the role of payload hydrophobicity in nanotherapeutic

Upload: tang-wei-lun

Post on 02-Jun-2018

216 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/11/2019 The Role of Payload Hydrophobicity in Nanotherapeutic

    1/10

    RESEARCH ARTICLE Pharmaceutical Nanotechnology

    The Role of Payload Hydrophobicity in NanotherapeuticPharmacokinetics

    POVILAS NORVAISAS, ARTURAS ZIEMYS

    Houston Methodist Research Institute, Department of Nanomedicine, Houston, Texas 77030

    Received 31 March 2014; revised 26 March 2014; accepted 7 April 2014

    Published online 6 May 2014 in Wiley Online Library (wileyonlinelibrary.com). DOI 10.1002/jps.23996

    ABSTRACT: Although drug delivery with nanovectors is regarded as one of the paradigm-shifting advances in modern medicine, thecompatibility and performance of drugvector formulations have not been systematically studied in terms of their physicochemistry andpharmacokinetics (PKs). The drug delivery systems (DDSs), currently available in clinics or trials, were analyzed based on hydrophobicityand anatomical therapeutic chemical (ATC) classification of drug payloads. Four major types of DDSs differentiated based on DDS structureand drug hydrophobicity, where payload hydrophobicity decreased: micelles, serum albumin, liposome membrane, and liposome interior.A strong relationship between the increase in half-life in DDS formulation and drug hydrophobicity was found with up to 200-fold greaterincrease for hydrophilic drugs. The analysis results seemingly integrated PKs, ATC, and hydrophobicity to reinforce the development oroptimization of drug delivery vectors and their formulations. C2014 Wiley Periodicals, Inc. and the American Pharmacists Association J

    Pharm Sci 103:21472156, 2014Keywords: log P; liposomes; albumin; micelles; nanoparticles; formulation vehicles; formulation; lipoproteins; pharmacokinetics; physic-ochemical

    INTRODUCTION

    Advances in nanotechnology and material sciences havespawned many approaches to enhance drug delivery.13 Themajority of these methods rely on nanoparticles and mi-croparticles, which span a wide range of sizes and shapes.These particles serve as delivery vectors46 for the trans-port of image-contrast agents7,8 and drugs. A large varietyof particles are available as vectors, such as lipid particles, 9

    liposomes,10 micelles,11 serum albumin particles,12 fullerenes,13

    carbon nanotubes,14 dendrimers,15 silica, metallic particles(gold, iron oxide, etc.),16 polymeric particles [e.g., poly(lactic-co-glycolic acid)],17 and hydrogels.18 Drugs that employ vector-based drug delivery systems (DDSs), such as Doxil

    TM, have been

    extensively used in clinical medicine.19 Over the last decade,more DDSs have entered clinical trials, and even more havebeen actively studied,20 especially in the treatment of cancers.

    Some drug delivery strategies involve the modulation anddifferentiation of vector biodistribution in organs or tissues.Therefore, such approaches increase the delivery of the ther-apeutic payload in certain organs.21 DDSs have gained ben-efits over classical drugs in pharmacokinetics (PKs), such asincreasing circulation time or reducing toxicity. However, the

    magnitude of benefits is sometimes poorly defined, as is refer-enced in several papers.22,23 Vectors have their own PK profilesthat may differ from those of therapeutic substances because

    Abbreviations used: ATC, anatomical therapeutic chemical; DDS, drugdelivery system; MPS, mononuclear phagocyte system; HSA, human serumalbumin; BSA, bovine serum albumin; MTD, maximum tolerated dose; AUC,area under the curve.

    Correspondence to: Arturas. Ziemys (Telephone: +713-441-7320; Fax:+713-441-7438; E-mail: [email protected])

    Povilas Norvaisas present address is Department of Biothermodynamics andDrug Design, Vilnius University, Vilnius LT-02241, Lithuania.

    Thisarticlecontains supplementary materialavailable fromthe authors uponrequest or via the Internet at http://onlinelibrary.wiley.com/.

    Journal of Pharmaceutical Sciences, Vol. 103, 21472156 (2014)C 2014 Wiley Periodicals, Inc. and the American Pharmacists Association

    of their size and surface properties. As a result, the vectors cir-culation and interactions with cells may be impacted, thus re-sulting in altered biodistribution. Carriers can also employ theenhanced permeability and retention (EPR) effect.24 The EPReffect is a result of two properties of tumor tissue: leaky vas-culature with fenestrations of up to several microns in size,25

    and greater than normal retention within the tumors intersti-tial fluid. Because of EPR, most polymeric drugs accumulatein tumor tissues at concentrations that are five to 10 times

    or 10 times higher than those in plasma or normal tissues,respectively.24 PEGylated (also referred to as sterically sta-bilized or Stealth

    TM) liposomes display inhibited interactions

    with plasma proteins and mononuclear phagocytes, resultingin prolonged circulation times and increased accumulation inthe interstitial fluid of tumors at levels comparable to thoseof reticuloendothelial system-rich organs.26 The mononuclearphagocyte system (MPS), which consists of phagocytic cells inthe lymph nodes, spleen, or Kupffer cells in the liver, sequestercirculating particles, including carriers.27 This effect is not de-sired frequently, and delivery agents can be chemically alteredto make them invisible to MPS. An example of this alterationis a modification with PEG.28 Because of the EPR effect andMPS, the PKs of vectors are very different from those of smallmolecule drugs. A drug that is associated with a carrier willadopt the carriers PK profile,29 both with advantages and neg-ative consequences.

    The efficacy of DDSs depends on many properties, includinghow much of a drug can be loaded into a carrier. If no covalentbinding is involved to retain drugs inside the carrier matrix, theloading and release of drugs from such DDSs rely on diffusionand concentration gradients, as well as the degradation of thecarrier. Because many different materials of varying composi-tions are possible for the production of vectors, each DDS willhave a specific affinity for a particular drug. Drug hydropho-bicity may influence its loading and release, making it more orless efficient.3038 Moreover, the hydrophobicity of a carrier may

    Norvaisas and Ziemys, JOURNAL OF PHARMACEUTICAL SCIENCES 103:21472156, 2014 2147

  • 8/11/2019 The Role of Payload Hydrophobicity in Nanotherapeutic

    2/10

    2148 RESEARCH ARTICLE Pharmaceutical Nanotechnology

    also have the same effect.3941 However, the carriers influenceis not significant because of the tendency for different immisci-ble systems to have comparable partitioning42 (Fig. A.1).

    LogP is the logarithm of a partitioning coefficient betweenthe waterand octanol phases for a chemical compound. The par-titioning of drugs is a complex process that depends on manyphysicochemical aspects, where logP provides an integratedphenomenological measure. Log D is an alternative parame-ter that accounts for the charge of drug at different pH levels.However, scarce experimental data are available for logD. Inaddition, it is less reliable than log P because of cumulativeerror in log P and pKa measurements.

    43 Log P is one of thefive parameters in the Lipinski rules for drug likeliness, and isemployed in drug discovery.44 Because logPcan be easily eval-uated with good accuracy using computational algorithms,45 itis widely accepted and can be found in many databases.

    In addition to the drugs interactions with the drug mediaand vector matrix, drug loading and release may also be af-fected by the surrounding physiological media, which can beexpressed in terms of the logP value of the drug. Studies in-

    vestigating the systemic basis of how drugs and vectors may

    be coupled are limited.46

    Because logP may affect both drugdelivery and PK, DDSs can be created in response to specifictherapeutic aims to maximize the synergism of the drugs and

    vectors physiochemical properties.We assume that there should be certain level of compati-

    bilities between drug vectors and drugs. Therefore, we haveanalyzed DDSs by investigating the physicochemical, PK, andtherapeutic properties of drugs to establish relations leading tobetter systemic knowledge about existing and future DDS.

    METHODS

    Drug Information

    DrugBank is a freely available database, which contained 6711total entries at the time of the study.47,48 The entire dataset, in-cluding small molecules and biomolecules, was parsed in XMLformat and analyzed with Knime software.49,50 Drug proper-ties, including log P, bioavailability, and half-life (t1/2), wereextracted with the accompanying structures. The parsing andanalysis of the dataset were automated to prepare informationfor further statistical evaluation. Most of the logP values forsalt-free drugs were taken from an extensive study by Han-sch and Leo.51 In this study, we will refer to experimentallyestablished logP values in the octanolwater system, unlessotherwise stated. Missing logP values in Table 1 were supple-mented by calculations with XlogP implementation in Knime

    software,49

    and they are marked with asterisks. The perfor-mance of XlogP was validated with DrugBank drugs, and theresults are displayed in Figure A.2. Drug bioavailability andt1/2 were extracted with regular expression text analysis tools,and then verified by visual inspection.

    After DrugBank was updated on August 2, 2013, the exper-imental logP values of several drugs (doxorubicin, daunoru-bicin, floxuridine, and fluorouracil) were altered, although theofficial database snapshot was not updated at the time ofmanuscript submission. The changes for both floxuridine andfluorouracil were very minor (0.2 and 0.1 logPunits). However,the logP values for doxorubicin and daunorubicin drasticallyincreased from0.5 and 0.1 to 1.27 and 1.83, respectively. Thischange seemed ambiguous because of the known solubility of

    these compounds. Therefore, we averaged the logPvalues thatwere documented in various studies. The calculated averagelogPvalue for doxorubicin and daunorubicin were0.26,44,5258

    and 0.72,5759 respectively. All of these values were determinedat pH 77.2 for salt-free drugs.

    Data regarding the anatomical therapeutic chemical (ATC)classification was collected, along with defined daily doses andadmission routes.60,61 Most of the drugs in DrugBank have

    ATC codes assigned to them, and some have several codes be-cause of the use of the same chemical compound for differentindications. The ATC code consists of several levels that de-scribe the anatomical, therapeutic, and pharmacological clas-sification of a specific chemical moiety (http://www.whocc.no/atc/structure and principles/). An extensive ATC code descrip-tion is given in Tables A.1A.2. In this study, drugs in Drug-Bank were grouped according to the first, ATC (1), and second,

    ATC (2), levels of ATC classification: anatomical location andtherapeutic action.

    Statistical Analysis

    The extracted data were analyzed using the StatSoft Statis-

    tica 10 software. We chose to directly analyze logP values in-stead of the partitioning coefficient (P), because logP exhibitsnormal distribution, which was also observed for all differentsubgroups of drugs. P distribution is highly asymmetric andcannot be easily parameterized. Datasets that were categorizedinto ATC (1) and ATC (2) classifications and exhibited normaldistribution was evaluated using parametric one-way analysisof variance. After the heterogeneous categories were identi-fied and homoscedasticity was proven by the Levene test, pair-wise comparisons of categories were performed with Tukeyshonest significant difference test for samples with unequal N(Spjotvoll/Stoline). This approach allowed us to identify sig-nificantly different drug groups at both ATC (1) and ATC (2)

    levels (Tables A.3. and A.4.). For box-and-whisker plots withboth normal and nonparametric distributions, only categorieswith more than four members were used.

    Pharmacokinetics

    Detailed PK analyses were performed for DDSs with sufficientdata. We used the mean values of PK parameters at the samedosage levels, and preferentially selected the maximum toler-ated dose (MTD) or the most commonly used dose of the freedrug if the MTD was unknown. In the case of paclitaxel, therewas no carrier-free formulation of the drug; therefore, we usedTaxol

    TMas a reference, regardless of its use of CremaphorEL.

    RESULTS

    This study depended on well-defined reference data. Whatmakes a good DDS depends on the purpose of that particularsystem. To provide the least-biased analysis, we assumed thatDDSs that are currently employed in clinics or clinical trialsare sufficiently well developed. Their usage in clinics suggeststhat the given DDS meets some clinical and engineering expec-tations. Therefore, the study began with a brief analysis of suchDDSs, where the physicochemical boundaries were evaluatedand compared against available drugs and therapeutic classi-fiers. The second part of the study dissected the PK propertiesof the DDSs, and established correlates to couple the propertiesof drugs and DDSs with their PK. Less relevant data related to

    Norvaisas and Ziemys, JOURNAL OF PHARMACEUTICAL SCIENCES 103:21472156, 2014 DOI 10.1002/jps.23996

  • 8/11/2019 The Role of Payload Hydrophobicity in Nanotherapeutic

    3/10

    RESEARCH ARTICLE Pharmaceutical Nanotechnology 2149

    Table 1. Classification of nanotherapeutics in clinical trials and clinics based on logP values and carrier types. The active ssubstance, brand

    name, average diameter of the carrier (), logP, indications, and information sources are given for each drug. Average log P values were

    calculated for each carrier type, and only unique drug substances were considered

    Type Drug Brand Name (nm) Log PD Average LogPD Indications

    Liposome interior Amikacin MiKasomeTM

    50 7.4 1.1 Infections

    Cytarabine CPX-351 100 2.8 Cancer

    Cisplatin SPI-077 110 2.2 Cancer

    Floxuridine CPX-1 110 1.2 CancerFluorouracil 5-FU 130170 0.9 Cancer

    Doxorubicin MyocetTM

    150180 0.3 Cancer

    Doxorubicin Doxil/Caelyx 100 0.3 Cancer

    Doxorubicin SarcodoxomeTM

    0.3 Cancer

    Doxorubicin MCC465 143 0.3 Cancer

    Lurtotecan* OSI/NX-211

  • 8/11/2019 The Role of Payload Hydrophobicity in Nanotherapeutic

    4/10

    2150 RESEARCH ARTICLE Pharmaceutical Nanotechnology

    Percentag

    e

    P

    ercentage

    P

    Figure 1. The distribution of experimental logPvalues for DrugBank

    drugs (columns). The Gaussian fit (red line) over all drugs is centered

    at 1.9. There is a significant overlap between the logP coverage of the

    DDSs (blue line) and the population of the drugs.

    Hydrophilic and hydrophobic vectors may comprise 29% and57% of all investigated drugs, respectively. As shown in Table1 and Figure 1, the vectors encompass the logPrange between7.4 and 4.3. The remaining 14% seem to be incompatible withcurrently available vectors. The major carriers are liposomes,which encompass 70% of all drugs, with 28% sequestered inthe liposome aqueous core and 42% in the liposome membrane.

    Albumin appears to be the second major carrier type with logPvalues ranging from 1.8 to 4.3, thus representing 42% of allDrugBank drugs. Finally, micelles appear to have a slightlynarrower range of compatible logPvalues between 2.4 and 4.2.However, this group coincides with the peak of the histogramin Figure 1 and encompasses 29% of potential drugs. The log

    P range between 2.4 and 4.2 not only contains most of the

    drugs, but is shared among micelles, liposome membranes, andalbumin particles. Therefore, these drugs possess the highestpotential of success with vectors, as one can select from a large

    variety of PK and biodistribution profiles that are associatedwith different DDS types.

    The majority of drugs demonstrate low water solubility intheir salt-free forms, because of comparably high log P val-ues. This coincides with the active components of carriers,such as the internal milieu of liposome membranes and mi-celles. The partitioning of drugs and hydrocarbons between wa-ter and different lipophilic phases are similar for compoundswith low logP values (up to logP = 3), whereas larger par-titioning differences appear in compounds with larger log P

    values.62,63

    The partitioning ofn-alkanes in water-octanol andwater-hexadecane is only slightly higher in the lipophilic phaseof hexadecane42 (Fig. A.1). This suggests that the carrier ma-trix should be of a similar or larger log P value to provide aphase that would offer efficient hydrophobic drug loading andcompatibility. Results from our analysis reveal that drugs withspecific logP values are largely being used with a particularcarrier type.

    Differences in log P among drugs with various therapeu-tic aims can lead to alterations in the loading and release ofthose drugs from the carriers. The release rate seems to beclosely related to the partitioning of the drug. For example,more hydrophobic drugs are released at slower rates. The re-lease of Ftorafur is slower than that of 5-FU, which is less

    lipophilic.30 Lipophilic compounds, including rapamycin and cy-closporine A, partition predominantly into micelle cores and arereleased very slowly.39 For antiepilepticdrugs, rufinamide is themost hydrophobic with the slowest release.36 The release of dif-ferent corticosteroids decreases with increasing lipophilicity.37

    The properties of the matrix of the carrier itself also modulatethe release rate depending on its hydrophobicity. However, ac-cording to the data for linear alkanes whose partitioning wasinvestigated in different immiscible systems, such modulationshould be minor42 (Fig. A.1). Therefore, the drug release pro-cess depends heavily on the partitioning of the drug (Fig. A.4.),which can be more important than the reduction of drug diffu-sion by the carrier matrix.64 Further discussion on this topic isprovided in the Appendix section Diffusion Mass Release.

    Anatomical Therapeutic Chemical

    The analysis was extended to determine whether the thera-peutic and anatomical groups of drugs have divergent log P

    values and exhibit hydrophobicity associated correlations be-tween ATC and carriers. Experimental logP values of Drug-

    Bank entries were grouped according to their ATC (1) (Fig. A.4;Table A.8) and ATC (2) codes, which represent the anatomicallocations of drug action and therapeutic grouping correspond-ingly.

    By following the ATC (2) classification, 88 therapeutic druggroups were obtained. Seventy one out of the 88 groups hadat least five members and were used for further analysis.Fifty-eight groups were significantly divergent with the meansspread in seven logP units (Tables A.4 and A.9). The distribu-tion of all 71 groups is shown in Figure 2. The variance of log

    P within a single set was very different at times. In some ofthe groups, high variances in logP (e.g., C05 and B01) coveredalmost the entire range of values found in DrugBank, whereasother groups had very tight distribution (L03 withN= 12, D07

    with N= 39, N01 with N= 33, and A14 with N= 5). Thecorrelation of logP values with ATC suggests that particulardrug groups may be used with a specific carrier type (Table

    A.9). Therefore, the degree of hydrophobicity may be used todiscriminate drugs by anatomical locations of their actions andtherapeutic grouping, as well as to identify the most optimalcarrier (Table 1).

    Pharmacokinetics

    We further extended our study of DDSs by analyzing their PKsandcomparing them to those of free drugs. The potentialof drug

    vectors to prolong the half-lives of drugs is shown in Figure 3.

    Our analysis revealed that many drugs have relatively shorthalf-lives: 68% of DrugBank drugs have a t1/2 shorter than12 h, 50% shorter than 6 h, and 10% less than 1 h (Table A.10.).Figure 3 shows a comparison oft1/2changes for compounds thatare currently used in trials and clinics, where the change int1/2is depicted by arrows and sorted by drug hydrophobicity. Mostof these drugs have short half-lives (

  • 8/11/2019 The Role of Payload Hydrophobicity in Nanotherapeutic

    5/10

    RESEARCH ARTICLE Pharmaceutical Nanotechnology 2151

    Figure 2. The logPdistribution in different therapeutic groups of DrugBank drugs based on ATC(2) levels. The group all represents all drugs

    in the DrugBank. Compatibility ranges for different delivery vehicles are color-coded in the background with the number of drugs for each group

    shown by columns at the top.

    ( )

    Figure 3. The histogram shows the distribution oft1/2 of DrugBank

    drugs with the percentage of the whole population displayed on top of

    the bars. The arrows illustrate the change in the t1/2 of a particular

    drug substance following DDS delivery, starting at the t 1/2 of a pure

    drug and ending at the t 1/2 of vector formulation. The text on top of

    the arrows indicates the brand names (active substance, log P) and

    multiplicator oft1/2 change.

    DDSs depends on many important aspects, each carrier groupis briefly reviewed below, including representative cases.

    Liposome-Based Formulations

    AmBisome carries the anti-infective drug, amphotericin B, ina liposome membrane. The mean t1/2 of total amphotericin Bafter liposomal administration is 152 h, compared with 362h of the free drug in plasma.65 However, with a threefold in-crease in the dose, the Cmaxis eightfold higher and the medianarea under the curve (AUC) is ninefold higher under conditionswhere the total available drug pool is analyzed.66 Doxorubicin,

    a popular anticancer drug, is already used with carriers in twoformulations (Doxil/Caelyx, Myocet) with one undergoing clin-ical trials (MMC-465).t1/2 was prolonged in all three formula-tions; however, the PEGylation of Doxil causes toxicity in theform of hand-foot syndrome, as well as other dose-limiting sideeffects.67 All three DDSs of doxorubicin exhibit different ad-

    vantages over the free formulation of the drug. CPX-351 is abilameral liposomal formulation, in which cytarabine and dox-orubicin are maintained in the aqueous liposome interior at aratio of 5:1. This carrier prolongs the terminal t1/2 of cytara-bine from 12 min to an average of 34 h.68,69 The terminal t1/2of daunorubicin increases from 17 to 25 h on average.68,70,71

    Equivalent therapeutic efficacy is achieved with doses lowerthan conventional formulations.72 CPX-351 employs a synergyof the drugs.68

    Albumin-Based Formulations

    Albumin-bound paclitaxel (nab-paclitaxel; Abraxane) uses analbumin nanoparticle as its carrier.73,74 Human serum albumin(HSA) nanoparticles possess high drug loading efficiency with-out the risk of toxicity. However, circulation of the drug cannotbe substantially prolonged, as the mean t1/2for Abraxane is sim-ilar to that of the solution-based paclitaxel (Taxol). Even though

    a higher initial concentration of the drug can be reached, sys-tematic exposure remains the same as with Taxol, because ofits increased rate of clearance.75,76 The main advantages of the

    Abraxane formulation include reductions in toxicity, a 3.8-foldincrease in peak plasma concentration, and a 10-fold increasein the maximum concentration of the unbound drug.75,77

    Micelle-Based Formulations

    Genexol-PM is a PEGpoly-(D,L-lactide) polymeric micellar for-mulation of paclitaxel. It was developed to overcome toxicitiescaused by CremaphorEL (Taxol). Genexol-PM has a lower peakplasma concentration and lower systematic exposure (AUC)with a slight increase in t1/2 on average. Similar to Abraxane,

    DOI 10.1002/jps.23996 Norvaisas and Ziemys, JOURNAL OF PHARMACEUTICAL SCIENCES 103:21472156, 2014

  • 8/11/2019 The Role of Payload Hydrophobicity in Nanotherapeutic

    6/10

    2152 RESEARCH ARTICLE Pharmaceutical Nanotechnology

    Figure 4. Augmentation of drugt1/2 in vectors, according to drug hy-

    drophobicity. Amikacin in MiKasome was excluded from the regression

    as an outlier (marked by an asterisk).

    it has a good toxicity profile with no known side effects. Un-like Taxol, Genexol-PM does not require premedication withsteroids or antihistamines.76 A threefold increase in MTD isobserved, and the drug levels are two to three times higher in

    various tissues, including tumors.78

    Competitive Drug Binding in Blood Plasma

    While summarizing the changes in t1/2 in all analyzed DDSs,we noticed that the hydrophilic and hydrophobic drugs havecompletely different PKs when used with vectors. As shown inFigure 4, the t1/2 of hydrophilic drugs is prolonged to a greaterextent than that of hydrophobic drugs, regardless of a carriertype. The t1/2 of hydrophobic drugs is barely prolonged by the

    vector. This may be explained by the observation that hydropho-

    bic drugs can quickly associate with blood serum proteins.7981

    Findings from earlier studies suggest that HSA, alpha-1-acidglycoprotein, and low- and high-density lipoproteins (LDL andHDL) may be responsible for the lack of change in the t1/2 ofhydrophobic drugs.82

    Human serum albumin is the most common protein in theblood with a circulation time of up to 19 days.73 It binds hy-drophobic drugs in several pockets on its surface,73,83,84 andmay also trigger the dissociation of certain micelle systems.85

    Using results on drug binding to HSA,86 our analysis confirmedthat logPvalues and drug binding have a strong positive corre-lation (Fig. A.5). The presence of bovine serum albumin (BSA)(almost identical to HSA) in solution (4% [w/w]) doubles the

    release of rapamycin from PEGPCL micelles.39

    The associa-tion of BSA with the free drug allows more of the hydrophobicdrug to be released, compared with that observed by parti-tioning alone.39 Drugs with a high affinity to serum albumin(>95% bound) require correspondingly higher doses to achieveeffective concentrations in vivo.79 The amount of free hydropho-bic drug can be significantly reduced67; in the case of ampho-tericin B, more than 90% of the drug that is injected with lipo-somes is recovered as HSA bound.65 LDL and HDL may alsoaccount for the different PKs of hydrophobic drugs. The asso-ciation or interaction with cholesterol, triglycerides, phospho-lipids, and LDL/HDL seem to be general property of vectors. 87

    For liposomes incubated with human plasma, up to 67% ofhydrophobic drugs are recovered in the HDL fraction.82 The

    t

    (

    (

    (

    (

    (

    (

    Figure 5. The changes in PK parameters of drugs after they are used

    in DDSs. The change in AUC was kept in a linear scale for clarity. The

    red lines separate DDSs into groups, based on changes in their t1/2and

    Cmax.

    above-mentioned blood components eventually serve as naturaldelivery systems (NDSs) that sequester drugs from synthetic

    vectors, and can be employed for drug delivery.82,88

    Hydrophobicity and PKs

    We analyzed hydrophobicity of the DDSs and changes in theoverall PK profiles of drugs in formulations with and withoutcarrier. For 10 formulations with sufficient data, we comparedthe average values of AUC, Cmax, and t1/2(Table A.11). All DDSsshowed an increase in at least one of the parameters of PK,as seen in Figure 5. On the basis of the changes in t1/2 andCmax, three DDS groups were identified. The first group had anincreased t1/2 and decreased Cmax, like Genexol-PM CPX-351(daunorubicin). The second group had a decreased t1/2 and in-creasedCmax, like AmBisome and DaunoXome. The third grouphad an increased t1/2 and Cmax, like Doxil, Myocet, MCC465,CPX-351 (cytarabine), and SPI-077. All DDSs with drugs for-mulated in the liposome interior showed a significant increase

    in the AUC, except for Abraxane, which had its AUC reducedcompared with Taxol.The hydrophilic drugs had a greater increase in the AUC

    compared with the hydrophobic drugs. Even though the AUCis considered to be a measure of exposure to the drug, it can-not be generalized whether the increase in this parameter isfavorable. All of the 10 DDS formulations have benefited fromreductions in systemic toxicities, regardless of changes in PK.Both Genexol-PM andAbraxane were created to avoid toxicitiesassociated with CremaphorEL in Taxol. Small changes in PKparameters are likely to be caused not only by hydrophobicity,but also by the fact that we are comparing parameters againstthose of Taxol, which already has solubilizing agent. AmBi-some has benefited because amphotericin B is more effectively

    Norvaisas and Ziemys, JOURNAL OF PHARMACEUTICAL SCIENCES 103:21472156, 2014 DOI 10.1002/jps.23996

  • 8/11/2019 The Role of Payload Hydrophobicity in Nanotherapeutic

    7/10

    RESEARCH ARTICLE Pharmaceutical Nanotechnology 2153

    Table 2. The models of relation between PK and logP. The asterix

    denotes a base 10 logarithm of a ratio in a parameter

    LogP and PK Relations R2 p

    AUC 0.52Cmax + 0.38t1/2 0.31 logP + 0.41 0.87

  • 8/11/2019 The Role of Payload Hydrophobicity in Nanotherapeutic

    8/10

    2154 RESEARCH ARTICLE Pharmaceutical Nanotechnology

    6. Blanco E, Sangai T,Hsiao A, Ferrati S, Bai L, Liu X, Meric-Bernstam

    F, Ferrari M. 2012. Multistage delivery of chemotherapeutic nanopar-

    ticles for breast cancer treatment. Cancer Lett 334:245251.

    7.Karathanasis E, Suryanarayanan S, Balusu SR, McNeeley K,

    Sechopoulos I, Karellas A, Annapragada AV, Bellamkonda RV.

    2009. Imaging nanoprobe for prediction of outcome of nanoparticle

    chemotherapy by using mammography. Radiology 250:398406.

    8.Na HB, Song IC, Hyeon T. 2009. Inorganic nanoparticles for MRI

    contrast agents. Adv Mater 21:21332148.9.Mehnert W, Mader K. 2012. Solid lipid nanoparticles produc-

    tion, characterization and applications. Adv Drug Del Rev 47:165

    196.

    10.Allen TM, Cullis PR. 2012. Liposomal drug delivery systems: From

    concept to clinical applications. Adv Drug Del Rev 65:3648.

    11.Nishiyama N, Cabral H, Kataoka K. 2012. Micellar structures as

    drug delivery systems. Drug Deliv Oncol 10511069.

    12.Miele E, Spinelli GP, Tomao F, Tomao S. 2009. Albumin-bound for-

    mulation of paclitaxel (Abraxane RABI-007) in the treatment of breast

    cancer. Int J Nanomed 4:99.

    13.Montellano A, Da Ros T, Bianco A, Prato M. 2011. Fullerene C60

    as a multifunctional system for drug and gene delivery. Nanoscale

    3:40354041.

    14.Liu Z, Tabakman S, Welsher K, Dai H. 2009. Carbon nanotubes in

    biology and medicine: In vitro and in vivo detection, imaging and drugdelivery. Nano Res 2:85120.

    15.Nanjwade BK, Bechra HM, Derkar GK, Manvi F, Nanjwade VK.

    2009. Dendrimers: Emerging polymers for drug-delivery systems. Eur

    J Pharm Sci 38:185196.

    16.Duncan B, Kim C, Rotello VM. 2010. Gold nanoparticle platforms

    as drug and biomacromolecule delivery systems. J Control Release

    148:122127.

    17. Acharya S, Sahoo SK.2011.PLGA nanoparticles containing various

    anticancer agents and tumour delivery by EPR effect. Adv Drug Deliv

    Rev 63:170183.

    18. Qiu Y, Park K. 2012. Environment-sensitive hydrogels for drug

    delivery. Adv Drug Del Rev 64:4960.

    19.Barenholz YC. 2012. Doxil RThe first FDA-approved nano-drug:

    Lessons learned. J Control Release 160:117134.

    20.Wang AZ, Langer R, Farokhzad OC. 2012. Nanoparticle delivery ofcancer drugs. Annu Rev Med 63:185198.

    21. Brannon-Peppas L, Blanchette JO. 2012. Nanoparticle and tar-

    geted systems for cancer therapy. Adv Drug Deliv Rev 64:206

    212.

    22.Jain RK, Stylianopoulos T. 2010. Delivering nanomedicine to solid

    tumors. Nature Rev Clin Oncol 7:653664.

    23. Davis ME. 2008. Nanoparticle therapeutics: An emerging treat-

    ment modality for cancer. Nat Rev Drug Discov 7:771782.

    24.Maeda H. 2001. The enhanced permeability and retention (EPR)

    effect in tumor vasculature: The key role of tumor-selective macro-

    molecular drug targeting. Adv Enzyme Regul 41:189207.

    25. Hashizume H, Baluk P, Morikawa S, McLean JW, Thurston G,

    Roberge S, Jain RK, McDonald DM. 2000. Openings between defec-

    tive endothelial cells explain tumor vessel leakiness. Am J Pathol 156:

    1363.26.Park JW. 2002. Liposome-based drug delivery in breast cancer

    treatment. Breast Cancer Res 4:9599.

    27.Geissmann F, Gordon S, Hume DA, Mowat AM, Randolph GJ.2010.

    Unravelling mononuclear phagocyte heterogeneity. Nat Rev Immunol

    10:453460.

    28.Li SD, Huang L. 2010. Stealth nanoparticles: High density but

    sheddable PEGis a keyfor tumor targeting. J Control Release 145:178

    181.

    29. Simovic S, PrestidgeCA. 2007. Nanoparticle layers controlling drug

    release from emulsions. Eur J Pharm Biopharm 67:3947.

    30.Arias JL, Linares-Molinero F, Gallardo V, Delgado AV. 2008. Study

    of carbonyl iron/poly (butylcyanoacrylate)(core/shell) particles as anti-

    cancer drug delivery systems: Loading and release properties. Eur J

    Pharm Sci 33:252261.

    31.Bromberg L, Temchenko M, Hatton TA. 2002. Dually responsive

    microgels from polyether-modified poly (acrylic acid): Swelling and drug

    loading. Langmuir 18:49444952.

    32.Gomez-Gaete C, Fattal E, Silva L, Besnard M, Tsapis N. 2008.

    Dexamethasone acetate encapsulation into Trojan particles. J Control

    Release 128:4149.

    33.Redhead H, Davis S, Illum L. 2001. Drug delivery in poly (lactide-

    co-glycolide) nanoparticles surface modified with poloxamer 407 and

    poloxamine 908: In vitro characterisation and in vivo evaluation. J Con-trol Release 70:353363.

    34.Arnold MM, Gorman EM, Schieber LJ, Munson EJ, Berkland C.

    2007. NanoCipro encapsulation in monodisperse large porous PLGA

    microparticles. J Control Release 121:100109.

    35.Lammel A, Schwab M, Hofer M, Winter G, Scheibel T. 2011. Re-

    combinant spider silk particles as drug delivery vehicles. Biomaterials

    32:22332240.

    36.Thomas M, Slipper I, Walunj A, Jain A, Favretto M, Kallinteri P,

    Douroumis D. 2010. Inclusion of poorly soluble drugs in highly ordered

    mesoporous silica nanoparticles. Int J Pharm 387:272277.

    37.Jensen LB, Magnussson E, Gunnarsson L, Vermehren C, Nielsen

    HM, Petersson K. 2010. Corticosteroid solubility and lipid polarity con-

    trol release from solid lipid nanoparticles. Int J Pharm 390:5360.

    38.Bailey MM, Berkland CJ. 2009. Nanoparticle formulations in pul-

    monary drug delivery. Med Res Rev 29:196212.39. Forrest ML, Won CY, Malick AW, Kwon GS. 2006. In vitro re-

    lease of the mTOR inhibitor rapamycin from poly(ethylene glycol)-b-

    poly(epsilon-caprolactone) micelles. J Control Release 110:370377.

    40.Chen CC, Tsai TH, Huang ZR, Fang JY. 2010. Effects of lipophilic

    emulsifiers on the oral administration of lovastatin from nanostruc-

    tured lipid carriers: Physicochemical characterization and pharmacoki-

    netics. Eur J Pharm Biopharm 74:474482.

    41.Dalpiaz A, Mezzena M, Scatturin A, Scalia S. 2008. Solid lipid

    microparticles for the stability enhancement of the polar drug N6-

    cyclopentyladenosine. Int J Pharm 355:8186.

    42.Khadikar PV, Mandloi D, Bajaj AV, Joshi S. 2003. QSAR study on

    solubilityof alkanes in water andtheir partition coefficients in different

    solvent systems using PI index. Bioorg Med Chem Lett 13:419422.

    43.Tetko IV, Bruneau P. 2004. Application of ALOGPS to predict 1-

    octanol/water distribution coefficients, logP, and logD, of AstraZenecain-house database. J Pharm Sci 93:31033110.

    44.Lipinski CA, Lombardo F, Dominy BW, Feeney PJ. 2012. Experi-

    mental and computational approaches to estimate solubility and per-

    meability in drug discovery and development settings. Adv Drug Del

    Rev 23:325.

    45. Mannhold R, Poda GI, Ostermann C, Tetko IV. 2009. Calcula-

    tion of molecular lipophilicity: State-of-the-art and comparison of log

    P methods on more than 96,000 compounds. J Pharm Sci 98:861

    893.

    46. Zucker D, Marcus D, Barenholz Y, Goldblum A. 2009. Liposome

    drugs loading efficiency: A working model based on loading conditions

    and drugs physicochemical properties. J Control Release 139:7380.

    47. Wishart DS, Knox C, Guo AC, Cheng D, Shrivastava S, Tzur D,

    Gautam B, Hassanali M. 2008. DrugBank: A knowledgebase for drugs,

    drug actions and drug targets. Nucleic Acids Res 36:D901D906.48.Wishart DS, Knox C, Guo AC, Shrivastava S, Hassanali M,

    Stothard P, Chang Z, Woolsey J. 2006. DrugBank: A comprehensive

    resource for in silico drug discovery and exploration. Nucleic Acids Res

    34:D668D672.

    49.Berthold MR, Cebron N, Dill F, Gabriel TR, Kotter T, Meinl T,

    Ohl P, Sieb C, Thiel K, Wiswedel B. 2008. KNIME: The Konstanz

    informationminer. In Data analysis,machine learning and applications

    studies in classification, data analysis, and knowledge organization;

    Preisach C, BurkhardtH, Schmidt-Thieme L, Decker R, Eds. New York:

    Springer, pp 319326.

    50.Berthold MR, Cebron N, Dill F, Gabriel TR, Kotter T, Meinl T,

    Ohl P, Thiel K, Wiswedel B. 2009. KNIMEThe Konstanz information

    miner. Version 2.0 and beyond. ACM SIGKDD Explor Newslett 11:26

    31.

    Norvaisas and Ziemys, JOURNAL OF PHARMACEUTICAL SCIENCES 103:21472156, 2014 DOI 10.1002/jps.23996

  • 8/11/2019 The Role of Payload Hydrophobicity in Nanotherapeutic

    9/10

    RESEARCH ARTICLE Pharmaceutical Nanotechnology 2155

    51.Hansch C, Leo A. 1995. Exploring QSAR: Fundamentals and ap-

    plications in chemistry and biology. Washington: American Chemical

    Society.

    52.Leo A, Hansch C, Elkins D. 1971. Partition coefficients and their

    uses. Chem Rev 71:525616.

    53.Melik-Nubarov N, Kozlov MY. 1998. Evaluation of partition coeffi-

    cients of low molecular weight solutes between water and micelles of

    block copolymer of ethylene oxide based on dialysis kinetics and fluo-

    rescence spectroscopy. Colloid Polym Sci 276:381387.54.Gallois L, Fiallo M, Laigle A, Priebe W, Garnier-Suillerot A. 1996.

    The overall partitioning of anthracyclines into phosphatidyl-containing

    model membranes depends neither on the drug charge nor the presence

    of anionic phospholipids. Eur J Biochem 241:879887.

    55.Knox C, Law V, Jewison T, Liu P, Ly S, Frolkis A, Pon A, Banco K,

    Mak C, Neveu V. 2011. DrugBank 3.0: A comprehensive resource for

    omics research on drugs. Nucleic Acids Res 39:D1035D1041.

    56.Levin VA. 1980. Relationship of octanol/water partition coefficient

    and molecular weight to rat brain capillary permeability. J Med Chem

    23:682684.

    57.Hoffmann D, Berscheid H, Boettger D, Hermentin P, Sedlacek H,

    Kraemer H. 1990. Structureactivity relationship of anthracyclines in

    vitro. J Med Chem 33:166171.

    58. Zamora JM, Pearce H, Beck WT. 1988. Physicalchemical prop-

    erties shared by compounds that modulate multidrug resistance inhuman leukemic cells. Mol Pharm 33:454462.

    59.Knox C, Law V, Jewison T, Liu P, Ly S, Frolkis A, Pon A, Banco K,

    Mak C, Neveu V. 2011. DrugBank 3.0: A comprehensive resource for

    omics research on drugs. Nucleic Acids Res 39:D1035D1041.

    60.NatschS, Hekster Ya, de Jong R, Heerdink ER,Herings RM,van der

    Meer JW. 1998. Application of the ATC/DDD methodology to monitor

    antibiotic drug use. Eur J Clin Microbiol Infect Dis 17:2024.

    61.Bergman U. 2006. Thehistory of thedrug utilization research group

    in Europe. Pharmacoepidemiol Drug Saf 15:9598.

    62.AvdeefA, Box K, Comer J, Hibbert C, Tam K. 1998. pH-Metric logP

    10. Determination of liposomal membranewater partition coefficients

    of lonizable drugs. Pharm Res 15:209215.

    63.Gobas FAPC, Mackay D, Shiu WY, Lahittete JM, Garofalo G. 1988.

    A novel method for measuring membrane-water partition coefficients

    of hydrophobic organic chemicals: Comparison with 1-octanolwaterpartitioning. J Pharm Sci 77:265272.

    64.Bunjes H. 2010. Lipid nanoparticles for the delivery of poorly water-

    soluble drugs. J Pharm Pharmacol 62:16371645.

    65. Bekersky I, Fielding R. 2002. Plasma protein binding of ampho-

    tericin B and pharmacokinetics of bound versus unbound amphotericin

    B after administration of intravenous liposomal amphotericin. Antimi-

    crob Agents Chemother 46:834840.

    66.Heinemann V, Bosse D, Jehn U. 1997. Pharmacokinetics of liposo-

    mal amphotericin B (Ambisome) in critically ill patients. Antimicrob

    Agents Chemother 41:12751280.

    67. Li S, Huang L. 2008. Pharmacokinetics and biodistribution of

    nanoparticles. Mol Pharm 5:496504.

    68.Feldman EJ, Lancet JE, Kolitz JE, Ritchie EK, Roboz GJ, List AF,

    Allen SL, Asatiani E, Mayer LD, Swenson C, Louie AC. 2011. First-

    in-man study of CPX-351: A liposomal carrier containing cytarabineand daunorubicin in a fixed 5:1 molar ratio for the treatment of re-

    lapsed and refractory acute myeloid leukemia. J Clin Oncol 29:979

    985.

    69.Tardi P, Johnstone S, Harasym N, Xie S, Harasym T, Zisman N,

    Harvie P, Bermudes D, Mayer L. 2009. In vivo maintenance of syn-

    ergistic cytarabine:daunorubicin ratios greatly enhances therapeutic

    efficacy. Leukemia Res 33:129139.

    70.Gill P, Espina B, Muggia F. 1995. Phase I/II clinical and pharma-

    cokinetic evaluation of liposomal daunorubicin. J Clin Oncol 13:996

    1003.

    71.Bellott R, Auvrignon A, Leblanc T, Perel Y, Gandemer V, Bertrand

    Y, Mechinaud F, BellengerP, Vernois J, Leverger G, Baruchel A, Robert

    J. 2001. Pharmacokinetics of liposomal daunorubicin (DaunoXome)

    duringa phase III study in children with relapsed acute lymphoblastic

    leukaemia. Cancer Chemother Pharmacol 47:1521.

    72.Bayne W. 2008. Pharmacokinetics of CPX-351 (cytara-

    bine/daunorubicin HCl) liposome injection in the mouse. J Pharm Sci

    98:25402548.

    73.Elsadek B, Kratz F. 2012. Impactof albumin on drug deliveryNew

    applications on the horizon. J Control Release 157:428.

    74.Kratz F. 2008. Albumin as a drug carrier: Design of prodrugs, drug

    conjugates and nanoparticles. J Control Release 132:171183.75.Gardner ER, Dahut W, Figg WD. 2008. Quantitative determination

    of total and unbound paclitaxel in human plasma following Abraxane

    treatment. J Chromatogr B 862:213218.

    76.Sparreboom A, Scripture CD, Trieu V, Williams PJ, De T, Yang A,

    Beals B, Figg WD, Hawkins M, Desai N. 2005. Comparative preclin-

    ical and clinical pharmacokinetics of a cremophor-free, nanoparticle

    albumin-bound paclitaxel (ABI-007) and paclitaxel formulated in Cre-

    mophor (Taxol). Clin Cancer Res 11:41364143.

    77.Gardner ER, Dahut WL, Scripture CD, Jones J, Aragon-Ching JB,

    Desai N, Hawkins MJ, Sparreboom A, Figg WD. 2008. Randomized

    crossover pharmacokinetic study of solvent-based paclitaxel and nab-

    paclitaxel. Clin Cancer Res 14:42004205.

    78.Kim T-Y, Kim D-W, Chung J-Y, Shin SG, Kim S-C, Heo DS, Kim

    NK, Bang Y-J. 2004. Phase I and pharmacokinetic study of Genexol-

    PM, a cremophor-free, polymeric micelle-formulated paclitaxel, in pa-tients with advanced malignancies. Clin Cancer Res 10:37083716.

    79.Ghuman J, Zunszain Pa, Petitpas I, Bhattacharya Aa, Otagiri M,

    Curry S. 2005. Structural basis of the drug-binding specificity of human

    serum albumin. J Mol Biol 353:3852.

    80.Colmenarejo G. 2003. In silico prediction of drug-binding strengths

    to human serum albumin. Med Res Rev 23:275301.

    81.Dong X, Mattingly CA, Tseng MT, Cho MJ, Liu Y, Adams

    VR, Mumper RJ. 2009. Doxorubicin and paclitaxel-loaded lipid-

    based nanoparticles overcome multidrug resistance by inhibiting P-

    glycoprotein and depleting ATP. Cancer Res 69:39183926.

    82.Wasan KM, Brocks DR, Lee SD, Sachs-Barrable K, Thornton SJ.

    2008. Impact of lipoproteins on the biological activity and disposition

    of hydrophobic drugs: Implications for drug discovery. Nat Rev Drug

    Discov 7:8499.

    83.Paal K, Shkarupin A, Beckford L. 2007. Paclitaxel binding to hu-man serum albuminAutomated docking studies. Bioorg Med Chem

    15:13231329.

    84.Fasano M, Curry S, Terreno E, Galliano M, Fanali G, Narciso P,

    Notari S, Ascenzi P. 2005. The extraordinary ligand binding properties

    of human serum albumin. IUBMB Life 57:787796.

    85.Elzoghby AO, Samy WM, Elgindy Na. 2012. Albumin-based

    nanoparticles as potential controlled release drug delivery systems.

    J Control Release 157:168182.

    86.Colmenarejo G, Alvarez-Pedraglio A, Lavandera JL. 2001. Chemin-

    formatic models to predict binding affinities to human serum albumin.

    J Med Chem 44:43704378.

    87.Hellstrand E, Lynch I, Andersson A, Drakenberg T, Dahlback B,

    Dawson Ka, Linse S, Cedervall T. 2009. Complete high-density lipopro-

    teins in nanoparticle corona. FEBS J 276:33723381.

    88.Oda MN, Hargreaves PL, Beckstead Ja, Redmond Ka, van Antwer-pen R, Ryan RO. 2006. Reconstituted high density lipoprotein enriched

    with the polyene antibiotic amphotericin B. J Lipid Res 47:260267.

    89.Tsukioka Y, Matsumura Y, Hamaguchi T, Koike H, Moriyasu F,

    Kakizoe T. 2002. Pharmaceutical and biomedical differences between

    micellar doxorubicin (NK911) and liposomal doxorubicin (Doxil). Can-

    cer Sci 93:11451153.

    90.Duncan R. 2006. Polymer conjugates as anticancer nanomedicines.

    Nat Rev Cancer 6:688701.

    91.Boddy AV, Plummer ER, Todd R, Sludden J, Griffin M, Robson L,

    Cassidy J, Bissett D, Bernareggi A, Verrill MW, Calvert AH. 2005. A

    phase I and pharmacokinetic study of paclitaxel poliglumex (XYOTAX),

    investigating both 3-weekly and 2-weekly schedules. Clin Cancer Res

    11:78347840.

    DOI 10.1002/jps.23996 Norvaisas and Ziemys, JOURNAL OF PHARMACEUTICAL SCIENCES 103:21472156, 2014

  • 8/11/2019 The Role of Payload Hydrophobicity in Nanotherapeutic

    10/10

    2156 RESEARCH ARTICLE Pharmaceutical Nanotechnology

    92. Hamaguchi T, Kato K, Yasui H, Morizane C, Ikeda M, Ueno H,

    Muro K, Yamada Y, Okusaka T, Shirao K, Shimada Y, Nakahama H,

    Matsumura Y. 2007. A phase I and pharmacokinetic study of NK105, a

    paclitaxel-incorporating micellar nanoparticle formulation. Br J Can-

    cer 97:170176.

    93.Hamaguchi T, Matsumura Y, Suzuki M, Shimizu K, Goda R, Naka-

    mura I, Nakatomi I, Yokoyama M, Kataoka K, Kakizoe T. 2005. NK105,

    a paclitaxel-incorporating micellar nanoparticle formulation, can ex-

    tend in vivo antitumour activity and reduce the neurotoxicity of pacli-

    taxel. Br J Cancer 92:12401246.

    94. Blanco E, Hsiao A, Mann AP, Landry MG, Meric-Bernstam F,

    Ferrari M. 2011. Nanomedicine in cancer therapy: Innovative trends

    and prospects. Cancer Sci 102:12471252.

    95.Gaucher G, Dufresne M-H, Sant VP, Kang N, Maysinger D, Leroux

    J-C. 2005. Block copolymer micelles: Preparation, characterization and

    application in drug delivery. J Control Release 109:169188.

    Norvaisas and Ziemys, JOURNAL OF PHARMACEUTICAL SCIENCES 103:21472156, 2014 DOI 10.1002/jps.23996