese Pharmaceutical Association

    Institute of Materia Medica, Chinese Academy of Medical Sciences

    www.elsevier.com/locate/apsbwww.sciencedirect.com

    Acta Pharmaceutica Sinica B

    http://dx.doi.org/10.1016/j.apsb.2015.07.0032211-3835 & 2015 Chinese Pharmaceutical Association and Institute of Materia Medica, Chinese Academy of Medical Sciences. Production and hosting byElsevier B.V. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

    nCorresponding author. Tel.: 91 9948444546; fax: 91 8816 250863.

    E-mail address:[email protected](Sandeep Kalepu).

    Peer review under responsibility of Institute of Materia Medica, Chinese Academy of Medical Sciences and Chinese Pharmaceutical Association.

    Acta Pharmaceutica Sinica B 2015;5(5):442453

    http://www.elsevier.com/locate/apsbhttp://www.sciencedirect.com/http://localhost/var/www/apps/conversion/tmp/scratch_1/dx.doi.org/10.1016/j.apsb.2015.07.003mailto:[email protected]://localhost/var/www/apps/conversion/tmp/scratch_1/dx.doi.org/10.1016/j.apsb.2015.07.003http://localhost/var/www/apps/conversion/tmp/scratch_1/dx.doi.org/10.1016/j.apsb.2015.07.003http://localhost/var/www/apps/conversion/tmp/scratch_1/dx.doi.org/10.1016/j.apsb.2015.07.003http://localhost/var/www/apps/conversion/tmp/scratch_1/dx.doi.org/10.1016/j.apsb.2015.07.003mailto:[email protected]://crossmark.crossref.org/dialog/?doi=10.1016/j.apsb.2015.07.003&domain=pdfhttp://crossmark.crossref.org/dialog/?doi=10.1016/j.apsb.2015.07.003&domain=pdfhttp://crossmark.crossref.org/dialog/?doi=10.1016/j.apsb.2015.07.003&domain=pdfhttp://localhost/var/www/apps/conversion/tmp/scratch_1/dx.doi.org/10.1016/j.apsb.2015.07.003http://localhost/var/www/apps/conversion/tmp/scratch_1/dx.doi.org/10.1016/j.apsb.2015.07.003http://localhost/var/www/apps/conversion/tmp/scratch_1/dx.doi.org/10.1016/j.apsb.2015.07.003http://www.sciencedirect.com/http://www.elsevier.com/locate/apsb
  • 7/25/2019 Insoluble Drug Delivery Strategies

    2/12

    1. Introduction

    The search for innovative medicines in disease management withoutcompromising on safety and efcacy is a challenge. In spite ofsignicant success in the discovery of new drugs, there are still unmetmedical conditions which need effective therapy. Market potential,competition among companies, dry pipeline of developmental candi-dates of various companies have hastened the drug discovery anddevelopment process. As a result, a signicant number of drugs gettingapprovals have poor biopharmaceutical properties. An estimated 40%of approved drugs and nearly 90% of the developmental pipeline drugsconsist of poorly soluble molecules1. Several marketed drugs sufferfrom poor solubility, low permeability, rapid metabolism and elimina-tion from the body along with poor safety and tolerability2.

    Recent studies have revealed that discovery and development ofnew drugs alone are not sufcient to achieve therapeutic excellenceand capture market economies3. Therefore, modied formulations ofexisting drugs are gaining more importance. The improved for-mulation of existing drugs is turning out to be lucrative business forpharmaceutical industry which is facing innovation decit thesedays for new molecules4. New dosage form, change of forms of

    drugs (ester/salt), prodrug/active metabolite of drug, different routesof administration are few changes that pharmaceutical companiesare exploring for 505(b)(2) llings5. Signicant number of insolubledrugs in the market provides protable strategies for pharmaceuticalcompanies to le NDA under 505(b)(2) with improved formulationsproviding faster dissolution and enhanced bioavailability. Hence thisreview summarizes various solubilization technologies. The recentadvances, clinical benets and business potentials of these technol-ogies are discussed in detail. The potential benets of insoluble drugdelivery technologies are depicted inFig. 1.

    2. Insoluble drug delivery technologies

    2.1. pH modication and salt forms

    Nearly 70% of drugs are reported to be ionizable, of which amajority are weakly basic. A pH-dependent solubility is exhibitedby ionizable drugs, wherein weakly acidic drugs are more soluble

    at pH4pKa (ionization constant) and weakly basic drugs aresoluble at pHopKa6. This pH dependent solubility was exploredextensively to formulate insoluble drugs. On the other hand, saltformation of weakly acidic or basic drugs provided alternatestrategies for formulation of drugs which have pH dependentsolubility. Pharmaceutically acceptable counter ions in the salt canprovide favorable pH conditions upon dissolution in water, andthus the pH of resulting solution would be close to maximum pHof drugs. Hence salt forms may sometimes avoid pH adjustmentsnecessary for solubilization of drugs. In addition, salt formationhas been reported to improve crystallinity, stability and pharma-ceutical processibility of drugs6.

    There are many insoluble drugs on the market which areformulated with pH modication technology. Ciprooxacin is aclassic drug which is weakly basic and practically insoluble inwater at neutral pH. However it exhibits pH-dependent solubilitywith higher solubility at acidic condition. Most of the intravenousformulations contain lactic acid as pH modier to improvesolubility7. Intravenous ciprooxacin infusions are essential fortreating different kinds of severe bacterial infections. Telmisartanis another drug which exhibits pH-dependent solubility. The

    currently marketed oral formulation of telmisartan contain alkalis,such as sodium hydroxide and meglumine for pH modication710.Telmisartan formulation marketed under brand name Micardiss ismanufactured using a expensive spray-drying process, whereindrug and alkalis along with other excipients are dissolved in waterand spray-dried to produce granules11. The spray-dried granulesobtained were reported to have a pH-independent dissolutionprole. However, generic versions of the telmisartan formulationare hard to come by, owing to the insoluble nature of the drug'sfree-acid-form and the critical steps involved in its manufacturingprocess that provided an additional market capitalization to theinnovator12.

    Repaglinide is an example of Zwitterion drug with poor watersolubility of 37mg/mL13. Currently repaglinide, marketed as

    Prandins in USA, is formulated with meglumine as pH modier.Various patents disclose the use of meglumine in the formulationand spray-drying as the process for preparing the granules1417.Tricky process and critical formulation sometimes prove to be hardto make generic copies. In case of both telmisartan and repagli-nide, actual salt forms of drugs are not used in the formulation,instead the bases such as meglumine and sodium hydroxide wereadded to the formulation. This could be due to technical reasons,such as lack of crystallinity, poor stability and deliquescent natureof resulting salts. On other hand, including bases in the formula-tion could be due to commercial reasons, in order to buildcomplexity in the process and product, such that it is hard tomake generic versions. These are a few examples of how a

    clinically and commercially bene

    cial drug product could belaunched in the market by altering the formulation strategies.Aspirin is century old non-steroidal anti-inammatory drug

    (NSAID), yet currently explored by various companies forcommercial benets. Soluble formulations of aspirin are currentlyavailable on the market. Aspro Clear, is soluble, effervescent tabletcontaining aspirin. The effervescence and favorable pH conditionrequired for solubility of aspirin are facilitated by incorporatingsodium bicarbonate and citric acid in the formulation. Aspro Clearreported to provide faster relief of pain than plain aspirin tablets18.This is another example, how insoluble drug formulation technol-ogy can be explored for commercial and clinical benets.

    Insoluble drugs are mostly formulated using the salt forms ofweakly acid and basic drugs. Various salt forms of drugs haveFigure 1 Benets of insoluble drug delivery strategies.

    Insoluble drug delivery strategies 443

  • 7/25/2019 Insoluble Drug Delivery Strategies

    3/12

    been the area of interest for pharmaceutical companies forcommercial and clinical benets. In the following section, fewexamples of such inventions are discussed. Identication ofbisulfate salt form of atazenavir is an interesting example ofhow salt screening could help a molecule to progress from beingdropped at preclinical development to clinical studies and nally tomarketing approval. Atazenavir as free base is practically insolublein water (o1mg/mL) and had poor oral bioavailability inpreclinical animal models19. Lack of sufcient absorption wasreported to be a hurdle in the development of this molecule. In aneffort to identify viable option to improve the bioavailability,series of salts were screened and nally atazenavir bisulfate wasselected for further development19,20. Atazenavir bisulfate exhib-ited distinct advantage over other salts such as methane sulfonateand hydrochloride in terms of solubility and solid state stability.Hydrochloride and methane sulfonate salts of atazenavir, whendissolved in water beyond saturation solubility of salts, there wassolid state transformation leading to dissociation of salt to freebase at pH4pHmax. Analysis of excess of solid in the suspensionrevealed that material was indeed free base. Under similarexperimental conditions bisulfate was found to be stable and did

    not convert to free base,and rather excess of solid was found to bein hydrated sulfate salt20. Therefore, the absolute bioavailability ofbisulfate was multifold-higher than the free base. This inventionnot only lead to superior protease inhibitor on the market butprovided additional patent protection and marketing exclusivity toinventor company.

    One of largest-selling anticancer drugs imatinib is marketed as asalt form, imatinib mesylate. The drug exhibits poor solubility andhence mesylate salt was used for its development, which is soluble inwater at pHo5.521,22. Among the two polymorphic forms (and ),generated by imatinib salt, the form is more stable with acceptablepharmaceutical properties. However, additional marketing rights wereassigned to the innovator due to their patent protection of the form23. Many old drugs have been reformulated as salt forms for

    commercial purposes. One such example is fenobrate, which wasapprovedin 1993 and was included in generic competition from theyear 200024. Since then, Abbott laboratories24 continuedling NDA'saltering the dose in order to gain market exclusivity. Interestingly, theactive form of all fenobrate formulations was found to be fenobricacid, an active metabolite of fenobrate which was responsible for thetherapeutic activity. This fact was well explored by Abbot anddeveloped cholin-fenobrate, a soluble and light-stable salt offenobric acid25. This salt form was developed into a delayedrelease capsule formulation and was approved by the FDA. Thisdelayed release formulation was proved to be one of blockbusterproduct in the recent time. In the times of innovation drought,such inventions are becoming huge commercial success thus

    improving overall investment drive in pharmaceutical research anddevelopment.The use of aspirin for clinical management of migraine was tested

    recently. The soluble aspirin-D,L-lysine salt was formulated forintravenous injection (IV). The clinical studies revealed that intrave-nous administration of aspirin was effective in relieving migraineattack. Although sumatriptan was slightly more effective than aspirinIV in headache relief, aspirin was well tolerated26,27. Hence the newsalt form of aspirin demonstrated safe, effective and affordablealternative therapy for the treatment of migraine. Similarly aspirin-calcium was utilized in the formulation of soluble tablet (Solorpins).The formulationshowed faster onset of action compared to the tabletwith plain aspirin28. Improved clinical benets, as well as commercialprots, were accomplished with these salt forms.

    Clopidogrel is an anti-platelet agent that works throughirreversible binding of its active metabolite to the P2Y12subtypeof adenosine diphosphate (ADP) chemoreceptors on platelets cellmembrane. Initially, it was available as Plavixs, consisting of thesalt form, clopidogrel bisulfate. However, other salt forms likeclopidogrel besilate and clopidogrel hydrochloride were approvedin Europe. In this case salt forms are explored by generic makerfor market exclusivity29. Recently FDA approved Advils, asodium salt of ibuprofen. This product is superior in terms of itsrapid onset of action as compared to Advil Liqui-Gels capsulescontaining ibuprofen30. Apart from enabling faster pain-relief topatients, this new salt form of ibuprofen provided marketexclusivity of at least 35 years for the manufacturer.

    Rosuvastatin (sparingly soluble) is available in the market as itscalcium salt. Recently generic-maker Watson pharmaceuticals, Inc.,gained approval for its NDA containing rosuvastatin zinc undersection 505(b)(2)31, thus getting marketing exclusivity more thantypical ANDA. However, the approval is subject to a court decisiondue to a legal petition led by AstraZeneca. Pharmaceutical companiesare continuously exploring the salt forms of drugs for better clinicalperformance. Sometimes it seems like reformulation is an alternative

    path for the pharmaceutical companies to exploit marketing exclusivityand captivity32. Further advancements in this technology will be moreinteresting, since there would be many more NDA's drugs to beapproved with new salt forms in the future.

    2.2. Co-solvency and surfactant solubilization

    Formulation of insoluble drugs using co-solvents is also one of theoldest and widely used technique, especially for liquid formulationintended for oral and intravenous administration. Reduction of thedielectric constant is possible by the addition of co-solvents, whichfacilitates increased solubilization of non-polar drug molecules. In orderto maximize the solubility and prevent precipitation upon dilution, co-

    solvents are used in conjunction with surfactants and pH modiers33,34.Taxol, an intravenous injection of paclitaxel, is the most

    debated formulation using this approach. This was developedusing 49% of dehydrated alcohol and 527 mg of cremophore EL35,which must be diluted before infusion. Additionally, pretreatmentof patients with antihistamines is essential owing to a hypersensi-tivity reaction due to higher content of cremophore EL in theformulation. Later, several formulations of paclitaxel excludingcremophore EL were attempted and couple of them gained FDAapproval after making a smooth means of access through clinicaltesting. Formulations devoid of cremophore EL included Abraxane(albumin microspheres containing Paclitaxel)and Genexol (PEG-PLA polymeric micelles with Paclitaxel)36,37.

    Similarly, docetaxel is another widely used anticancer drug andthe original formulations of taxotere contains ethanol and Tween80 to solubilize the drug (0.54 g polysorbate 80 and 0.395 gdehydrated alcohol)38. However, hypersensitivity reactions usingthis product were reported due to the surfactants in the formula-tion. Sandoz, Inc., Hospira Inc. and Apotex Inc. each hasdocetaxel containing a new drug product approved under section505(b)(2)3941. Most of the new formulations have PEG 300 asadditional cosolvent and Tween 80 content signicantly less thantaxotere. These new formulations were claimed to be safer andstable than taxotere. Insoluble drug delivery technology utilizingthe co-solvent-surfactant approach had indeed proved vital inproviding an effective treatment option for cancer patients. Furtherimprovement in the formulation of taxol's resulted in more patient

    Sandeep Kalepu, Vijaykumar Nekkanti444

  • 7/25/2019 Insoluble Drug Delivery Strategies

    4/12

    compliance and new intellectual properties for pharmaceuticalcompanies. A list of pharmaceutical formulations containing thehighest amounts of co-solvents and surfactants are provided in

    Table 1 42. Though co-solvency and surfactant solubilizationtechniques are widely used for enhancing the solubility ofhydrophobic drugs, they have some disadvantages: tolerability offormulations with high levels of synthetic surfactants may be poorin cases where long term chronic administration is intended;uncontrolled precipitation may occur upon dilution with aqueousmedia or physiological uids. Precipitates may be amorphous orcrystalline and can vary in size; precipitation of drug from a co-solvent mixture may result in embolism and local adverse effectsat the injection site; concomitant solubilization of other ingredientssuch as preservatives may lead to consequent alteration in stabilityand effectiveness of the drug product.

    2.3. Amorphous forms, solid dispersions and cocrystals

    Stable crystal forms of drugs pose problem in solubilization due tohigh lattice energy. Thus, disordered amorphous forms offer distinctadvantage over crystal forms with regards to solubility. Hence,changing the solid state characteristics of active pharmaceuticalingredient (API) renders the molecule more water soluble. But,excess of enthalpy, entropy and free energies of amorphous formsmakes them prone to crystallization, leading to the formation of stablecrystals43. However, the advent of new techniques to improvestability of amorphous forms improved chances of their use inpharmaceutical formulations44. Complicated process of makingamorphous drug systems and various factors affecting the stability

    of those forms resulted in reduced generic competition for alreadyapproved amorphous products. Cefuroxime axetil practically wasinsoluble in water and introduced as Ceftins by GSK in amorphousform and was protected by a couple of patents, which barred the entryof generic players for a reasonable period45,46. Another drug product,the amorphous zarlukast is available commercially as Accolates.The amorphous form issubject to various patents which precludedearly generic entry47,48. Amorphous forms of other drugs likenelnavir mesylate, quinapril hydrochloride and rosuvastatin calciumare also commercially available in the market.

    Solid dispersion technology was extensively explored in recentdecades for the delivery of insoluble drugs. Physically, soliddispersions are eutectic mixtures or solid solutions in which drugs

    exist either in an amorphous form dispersed in the carrier or as amolecular dispersion in the carrier4951. Solid dispersions favorenhanced dissolution of drugs due to the formation of a high-

    energy amorphous form or increased solubility leading to super-saturation. The increased solubility can be attributed to the dispersionof drugs at the molecular level and/or solubilization effects of thepolymer. The drug remains in a metastable form for considerable timein the supersaturated state and polymeric carrierin turn can stabilizethe metastable state by preventing nucleation51. Advances in melt-extrusion and spray-drying have accelerated industrial applications ofsolid dispersions for the delivery of insoluble drugs.

    Sporanoxs is a classic example of a drug (itraconazole)formulated using solid dispersion technology. At neutral pHitraconazole has a negligible solubility of 1 ng/mL52. For preparingsolid dispersions of itraconazole, spray-layering technology wasused in which an organic solution of drug and hydroxylpropyl

    methylcellulose (HPMC) was sprayed over sugar beads to form athin lm consisting of molecularly dispersed drug and polymer.This amorphous formulation signicantly enhanced bioavailabilitycompared to crystalline itraconazole. Apart from spray layering,itraconazole solid dispersions were also prepared using hot-meltextrusion with varying polymers such as HPMC, Eudragit andpolyvinyl pyrrolidone (PVP) mixture. In vitro studies revealed afaster dissolution of solid dispersions containing Eudragit incomparison to HPMC and sporanox52. In contrast, clinical studiesrevealed a similarity between solid dispersions containing HPMCand sporanox, which can be attributed to the solubilization andstabilization effects of HPMC in physiological conditions (in vivo).

    A list ofcurrently marketed solid-dispersion products is shownin Table 2 51. All the listed products have generated clinically

    benecial results by producing adequate drug levels in the body atdesired therapeutic concentration, leading to improved bioavail-ability. Apart from potential clinical benets, these products havegenerated considerable intellectual property and commercial suc-cess to the manufacturer.

    Pharmaceutical cocrystal technology has received greater atten-tion in the last decade owing to its successful delivery of insolubledrugs. Stoichiometric solids of drug and conformer (secondcomponent), which exist as crystals at ambient temperature arereferred to as cocrystals. Non-covalent forces like acidamide,acidacid, and amideamide interactions, usually of hydrogenbonding nature, hold the drug and conformer together in thecocrystal. The enhanced solubility of drug in cocrystal is achieved

    Table 1 List of parenteral drug formulations containing co-solvents and surfactants.

    Solvent Percentage in marketed formulation(%)

    Percentage administered(%)

    Route ofadministration

    Example

    Cremophor EL 1165 r10 IV infusion PaclitaxelCremophor RH 60 20 r0.08 IV infusion TacrolimusDimethylacetamide

    (DMA)

    6 r3 IV infusion Teniposide

    Ethanol 580 r6 SC DihydroergotamineGlycerin 1532 r15 IM, SC, IV Dihydroergotamine

    N-methyl-2-pyrrolidone 100 100 Subgingival DoxycyclinPEG 300 r60 r50 IM, IV bolus MethocarbamilPEG 400 1867 r18 IM LorazepamPolysorbate 80 0.075100 r4 IM ChlordiazepoxidePropylene glycol 1080 r80 IM LorazepamSolutol HS-15 50 50 IV Propanidid

    IM: intramuscular; IV: intravenous; PEG: polyethylene glycol; SC: subcutaneous.

    Insoluble drug delivery strategies 445

  • 7/25/2019 Insoluble Drug Delivery Strategies

    5/12

    by lower lattice energy and higher solvent afnity53. Any of thegenerally regarded as safe (GRAS)-listed excipients, organic acids(such as fumaric acid, malic acid, glutaric acid, succinic acid,oxalic acid), nutraceuticals (such as pterostilbene, quercetin, p-coumaric acid and saccharine) can act as a conformer. Co-crystaltechnology has been explored for solubility enhancement of drugslike itraconazole, carbamazepine, gabapentinin, modanil, piroxi-cam, caffeine, etc.53. The cocrystal technology have been used tocreateintellectual property and large number of patents have been

    led54. However there is no approved product with drug cocrys-tals, with enormous potential for delivery of insoluble drugs tilltoday, but the future of cocrystals is promising.

    2.4. Polymeric micelles

    Water insoluble drugs often have greater afnity for hydrophobicsolvents because of hydrophobichydrophobic interactions andalso have afnity for hydrophobic region of micelles. Henceencapsulation of those drugs in micelles enables their formulationin aqueous vehicle. Initially the hydrophilic surfactants were usedto solubilize the drug for oral and intravenous administration.However, limited solubilization, higher critical micellear concen-

    trations (CMC) and potential adverse events after intravenousadministration have limited their application. Polymeric micelleson other hand, offer greater advantage in terms of solubilizationcapacity, lower CMC and greater tolerability. Polymeric micellesare formed using diblcok polymers such as PEG-PLA or triblockpolymers PLA-PEG-PLA. The PEG is usually the hydrophiliccomponent in the polymer for micelles and hydrophobic chain canbe of poly lactic acid, poly aspartic acid, polycaprolactic acid,etc.37,55. Due to low CMC, the polymeric micelles remain stable atlow polymer concentration after dilution with body uids. Thenano-sized nature of polymeric micelles provides opportunity fortumor-targeting via enhanced permeation and retention effect(EPR). The hydrophilic PEG surface makes micelles less suscep-

    tible for reticulo-endothelial scavenging, and thus drugs havelonger circulation time. Polymeric micelles can also be tailored forpH-responsive release of drugs at specic tissues and for active-targeting using targeting ligands37.

    The Genexol-PM is polymeric micelles comprising of PEG-(D,L-lactide) polymer with paclitaxel encapsulated in the micelles.This is the rst polymeric micelle formulation approved by FDA56

    and is reported to be superior in terms of safety and tolerabilitycompared to other marketed formulations (ethanol/CremophoreEL). The pluronics-based polymeric micelles containing doxoru-bicin (SP1049C) are currently in phase III clinical trial and havebeen granted orphan status by FDA. Another paclitaxel polymericmicelles (NK105) and cisplatin micelles are in phase II clinicaltrials37. The polymeric micelles for delivery of insoluble drugs,

    especially parenteral formulations can offer intellectual propertyfor companiesand better treatment options for the patients in needof those drugs37. Table 3 presents the representative list of drug-loaded polymeric micelles products and their progress57,58.

    2.5. Inclusion complexation

    Cyclodextrins (CD) are the versatile excipients studied extensively

    for pharmaceutical applications59

    . These are cyclic oligosacchar-ides consisting of glucopyranose units that are united via 1,4-linkage. Three major types of CDs include ,and , varying with6, 7 and 8 glucopyranose units, respectively. CDs have atruncated-cone structure with a hydrophobic interior and a hydro-philic exterior due to the cyclic orientation of pyranose units.Central cavity of cyclodextrin is hydrophobic due to skeletalcarbon atoms and ethereal oxygen. Polarity of cavity is estimatedto be somewhere close to aqueous ethanolic solution59. Thehydrophobic nature of cavity enables entrapment of hydrophobicmolecules of suitable size inside the cavity and hydrophilic surfaceof CD makes complex soluble in water. Apart from solubilization,cyclodextrins are also used for drug stabilization, drug protectionfrom light, thermal and oxidative stress, taste masking of drugs,

    and reduced dermal, ocular or gastrointestinal irritation.The relative size of CD to the guest molecule, the presence of

    key functional groups on the guest molecule, and thermodynamicinteractions between CD, guest molecule and solvent are the keyfactors that enable the formation of an inclusion complex. Inaddition to natural CDs, insoluble drugs are formulated usingsynthetic CDs like hydroxy propyl--cyclodextrins, hydroxypropyl--cyclodextrins and sulfobutyl cyclodextrin (Captisols),since the latter have higher solubility and safety proles whencompared to the former59,60.

    The use of cyclodextrins in the formulation has enabled manyproduct containing insoluble drugs to reach the market andeventually helped to treat many life-threatening disease conditions.

    Recently cyclodextrins are explored to reformulate existing drugsfor better clinical applications and also for revenue generation viaNDAs under section 505(b)(2). There were several cyclodextrincontaining drug products in Japan, Germany and other Europeancountries; however, Janssen Pharmaceuticals, Inc. was the rstcompany to get US FDA approval for its antifungal drug product(sporanox oral and IV solution) containing itraconazole with 40%of hydroxy propyl--cyclodextin in the year 199958. This wasproved to be huge commercial successes for Janssen Pharmaceu-ticals, Inc. and their efforts in nding modied CD were paid off.The introduction of spornox oral solution lead to effectivetreatment of fungal throat infections and intravenous formulationfor severe systemic fungal infections. The intravenous formula-tions of ziprazidone mesylate and voriconazole formulated with

    Table 2 List of marketed products in United States utilizing solid dispersion technology.

    Drug Brand name Carrier Manufacturer Year of FDA approval

    Itraconazole Sporanoxs HPMC Janssen Pharmaceuticals, Inc., USA 1992Tacrolimus Prograf s HPMC AstellasPharma, US Inc. 1994Lopinavir/Ritonavir Kaletras PVP/VA Abbot Labarotaries, USA 2005Nabilone Casamet s PVP Meda Pharmaceuticals Inc., USA 2006

    Nimodipine Nimotops

    PEG Bayer (Pty) Ltd., USA 2006Fenobrate Fenoglides PEG/Poloxamer Santarus, Inc. 2007Etravirine Intelences HPMC Janssen Therapeutics, USA 2008

    Sandeep Kalepu, Vijaykumar Nekkanti446

  • 7/25/2019 Insoluble Drug Delivery Strategies

    6/12

    sulphobutyl cyclodextrin are available in many countries includingUSA. The reformulation of existing drugs using cyclodextrin hasbeen explored to get approval of new drug applications withgreater marketing exclusivity and patent protection. Drugs such asaripriprazole, mitomycine, diclofenac sodium, chlodizepoxide,meloxicam, alfaxalone, cisapride, indomethacine, insulin (nasalspray) and omeprazole have been reformulated using cyclodextrinsfor both commercial and health benets61.

    2.6. Size reduction and nanonization

    Over the past two decades, nanoparticle technology has become awell-established and proven formulation approach for poorly-solubledrugs. Reducing a drug's particle size to sub-micron range is referredto as nanonization. In the eld of pharmaceuticals, the termnanoparticleis applied to structures less than 1 m in size. Higherintracellular uptake of nanoparticles due to their sub-micron-size

    range offers a distinct advantage over microparticles. Nanoparticlesoffer a potential opportunity to overcome the challenges associatedwith the formulation of insoluble drugs. Drug nanoparticles can beproduced by various technologies, which can be broadly categorizedinto bottom up and the top downtechnologies.

    In bottom-up technologies controlled precipitation of the solubi-lized drug is achieved by adding a suitable non-solvent. Hydrosoldeveloped by Sandoz (presently Novartis) is an example fornanoformulation prepared using the precipitation technique6264.In this process, the drug is dissolved in a solvent and this solution issubsequently added to a non-solvent solution. This results in highsupersaturation, rapid nucleation and the formation of many smallnuclei65. Upon solvent removal, the dispersion can be ltered and

    lyophilized to obtain amorphous nanocrystals having a highsolubility and dissolution rate.High-pressure homogenization and milling methods are the

    alternate technologies that are frequently used for producing drugnanoparticles. However, a combination approach, with a pre-processing step followed by size reduction is also in application.Supercritical uid technology is another approach for nanosizing,but it is industrially less successful when compared to theaforementioned technologies.

    In early 19th century, heterogeneouscatalysts were rst amongthe reported techniques for nanosizing66,67. Preclinical studies ofdanazol nanosuspension with a median diameter of 169 nmshowed enhanced oral bioavailability 82.3710.1%, when com-pared to conventional as-isdrug suspension 5.171.9%68,69. Fine

    particles of atovaquone in the range of 100300 nm have beensuccessfully produced using the homogenization (microuidiza-tion) technique. Following oral administration, the nanoparticleformulation enhanced the drug concentration in plasma from 15%to 40% in comparison to micronized Wellvones, at equivalentdoses. These results reect the potency of the nanonizationtechnique in terms of reducing drug load from 22.5 mg/kg (Well-vones) to 7.5 mg/ kg, and increasing the activity 2.5-fold70.

    In August 2000, the rst product incorporating the NanoCrystals

    technology was approved by the US FDA. Wyeth's Rapamunes

    (sirolimus, an immunosuppressant) developed using similar technol-ogy captured the market after its approval. Rapamunes was marketedas an oral solution and stored at refrigerated condition. The oralsolution was given with orange juice prior to dosing. The develop-ment of a NanoCrystals dispersion of sirolimus provided a drugproduct with enhanced bioavailability and improved stability.

    In April 2003 an antiemetic drug, Emends (aprepitant, MK

    869) was approved and introduced into the market. Emend iscapsule dosage form containing 80 or 125 mg of aprepitantformulated as drug nanoparticles. Following oral administration,the nanosuspension was able to overcome the signicant foodeffect observed with the microsuspension formulation. Abraxanes

    (a reformulation of paclitaxel) is a nanoparticle-based product andwas approved by FDA in 2006 for intravenous administration. It isa novel formulation consisting of lyophilized particles with 10%(w/w) paclitaxel and 90% (w/w) albumin71. The particle size of thenanosuspension is about 130 nm. The maximum tolerated doseobserved from this study was higher than the commercial Taxols

    formulation. Further studies conrmed that the nanoparticleformulation eliminated the need for premedication (since the toxicexcipient Cremophor EL was not used in the formulation). Studiesfrom intravenous and pulmonary applications of nanoparticlesreported good tolerability andprovided an alternative solution toinsoluble drug therapeutics72,73. A list of marketed products usingdrug nanoparticles is summarized inTable 4 74.

    Nanoparticle technology serves as a screening aid duringpreclinical efcacy and safety studies of new chemical entities(NCEs). Fabrication of existing drugs with maximal drug expo-sure, less toxicity, expanded intellectual property by drug life cyclemanagement and minimized competition during the drug's lifetime can be achieved through nanoparticle-based drug deliverysystems. In fact, viable formulations for poorly soluble drugs withmaximum drug exposure can be developed potentially by nano-particle technology, which has opened the stage gates for reviving

    Table 3 Representative list of drug-loaded polymeric micelles-based products.

    Product Incorporated drug Status Company

    Genexol PM Paclitaxel Marketed SamyangEstrasorb Estrogen Marketed NovavaxMedicelle DACH-platin-PEG-polyglutamic acid phase I/II NanoCarrier Flucide Anti-inuenza phase I/II Nano Viricides

    Basulin Insulin phase II/III Flamel TechnologiesDO/NDR/02 Paclitaxel phase I/II Dabur Research FoundationNK-911 Doxorubicin phase II Nippon Kayaku Co.NK-105 Paclitaxel phase II/III Nippon Kayaku Co.NK-012 SN-38 phase II Nippon Kayaku Co.NC-6004 Cisplatin phase III Nanocarrier Co.NC-4016 Oxaliplatin phase I/II Nanocarrier Co.SP-1049C Doxorubicin phase II/III SupratekPharma Inc.NC-6300 Epirubicin phase I/II Nanocarrier Co.

    Insoluble drug delivery strategies 447

  • 7/25/2019 Insoluble Drug Delivery Strategies

    7/12

    currently marketed products with suboptimal drug delivery,

    leading to better clinical and commercial bene

    ts. Some of thekey nanotechnology-based approaches for improving the oralbioavailability of poorly water-soluble drugs (according toSafe-Siebert and co-workers75) are highlighted inTable 569,7681.

    2.7. Solid lipid nanoparticles

    Solid lipid nanoparticles (SLN) are promising drug carriers withpotential applications in the delivery of poorly soluble drugs82,83.The lipid excipients used in the SLN formulations are biocompatibleand biodegradable and most of them are physiological components thatare generally regarded as safe (GRAS). Site-specic drug delivery,particularly for poorly soluble proteins and peptide drugs could be

    achieved by exploring SLN technology84. A signicant increase in

    bioavailability was achieved when a poorly soluble compound,ooxacin was formulated as SLN85. The enhancement in drug'sbioavailability is attributed to the increase in surface area of theparticles, improved dissolution rate and enhanced concentration ofooxacin in gastrointestinal tract (GIT) uids86,87. The drug in lipidnanoparticles may adhere to the intestinal wall and thereby increasesthe drug residence time in the GIT, resulting in improvedbioavailability88.

    Pandita et al.89 developed an SLN formulation for a poorly solublecompound, paclitaxel. Improved oral bioavailability as compared tothe control group was observed with the in vivo studies of SLNformulation. Studies also revealed an improved dissolution rate ofpoorly soluble drugs such as camptothecin, vinpocetineand feno-brate by their successful incorporation into SLNs90,91. Controlled

    Table 4 Overview of nanoparticle technology based marketed products.

    Trade name Drug Indication Drug delivery company Innovator company

    Rapamunes Rapamycin, sirolimus Immunosuppressant ElanNanosystems WyethEmends Aprepitant Anti-emetic ElanNanosystems Merck & Co.Tricors Fenobrate Hypercholesterolemia Abbott Laboratories Abbott laboratoriesMegace ESs Megestrol Anti-anorexic ElanNanosystems Par Pharmaceuticals

    Triglides

    Fenobrate Hypercholesterolemia IDD-P Skyepharma ScielePharma Inc. KingAvinzas Morphine sulfate Phychostimulant drug ElanNanosystems PharmaceuticalsFocalin Dexmethyl-phenidate HCl Attention decit hyperactivity

    disorder (ADHD)ElanNanosystems Novart is

    Ritalin Methyl phenidate HCl CNS stimulant ElanNanosystems NovartisZanaex Capsules Tizanidine HCl Muscle relaxant ElanNanosystems Acorda

    Table 5 Key nanotechnology-based approaches for the enhancement of drug solubility and oral bioavailability.

    Company Nanotechnology-based formulation approach Description and reference

    American Biosciences(Blauvelt, USA)

    Nanoparticle albumin-bound technology. e.g. paclitaxel-albumin nanoparticles Paclitaxel albuminnanoparticles76

    BaxterPharmaceuticals(Deereld, USA)

    Nanoedge technology: particle size reduction was achieved by homogenization,micro-precipitation, lipid emulsion and other dispersed systems.

    Nano-lipid emulsion77

    BioSantePharmaceuticals(Lincolnshire, USA)

    Calcium phosphate based nanoparticles were produced for improved oralbioavailability of hormones/proteins and vaccine adjuvants

    Calcium phosphatenanoparticles78

    ElanPharmaInternational(Dublin, Ireland)

    Nanoparticles (o1mm) were produced by Wet milling technique usingsurfactants and stabilizers. The technology was applied successfully indeveloping apprepitant and reformulation of Sirolimus.

    Nanocrystaldrugparticle79

    EurandPharmaceuticals(Vandalia, USA)

    Nanocrystal or amorphous drug is produced by breakdown of crystal lattice andstabilized by using biocompatible carriers (swellablemicroparticles orcyclodextrins)

    Cyclodextrinnanoparticle80

    iMEDDInc(Burlingame, USA)

    Implantable drug delivery system using silicon membrane with nano-pores(10100 nm)

    Stretchable siliconnanomembrane81

    pSivida Ltd.(Watertown, USA) The solubility and bioavailability of hydrophobic drugs was achieved byincorporating drug particles within the nano-width pores of biocompatiblesilicon membranes or bers.

    Silicon nanoparticles

    PharmaSol GmbH(Berlin, Germany)

    High pressure homogenization was used to produce nanostructured lipidparticles dispersions with solid contents that provide high-loading capacityfor hydrophilic drugs

    Drug encapsulated in lipidnanoparticles69

    SkyePharmaPlc,(Piccadily, London,UK)

    Nanoparticulate systems of water insoluble drugs were produced by applyinghigh shear or impaction and stabilization was achieved by usingphospholipids.

    A polymer stabilizing nano-reactor with theencapsulated drugcore69

    Sandeep Kalepu, Vijaykumar Nekkanti448

  • 7/25/2019 Insoluble Drug Delivery Strategies

    8/12

    release of drugs in the GIT with improved bioavailability bydecreasing the variability in absorption can be achieved by thesecarrier systems. Apart from these, avoidance of organic solutions,increased drug stability in GIT and feasibility to scale-up are a few ofthe potential therapeutic benets of solid lipid nanoparticles. How-ever, a product with SLN is yet to hit the market. A list of examplesof drugs developed using SLN technology and their biopharmaceu-tical applications are summarizedTable 6.

    2.8. Liposomes and proliposomes

    Liposomes are spherical closed vesicles of phospholipid bilayers withan entrapped aqueous phase, and may consist of one or more bilayers.Liposomes were rst prepared by A.D. Bangham in the early 1960sand demonstrated that a wide variety of molecules can be encapsulatedwithin aqueous spaces of liposomes or inserted into their membranes.Liposomes have been regarded as new drug delivery systems capableof transporting drug molecules to specic target site with enhancedefcacy and safety92. A potential advantage of liposomes is theencapsulation of hydrophobic as well as hydrophilic drugs, either inthe phospholipid bilayer, at the bilayer interface or in the entrappedaqueous volume. Recent developments in liposome technology aregenerating more effective strategies for improving the vesicle stabilityafter systemic administration93,94.

    Liposomal drug delivery offer signi

    cant therapeutic bene

    ts topoorly soluble compounds. One such example is the formulation ofcyclosporine and paclitaxel in which surfactants and organic co-solvents are used for systemic administration in humans. Thesesolubilizers may cause toxicity at the administered doses. Incomparison, liposomes are relatively non-toxic, non-immunogenic,biocompatible and biodegradable molecules, which can encapsulatea wide range of water-insoluble (lipophilic) compounds. Paclitaxelliposomes were able to deliver the drug systemically and increasethe therapeutic index of paclitaxel in human ovarian tumormodels95,96. Currently, liposomes are being used as excipients forpreparing better-tolerated clinical formulations of several lipophilic,sparingly water soluble drugs such as amphotericin B97. Developingliposome drug delivery improved solubility of lipophilic and

    amphiphilic drugs such as porphyrins, minoxidil, peptides andanthracyclines, respectively. Furthermore, in some cases anticanceragent such as acyclovir can be encapsulated in liposome interior atconcentrations above their aqueous solubility98. A representative listof liposomal based drug delivery products is summarized inTable 7.

    Proliposomes are dry, free owing powders which can formmultilamellar vesicles (MLVs) upon hydration with water. Prolipo-somes have been extensively studied as a potential carrier for oraldelivery of drugs with poor bioavailability99. It provides a novelsolution to product stability problems associated with the storage ofaqueous liposomal dispersions, wherein it produces a dry product that

    can be stored for long duration and hydrated immediately beforeuse100. Liposomes are either formed in vivo upon contact with thephysiological uids or prepared in vitrobefore administration using ahydrating solvent. The liposomes formed upon hydration are similar toconventional liposomes with uniform vesicle size101,102.

    Indomethacin proliposomes for oral administration werereported by Katare et al.103, in which the efcacy of the oralformulation was studied by measuring ulcerogenic index and anti-inammatory activity using carrageenan-induced paw edema testin rats. The liposomal formulation showed enhanced performancein vivowith reference to their cytoprotective and anti-inammatoryproperties.

    Greater efcacy and less toxicity were reported by encapsulat-

    ing vinpocetine in proliposomes. The study showed that the oralbioavailability of proliposomes was enhanced in New Zealandrabbits and thereby provided a new delivery platform to enhancethe absorption of poorly soluble drugs in the GIT104. Therapeuticbenets of proliposomes include enhanced bioavailability, protec-tion of drugs from degradation in the GIT, reduced toxicity andtaste masking. The proliposomes can also provide target drugdelivery and controlled drug release.

    2.9. Microemulsions and self-emulsifying drug delivery systems

    Micro-emulsions are thermodynamically stable, isotropic mixturesof oil, water, surfactant and a co-surfactant. In comparison to

    Table 6 List of examples of drugs developed using solid lipid nanoparticle technology.

    Drug Lipid used Biopharmaceutical application

    5-Fluoro uracil Dynasan 114 and Dynasan 118 Prolonged release in simulatedcolonic media

    Apomorphine Glycerylmonostearate, polyethylene glycol monostearate Enhanced bioavailability in ratsCalcitonin Trimyristin Improvement of the ef cacy of

    proteinsClozapine Trimyristin, Tristearin and Tripalmitin Improvement of bioavailabilityCyclosporin A Glycerylmonostearate and glycerylpalmitostearate. Controlled releaseGonadotropin release

    hormoneMonostearin Prolonged release

    Ibuprofen Stearic acid, Triluarin and Tripalmitin Stable formulation with lowtoxicity

    Idarubicin Emulsifying wax Delivery of oral proteinsInsulin Stearin acid, octadecyl alcohol, cetylpalmitate,

    glycerylpalmitostearate, glyceryltripalmitate,glycerylbehenate and glycerylmonostearate.

    Potential for oral delivery ofproteins.

    Lopinavir Campritol 888 ATO Bioavailability enhancedNimusulide Glycerylbehanate, palmitostearate, glyceryltristearate Sustained release of drugProgesterone Monostearin, stearic acid and oleic acid Potential for oral drug deliveryRepaglinide Glycerylmonostearate and tristearin Reduced toxicity

    Tetracycline Gycerylmonostearate and stearic acid Sustained release

    Insoluble drug delivery strategies 449

  • 7/25/2019 Insoluble Drug Delivery Strategies

    9/12

    conventional emulsions, micro-emulsions produce a clear emulsionon mild agitation. The advantage of micro-emulsions over conven-tional emulsion and solution formulations is that the former

    produces a stable heterogeneous system. Micro-emulsion technol-ogy is widely used to address the challenges associated with poorlysoluble compounds. Insoluble drugs can be administered throughparenteral route by formulating into micro-emulsions. The micro-emulsions for parenteral delivery comprise lipid droplets (10%20%), osmotic agent and an emulsier. Apart from these, anantimicrobial agent is incorporated if the emulsion is packed in amulti-dose container. Propofol injection is a classic example ofparenteral microemulsion formulation. Initially, Cremophore ELwas used for formulating propofol, and then ethanol was includedby changing the formulation. Finally, it was introduced into marketby formulating into a microemulsion with soybean oil105, having ahigher tolerable limit and safety prole. The representative list ofmarketed parenteral microemulsions is summarized inTable 8.

    Self-emulsifying drug delivery systems have gained importanceowing to theirability to enhance solubility and bioavailability ofinsoluble drugs106. Upon dilution by the aqueous environment in the

    GIT, these systems undergo rapid self-emulsi

    cation producingnano-sized globules of high surface area resulting in enhanced rateand extent of absorption with consistent plasma time proles. Anexample of drug product developed using self-micro-emulsifyingdrug delivery system (SMEDDS) is Neorals, an oral cyclosporineformulation which forms micro-emulsion in aqueous environment.The drug product showed improved bioavailability from 174%239% as compared to cyclosporine-A, Sandimmunes107.

    There are many examples and studies involving self-emulsifyingsystems for improving the in vitro and in vivo performances ofpoorly soluble drug candidates108. A signicant enhancement in thebioavailability was observed with vinpocetin and atorvastatin inself-emulsifying systems as compared to their conventional tabletformulation, indicating the criticality of surfactant concentration in

    Table 7 Representative list of liposomal based drug products.

    Product Drug Company Indication target

    Atragen Tretinoin Aronex Pharmaceuticals Inc. Acute myeloid leukemiaAmphotec Amphotericin B Sequus Pharmaceutical Inc. Fungal infections leishmaniasisAmbisome Amphotericin B NeXstar Pharmaceutical Inc. Co. Serious fungal infect ionsAmphocil Amphotericin B Sequus Pharmaceutical Inc. Serious fungal infections

    Abelcet

    Amphotericin B The Liposome Company, Inc. Serious fungal infectionsALEC Dry protein free powder of

    DPPC-PGBritannia Pharmaceuticals Ltd. Expanding lung diseases in infants

    Avian retrovirusvaccine

    Kil led avian retrovirus Vineland Laboratories, USA Chicken pox

    DaunoXome Daunorubicin citrate NeXstar Pharmaceutical Inc., Co. Kaposi sarcoma in AIDSDepoDur Morphine Pacira Pharmaceuticals Inc. Post-surgical pain reliever DaunoXome Daunorubicin citrate Galen Ltd. Kaposi sarcoma in AIDSDepocyt Cytarabin Pacira Pharmaceuticals Inc. Treatment of lymphomatous

    meningitisDoxil Doxorubicin SequusPharmaceutical Inc. Kaposi sarcoma in AIDSEstrasorb estradiol Novavax Menopausal therapyEvacet Doxorubicin The liposome company, USA Metastatic breast cancer EpaxalBernas Vaccine Inactivated hepatitis-A

    VirionsSwiss serum & vaccine institute,Switzerland.

    Hepatitis A

    Fungizone Amphotericin B Bristol-Myers Squibb, Netherland Serious fungal infectionsMiKasomes Amikacin NeXstar Pharmaceutical Inc., Co. Bacterial infectionNyotran Nystatin Aronex pharmaceuticals Inc. Systemic fungal infectionsTopex-Br Terbutalinesulphate Ozone Pharmaceuticals Ltd. AsthmaVentus Prostoglandin-E1 The liposome company, Inc. Systemic inammatory diseaseVincaXome Vincristine NeXstar Pharmaceutical Inc., Co. Solid tumors

    Table 8 Representative list of marketed parenteral microemulsion products.

    Drug Product name Company Therapeutic area

    Cyclosporine A Restasis Allergan Immunomodulation

    Diazepam Diazemuls Braun Melsungen SedationDexamethazonePalmitate Limethason Green Cross CarticosteroidEtomidate Etomidat Dumex (Denmark) AnesthesiaFlurbiprofen Lipfen Green Cross AnalgesiaProstaglandin-E1 Liple Green Cross Vasodilator Propofol Propofol Baxter Anesthesia Anesthesia

    Diprivan AstraZeneca AnesthesiaPerurodecalinPerurotripropylamine Fluosol-DA Green Cross AnalgesiaVitamins A, D, E and K Vitalipid Kabi Nutrition

    Sandeep Kalepu, Vijaykumar Nekkanti450

  • 7/25/2019 Insoluble Drug Delivery Strategies

    10/12

    formulation for yielding the smaller particles with concomitantenhancement in drug permeation and absorption109,110. The mar-keted oral products which yield an emulsion or micro-emulsion inthe gastrointestinal tract are summarized in Table 9.

    3. Conclusions

    A great opportunity as well as potential challenge is foreseen from thelarge number of insoluble drugs that are approved by FDA, as well asthose in the developmental pipeline. Exploring recent advances ofinsoluble drug delivery technologies will help in better therapeuticapplications with improved patient compliance. On the other hand, the

    insoluble drug delivery technologies are being effectively utilizedpredominantly for commercial benets through NDA route bydeveloping improved formulations. Therefore, further advancement inthe insoluble drug delivery technologies and their exploration for newdrug applications will be much more promising in coming years.

    References

    1. Loftsson T, Brewster ME. Pharmaceutical applications of cyclodex-trins: basic science and product development. J Pharm Pharmacol2010;62:160721.

    2. Hodgson J. ADMETturning chemicals into drugs. Nat Biotechnol2001;19:7226.

    3. The 505(b)(2) blog. 2012 approvals. Available from: http://blog.

    camargopharma.com/index.php/scorecard/2012-approvals.4. Drews J, Ryser S. Innovation decit in the pharmaceutical industry.

    Drug Inf J1996;30:97108.5. US Department of Health and Human Services, Food and Drug

    Administration, Center for Drug Evaluation and Research. Guidancefor industry, applications covered by section 505(b)(2). Rockville,MD 20857: 1999.

    6. Serajuddin AT. Salt formation to improve drug solubility.Adv DrugDeivl Rev 2007;59:60316.

    7. http://www.rxlist.com/cipro-iv-drug.htm.8. http://www.rxlist.com/micardis-drug.htm.9. Hauel N, Narr B, Ries U, Van Meel JC, Wienen W, Entzeroth M,

    inventors; Karl Thomae GmbH, assignee. Benzimidazoles useful asangiotensin-11 antagonists. United States Patent US 5591762.Januray 71997.

    10. Heinrich S, Schneider M, inventors; Schneider Heinrich, SchneiderMargarete, assignee. Polymorphs of telmisartan. United States Patent

    US 6358986. 2002 July 18.11. Giovanni G. Antihypertensive medicaments containing lacidipine and

    telmisartan. WIPO Patent No. WO/2000/027397A. 2000 May 18.12. Nakatani M, Ohki T, Takeshi S, Toyoshima K, inventors; Boehringer

    Ingelheim International GmbH, assignee. Solid pharmaceutical for-

    mulations comprising Telmisartan. European Patent EP 1545467.

    2007 December 9.13. Mandi Z, Gabelica V. Ionization, lipophilicity and solubility

    properties of repaglinide. J Pharm Biomed Anal2006;41:86671.14. Wolfgang G, Hurnaus R, Griss G, Sauter R, Reiffen M, Rupprecht E,

    inventors; Boehringer Ingelheim KG, assignee. Phenylacetic acid

    benzylamides. United States Patent US RE 37035. 2001 January 30.

    15. Wolfgang G, Hurnaus R, Griss G, Sauter R, Reiffen M, Ruprecht E,inventors; Karl Thomae GmbH, assignee. Phenylacetic acid benzy-

    lamides. United States Patent US 5312924. 1994 May 17.16. Wolfgang G, Hurnaus R, Griss G, Sauter R, Reiffen M, Ruprecht E,

    inventors; Karl Thomae GmbH, assignee. Phenylacetic acid benzy-

    lamides. United States Patent US 6143769. 2000 December 7.17. Mller PG, inventors; Novo, Nordisk A/s, assignee. NIDDM regi-

    men. United States Patent US 6677358. 2004 January 13.18. http://www.onlinepharmacynz.com/product/485/Aspro_Clear_Regu

    lar_Strength_300mg.html.19. Kazmierski WM. Antiviral drugs: from basic discovery through

    clinical trials. New Jersey: Wiley; 2011.20. Singh J, Pudipeddi M, Lindrud MD, inventors; Bristol-Myers Squibb

    Company, assignee. Bisulfate salt of HIV protease inhibitor. UnitedStates Patent US 6087383. 2000 July 11.

    21. Bni S, Szakcs Z, Csernk O, Barcza L, Noszl B. Cyclodextrin/

    imatinib complexation: binding mode and charge dependent stabi-lities. Eur J Pharm Sci2007;30:16774.

    22. http://www.rxlist.com/gleevec-drug.htm.23. Zimmermann J, Sutter B, Burger HM. Crystal modication of a

    N-phenyl-2-pyrimidineamine derivative, processes for its manufac-

    ture and its use. WIPO Patent WO/1999/003854. 1999.24. Downing NS, Ross JS, Jackevicius CA, Krumholz HM. Avoidance

    of generic competition by Abbott laboratories' fenobrate franchise.Arch Intern Med2012;172:72430.

    25. http://www.rxlist.com/trilipix-drug.htm.26. Weatherall MW, Telzerow AJ, Cittadini E, Kaube H, Goadsby PJ.

    Intravenous aspirin (lysine acetylsalicylate) in the inpatient manage-

    ment of headache. Neurology 2010;75:1098103.

    Table 9 Marketed oral products which yield an emulsion or microemulsion in the gastrointestinal tract.

    Drug Product name Company Therapeutic area

    Cyclosporine Sandimmune oral Novartis Immunosuppressant Cyclosporine Neoral Novartis Immunosuppressant Calcitrol Rocaltrol Roche Calcium regulator Clofazimine Lamprene Geigy Leprosy

    Doxercalciferol Hectoral Bone care Calcium regulator Dronabionol Marinol Roxane AnoxeriaDutasteride Avodart GSK Benign Prostatic Hyperplasia (BPH)Isotretionoin Accutane Roche AcneRitonavir Norvir Abbott AIDSRitonavir/lopinavir Kaletra Abbott AIDSParicalcitol Zemplar Abbott Calcium regulator Progesterone Prometrium Solvay Endometrial hyperplasiaSaquinavir Fortovase Roche AIDSSirolimus Rapumune Wyeth-ayerst Immunosuppressant Tritionoin Vesanoid Roche AcneTipranavir Aptivus Boehringer Ingelheim AIDSValproic acid Depakene Abbott Epilepsy

    Insoluble drug delivery strategies 451

    http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref1http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref1http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref1http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref1http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref1http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref1http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref1http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref1http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref1http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref2http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref2http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref2http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref2http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref2http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref2http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref2http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref2http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref2http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref2http://blog.camargopharma.com/index.php/scorecard/2012-approvalshttp://blog.camargopharma.com/index.php/scorecard/2012-approvalshttp://blog.camargopharma.com/index.php/scorecard/2012-approvalshttp://blog.camargopharma.com/index.php/scorecard/2012-approvalshttp://refhub.elsevier.com/S2211-3835(15)00106-9/sbref3http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref3http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref3http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref3http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref3http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref3http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref3http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref3http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref3http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref3http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref5http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref5http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref5http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref5http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref5http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref5http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref5http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref5http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref5http://www.rxlist.com/cipro-iv-drug.htmhttp://www.rxlist.com/cipro-iv-drug.htmhttp://www.rxlist.com/cipro-iv-drug.htmhttp://www.rxlist.com/micardis-drug.htmhttp://www.rxlist.com/micardis-drug.htmhttp://www.rxlist.com/micardis-drug.htmhttp://refhub.elsevier.com/S2211-3835(15)00106-9/sbref8http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref8http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref8http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref8http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref8http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref8http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref8http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref8http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref8http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref8http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref8http://www.onlinepharmacynz.com/product/485/Aspro_Clear_Regular_Strength_300mg.htmlhttp://www.onlinepharmacynz.com/product/485/Aspro_Clear_Regular_Strength_300mg.htmlhttp://www.onlinepharmacynz.com/product/485/Aspro_Clear_Regular_Strength_300mg.htmlhttp://www.onlinepharmacynz.com/product/485/Aspro_Clear_Regular_Strength_300mg.htmlhttp://refhub.elsevier.com/S2211-3835(15)00106-9/sbref10http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref10http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref10http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref10http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref10http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref11http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref11http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref11http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref11http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref11http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref11http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref11http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref11http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref11http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref11http://www.rxlist.com/gleevec-drug.htmhttp://www.rxlist.com/gleevec-drug.htmhttp://www.rxlist.com/gleevec-drug.htmhttp://refhub.elsevier.com/S2211-3835(15)00106-9/sbref13http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref13http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref13http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref13http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref13http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref13http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref13http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref13http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref13http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref13http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref13http://www.rxlist.com/trilipix-drug.htmhttp://www.rxlist.com/trilipix-drug.htmhttp://www.rxlist.com/trilipix-drug.htmhttp://refhub.elsevier.com/S2211-3835(15)00106-9/sbref15http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref15http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref15http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref15http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref15http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref15http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref15http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref15http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref15http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref15http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref15http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref15http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref15http://www.rxlist.com/trilipix-drug.htmhttp://refhub.elsevier.com/S2211-3835(15)00106-9/sbref13http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref13http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref13http://www.rxlist.com/gleevec-drug.htmhttp://refhub.elsevier.com/S2211-3835(15)00106-9/sbref11http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref11http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref11http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref10http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref10http://www.onlinepharmacynz.com/product/485/Aspro_Clear_Regular_Strength_300mg.htmlhttp://www.onlinepharmacynz.com/product/485/Aspro_Clear_Regular_Strength_300mg.htmlhttp://refhub.elsevier.com/S2211-3835(15)00106-9/sbref8http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref8http://www.rxlist.com/micardis-drug.htmhttp://www.rxlist.com/cipro-iv-drug.htmhttp://refhub.elsevier.com/S2211-3835(15)00106-9/sbref5http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref5http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref3http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref3http://blog.camargopharma.com/index.php/scorecard/2012-approvalshttp://blog.camargopharma.com/index.php/scorecard/2012-approvalshttp://refhub.elsevier.com/S2211-3835(15)00106-9/sbref2http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref2http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref1http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref1http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref1
  • 7/25/2019 Insoluble Drug Delivery Strategies

    11/12

    27. Diener H, Asasumamig Study Group. Efcacy and safety ofintravenous acetylsalicylic acid lysinate compared to subcutaneoussumatriptan and parenteral placebo in the acute treatment ofmigraine. A doubleblind, doubledummy, randomized, multicenter,

    parallel group study. Cephalalgia1999;19:5818.28. Seymour RA, Williams FM, Luyk NM, Boyle MA, Whiteld PM,

    Nicholson E, et al. Comparative efcacy of soluble aspirin andaspirin tablets in postoperative dental pain. Eur J Clin Pharmacol

    1986;30:4958.29. European Medicines Agency. Committee for medicinal products for

    human use, plenary meeting monthly report, EMEA/CHMP/389326/2009.

    30. http://www.accessdata.fda.gov/drugsatfda_docs/appletter/2012/201803s000ltr.pdf.

    31. http://www.accessdata.fda.gov/drugsatfda_docs/appletter/2011/202172s000ltr.pdf.

    32. Patel A, Jones SA, Ferro A, Patel N. Pharmaceutical salts: aformulation trick or a clinical conundrum? B r J Cardiol

    2009;16:2816.33. Lee YC, Zocharski PD, Samas B. An intravenous formulation

    decision tree for discovery compound formulation development. IntJ Pharm2003;253:1119.

    34. Kawakami K, Oda N, Miyoshi K, Funaki T, Ida Y. Solubilizationbehavior of a poorly soluble drug under combined use of surfactantsand cosolvents. Eur J Pharm Sci2006;28:714.

    35. http://www.rxlist.com/taxol-drug.htm.36. Hennenfent K, Govindan R. Novel formulations of taxanes: a review.

    Old wine in a new bottle? Ann Oncol2006;17:73549.37. Oerlemans C, Bult W, Bos M, Storm G, Nijsen JFW, Hennink WE.

    Polymeric micelles in anticancer therapy: targeting, imaging andtriggered release. Pharm Res2010;27:256989.

    38. http://www.rxlist.com/taxotere-drug.htm.39. http://www.accessdata.fda.gov/drugsatfda_docs/nda/2011/

    022234Orig1s000Approv.pdf.40. http://www.accessdata.fda.gov/drugsatfda_docs/nda/2011/

    201525Orig1s000ClinPharmR.pdf.41. http://www.accessdata.fda.gov/drugsatfda_docs/appletter/2012/

    022312s000ltr.pdf.42. Powell MF, Nguyen T, Baloian L. Compendium of excipients for

    parenteral formulations. PDA J Pharm Sci Technol 1998;52:238311.

    43. Yu L. Amorphous pharmaceutical solids: preparation, characteriza-tion and stabilization. Adv Drug Deliv Rev2001;48:2742.

    44. Laitinen R, Lbmann K, Strachan CJ, Grohganz H, Rades T.Emerging trends in the stabilization of amorphous drugs. Int JPharm 2013;453:6579.

    45. Crisp HA, Clayton JC, Elliott LG, Wilson EM, inventor; GlaxoGroup Limited, assignee. Preparation of a highly pure, substantiallyamorphous form of cefuroxime axetil. United States Patent US

    4820833. April 111989.46. Crisp HA, Clayton JC, inventors; Glaxo Group Limited, assignee.

    Amorphous form of cefuroxime ester. United States Patent US4562181. 1985 December 31.

    47. Holohan JJ, Edwards IJ, inventors; Imperial Chemical IndustriesPLC, assignee. Pharmaceutical agents. United States Patent US5319097. 1994 July 6.

    48. Corvari SJ, Creekmore JR, inventors; AstraZeneca UK Limited,assignee. Pharmaceutical compositions comprising Zarlukast. Eur-opean Patent EP 1041966. 2003 July 2.

    49. Sinha S, Ali M, Baboota S, Ahuja A, Kumar A, Ali J. Soliddispersion as an approach for bioavailability enhancement of poorlywater-soluble drug ritonavir. AAPS PharmSciTech2010;11:51827.

    50. Leuner C, Dressman J. Improving drug solubility for oral delivery

    using solid dispersions. Eur J Pharm Biopharm2000;50:4760.51. Durate I, Temtem M, Gil M, Gaspar F. Overcoming poor bioavailability

    through amorphous solid dispersions. Ind Pharm2011;30:46.

    52. Miller DA. Improved oral absorption of poorly water-soluble drugsby advanced solid dispersion systems [dissertation]. Austin: Uni-versity of Texas; 2007.

    53. Thakuria R, Delori A, Jones W, Lipert MP, Roy L, Rodrguez-Hornedo N. Pharmaceutical cocrystals and poorly soluble drugs. Int JPharm2013;453:10125.

    54. Almarsson , Peterson ML, Zaworotko M. The A to Z ofpharmaceutical cocrystals: a decade of fast-moving new science

    and patents. Pharm Pat Anal2012;1:313

    27.55. Rios-Doria J, Carie A, Costich T, Burke B, Skaff H, Panicucci R,

    et al. A versatile polymer micelle drug delivery system forencapsulation and in vivo stabilization of hydrophobic anticancerdrugs. J Drug Deliv 2012;2012:951741.

    56. Riggio C, Pagni E, Raffa V, Cuschieri A. Nano-oncology: clinicalapplication for cancer therapy and future perspectives. J Nanomater2011;2011:164506.

    57. Cabral H, Kataoka K. Progress of drug-loaded polymeric micellesinto clinical studies. J Control Release2014;190:46576.

    58. Souto EB. Patenting nanomedicines: legal aspects, intellectualproperty and grant opportunities. Berlin Heidelberg: SpringerScience& Business Media; 2012.

    59. Loftsson T, Brewster ME. Pharmaceutical applications of cyclodex-trins. 1. Drug solubilization and stabilization. J P harm Sci

    1996;85:101725.60. Nekkanti V, Karatgi P, Paruchuri S, Pillai R. Drug product

    development and pharmacological evaluation of a sparingly solublenovel camptothecin analog for peroral administration. Drug Deliv2011;18:294303.

    61. Loftsson T, Duchne D. Cyclodextrins and their pharmaceuticalapplications. Int J Pharm2007;329:111.

    62. Gassmann P, List M, Schweitzer A, Sucker H. Hydrosols: alter-natives for the parenteral application of poorly water soluble drugs.

    Eur J Pharm Biopharm1994;40:6472.63. List M, Sucker H. Pharmaceutical colloidal hydrosols for injection.

    GB Patent 2200048. 1998.64. Sucker H, Gassmann P. Improvements in pharmaceutical composi-

    tions. GB Patent 2269536A. 1994.65. Kipp JE, Wong JCT, Doty MJ, Rebbeck CL. Microprecipitation

    method for preparing submicron suspensions. Google Patents; 2003January 2.

    66. Rogers TL, Johnston KP, Williams RO. Solution-based particleformation of pharmaceutical powders by supercritical or compresseduid CO2and cryogenic spray-freezing technologies. Drug Dev IndPharm2001;27:100315.

    67. Kauffman GB. The origins of heterogeneous catalysis by platinum:Johann Wolfgang Dbereiner's contributions. Enantiomer1999;4:60919.

    68. Robertson AJB. The development of ideas on heterogeneouscatalysis. Platin Met Rev1983;27:319.

    69. Kaparissides C, Alexandridou S, Kotti K, Chaitidou S. Recentadvances in novel drug delivery systems. J Nanotechnol2006;2:111.

    70. Mller RH, Jacobs C, Kayser O. Nanosuspensions as particulate drugformulations in therapy: rationale for development and what we canexpect for the future. Adv Drug Deliv Rev2001;47:319.

    71. Wong J, Brugger A, Khare A, Chaubal M, Papadopoulos P, RabinowB, et al. Suspensions for intravenous (IV) injection: a review ofdevelopment, preclinical and clinical aspects. Adv Drug Deliv Rev2008;60:93954.

    72. Mouton JW, Van Peer A, De Beule K, Van Vliet A, Donnelly J,Soons PA. Pharmacokinetics of itraconazole and hydroxyitracona-zole in healthy subjects after single and multiple doses of a novelformulation. Antimicrob Agents Chemother2006;50:4096102.

    73. Kraft WK, Steiger B, Beussink D, Quiring JN, Fitzgerald N,Greenberg HE, et al. The pharmacokinetics of nebulized nanocrystalbudesonide suspension in healthy volunteers. J Clin Pharmacol2004;44:6772.

    Sandeep Kalepu, Vijaykumar Nekkanti452

    http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref16http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref16http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref16http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref16http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref16http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref16http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref16http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref16http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref16http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref16http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref16http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref16http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref16http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref16http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref16http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref16http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref16http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref16http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref17http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref17http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref17http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref17http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref17http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref17http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref17http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref17http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref17http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref17http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref17http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref17http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref17http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref17http://www.accessdata.fda.gov/drugsatfda_docs/appletter/2012/201803s000ltr.pdfhttp://www.accessdata.fda.gov/drugsatfda_docs/appletter/2012/201803s000ltr.pdfhttp://www.accessdata.fda.gov/drugsatfda_docs/appletter/2012/201803s000ltr.pdfhttp://www.accessdata.fda.gov/drugsatfda_docs/appletter/2012/201803s000ltr.pdfhttp://www.accessdata.fda.gov/drugsatfda_docs/appletter/2011/202172s000ltr.pdfhttp://www.accessdata.fda.gov/drugsatfda_docs/appletter/2011/202172s000ltr.pdfhttp://www.accessdata.fda.gov/drugsatfda_docs/appletter/2011/202172s000ltr.pdfhttp://www.accessdata.fda.gov/drugsatfda_docs/appletter/2011/202172s000ltr.pdfhttp://refhub.elsevier.com/S2211-3835(15)00106-9/sbref20http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref20http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref20http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref20http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref20http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref20http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref20http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref20http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref20http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref21http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref21http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref21http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref21http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref21http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref21http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref21http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref21http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref21http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref21http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref22http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref22http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref22http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref22http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref22http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref22http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref22http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref22http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref22http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref22http://www.rxlist.com/taxol-drug.htmhttp://www.rxlist.com/taxol-drug.htmhttp://www.rxlist.com/taxol-drug.htmhttp://refhub.elsevier.com/S2211-3835(15)00106-9/sbref24http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref24http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref24http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref24http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref24http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref24http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref24http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref24http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref24http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref25http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref25http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref25http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref25http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref25http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref25http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref25http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref25http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref25http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref25http://www.rxlist.com/taxotere-drug.htmhttp://www.rxlist.com/taxotere-drug.htmhttp://www.rxlist.com/taxotere-drug.htmhttp://www.accessdata.fda.gov/drugsatfda_docs/nda/2011/022234Orig1s000Approv.pdfhttp://www.accessdata.fda.gov/drugsatfda_docs/nda/2011/022234Orig1s000Approv.pdfhttp://www.accessdata.fda.gov/drugsatfda_docs/nda/2011/022234Orig1s000Approv.pdfhttp://www.accessdata.fda.gov/drugsatfda_docs/nda/2011/022234Orig1s000Approv.pdfhttp://www.accessdata.fda.gov/drugsatfda_docs/nda/2011/201525Orig1s000ClinPharmR.pdfhttp://www.accessdata.fda.gov/drugsatfda_docs/nda/2011/201525Orig1s000ClinPharmR.pdfhttp://www.accessdata.fda.gov/drugsatfda_docs/nda/2011/201525Orig1s000ClinPharmR.pdfhttp://www.accessdata.fda.gov/drugsatfda_docs/nda/2011/201525Orig1s000ClinPharmR.pdfhttp://www.accessdata.fda.gov/drugsatfda_docs/appletter/2012/022312s000ltr.pdfhttp://www.accessdata.fda.gov/drugsatfda_docs/appletter/2012/022312s000ltr.pdfhttp://www.accessdata.fda.gov/drugsatfda_docs/appletter/2012/022312s000ltr.pdfhttp://www.accessdata.fda.gov/drugsatfda_docs/appletter/2012/022312s000ltr.pdfhttp://refhub.elsevier.com/S2211-3835(15)00106-9/sbref30http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref30http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref30http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref30http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref30http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref30http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref30http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref30http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref30http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref31http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref31http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref31http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref31http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref31http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref31http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref31http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref31http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref31http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref32http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref32http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref32http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref32http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref32http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref32http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref32http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref32http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref32http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref32http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref33http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref33http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref33http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref33http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref33http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref33http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref33http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref33http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref33http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref33http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref34http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref34http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref34http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref34http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref34http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref34http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref34http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref34http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref34http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref35http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref35http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref35http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref35http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref35http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref35http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref35http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref35http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref35http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref37http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref37http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref37http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref37http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref37http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref37http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref37http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref37http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref37http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref37http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref38http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref38http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref38http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref38http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref38http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref38http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref38http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref38http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref38http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref38http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref39http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref39http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref39http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref39http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref39http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref39http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref39http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref39http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref39http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref39http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref39http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref40http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref40http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref40http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref40http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref40http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref40http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref40http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref41http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref41http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref41http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref41http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref41http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref41http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref41http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref41http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref41http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref42http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref42http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref42http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref42http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref42http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref42http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref42http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref43http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref43http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref43http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref43http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref43http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref43http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref43http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref43http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref43http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref44http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref44http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref44http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref44http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref44http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref44http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref44http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref44http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref44http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref44http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref45http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref45http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref45http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref45http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref45http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref45http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref45http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref45http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref45http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref46http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref46http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref46http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref46http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref46http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref46http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref46http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref46http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref46http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref47http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref47http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref47http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref47http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref47http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref47http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref47http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref47http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref47http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref47http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref47http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref47http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref47http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref47http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref48http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref48http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref48http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref48http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref48http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref48http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref48http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref48http://refhub.elsevier.com/S2211-3835(15)00106-9/sbref48http://refhub.elsevier.com/S2211