Sunteți pe pagina 1din 17

Article

pubs.acs.org/molecularpharmaceutics

Click Biotinylation of PLGA Template for Biotin Receptor Oriented


Delivery of Doxorubicin Hydrochloride in 4T1 Cell-Induced Breast
Cancer
Yuvraj Singh,† K.K. Durga Rao Viswanadham,†,# Arun Kumar Jajoriya,‡ Jaya Gopal Meher,† Kavit Raval,†
Swati Jaiswal,§ Jayant Dewangan,∥ H. K. Bora,⊥ Srikanta Kumar Rath,∥ Jawahar Lal,§
Durga Prasad Mishra,‡ and Manish K. Chourasia*,†

Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India

Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
§
Pharmacokinetics and Metabolism Division, CSIR-Central Drug Research Institute, Lucknow 226031, India

Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow 226031, India

Laboratory animals facility, CSIR-Central Drug Research Institute, Lucknow 226031, India
*
S Supporting Information

ABSTRACT: PLGA was functionalized with PEG and biotin using click
chemistry to generate a biotin receptor targeted copolymer (biotinylated−
PEG-PLGA) which in turn was used to fabricate ultrafine nanoparticles
(BPNP) of doxorubicin hydrochloride (DOX) for effective delivery in 4T1
cell induced breast cancer. However, adequate entrapment of a hydrophilic
bioactive like DOX in a hydrophobic polymer system made of PLGA is not
usually possible. We therefore modified a conventional W/O/W emulsion
method by utilizing NH4Cl in the external phase to constrain DOX in dis-
solved polymer phase by suppressing DOX’s inherent aqueous solubility as
per common ion effect. This resulted in over 8-fold enhancement in
entrapment efficiency of DOX inside BPNP, which otherwise is highly
susceptible to leakage due to its relatively high aqueous solubility. TEM
and DLS established BPNP to be sized below 100 nm, storage stability
studies showed that BPNP were stable for one month at 4 °C, and in vitro release suggested significant control in drug release.
Extensive in vitro and in vivo studies were conducted to propound anticancer and antiproliferative activity of BPNP. Plasma and
tissue distribution study supplemented by pertinent in vivo fluorescence imaging mapped the exact fate of DOX contained inside
BPNP once it was administered intravenously. A comparative safety profile via acute toxicity studies in mice was also generated
to out rightly establish usefulness of BPNP. Results suggest that BPNP substantially enhance anticancer activity of DOX while
simultaneously mitigating its toxic potential due to altered spatial and temporal presentation of drug and consequently deserve
further allometric iteration.
KEYWORDS: nanoparticles, common ion effect, PLGA, doxorubicin hydrochloride, click chemistry, active targeting

■ INTRODUCTION
Doxorubicin hydrochloride (DOX) is a drug of choice in a variety
tetracycline quinoid aglycone doxorubicinone and amino sugar
(daunosamine) linked together by a glycosidic linkage (Figure 1),
of cancerous ailments. DOX along with adjuvants is recom- which is extremely susceptible to acidic degradation prevalent in
mended as a first line anticancer drug in NCCN’s 2016 breast gastro intestinal tract.4
cancer guidelines.1 It interferes in DNA synthesis by binding to This attribute rules out oral administration and necessitates
topoisomerase II, damages preexisting DNA via intercalation, usage of higher parenteral dose which inadvertently contributes
generates reactive oxygen species, induces mitochondrial mem- toward cardiac toxicity. DOX is usually administered intrave-
brane depolarization by releasing cytochrome c, and obliterates nously as a hydrochloride salt and therein lays a major delivery
membranous integrity of treated cells.2 When used as a mono- related stumbling block. It is inconceivable to adequately entrap a
therapeutic option, after relapse or failure of combination hydrophilic moiety (aqueous solubility of DOX is 10 mg/mL) in
therapy, DOX induces response in 40% first-line cases and 20%
second-line cases. Despite these attributes, DOX is connected to Received: April 14, 2017
numerous side effects (dose-dependent cardiotoxicity, nausea, Revised: June 20, 2017
myelotoxicity, gastrointestinal shedding, and acute cramps) which Accepted: June 21, 2017
restrict its utility in a palliative environment.3 DOX consists of Published: June 21, 2017

© 2017 American Chemical Society 2749 DOI: 10.1021/acs.molpharmaceut.7b00310


Mol. Pharmaceutics 2017, 14, 2749−2765
Molecular Pharmaceutics Article

load of doxorubicin hydrochloride (BPNP) were prepared via


a common ion effect modified W/O/W emulsion method.
Extensive studies were conducted to propound anticancer
activity of developed nanoparticles in 4T1 breast cancer cells.
Pharmacokinetic intonation in DOX brought about by its load-
ing in nanoparticles was observed by both LC-MS and in vivo
imaging. A comparative safety profile via acute toxicity studies in
mice was also generated. In vivo anticancer activity in 4T1 cell
induced breast cancer carrying Balb/C mice model was done to
outrightly establish utility of nanoparticles.
Figure 1. Chemical structure of DOX. Equilibrium between salt form
and ionized form of drug is rapidly attained in water, yielding to
solubilization.
■ EXPERIMENTAL SECTION
Material. Doxorubicin hydrochloride (DOX) was received as
generous gift from Fresenius Kabi, Gurgaon, India. Poly lactide-
co-glycolide (PLGA) with molecular weight 50000 Da (50:50
a hydrophobic polymer like PLGA or its derivative as drug is grade) was bought from DURECT, Birmingham, AL, USA.
subject to leakage in surrounding media during aqueous pro- Pluronic 127 (PF 127), biotin-PEG-azide conjugate, propargyl
cessing of formulation (Figure 2C). Although we, along with alcohol, copper sulfate, sodium ascorbate, fetal bovine serum
others, have attempted to use base form of DOX to ensure its (FBS), antibiotic solution (penicillin/streptomycin, 0.1% v/v),
entrapment, it all adds to tediousness of formulation develop- dialysis membrane (10000−12000 Da), ribonuclease A, and
ment causing quantifiable drug loss as the process of convert- RPMI 1640 Medium, 3-[4,5-dimethylthiazol-2-yl]-2,5- diphe-
ing DOX to its base form is inefficient.5 To circumvent such nyltetrazolium bromide (MTT) were purchased from Sigma-
inefficient techniques we have utilized principle of common ion Aldrich, St. Louis, MO, USA. 4′,6-Diamidino-2-Phenylindole,
effect which was theoretically expounded during the last century dihydrochloride (DAPI), JC-1 dye were procured from Invitro-
itself, but has not been utilized anywhere in pharmaceutics. We gen, OR, USA. Three-stage purified water was obtained from
try to increase entrapment of a highly water-soluble salt form of a Milli-Q purifier (Millipore, Milli-Q plus 185, Bedford, MA,
drug (i.e., DOX) in a hydrophobic polymer nanoparticle system USA). All solvents used were of HPLC or LC-MS grade. The
(derived from PLGA), despite drug’s tendency to leach out, by chemicals were used as provided.
suppressing its aqueous solubility via introduction of a stronger Solubility Studies. Excess of DOX was added to 5 mL water
electrolyte (NH4Cl) which bears a chloride ion common to DOX or fixed strength aqueous solutions of different electrolytes (as
in the external phase of formulation. The proposed strategy, if mentioned in Table 1) in 10 mL capped vials. The dispersions so
successful, could work for other bioactives as well (because most formed were shaken on a benchtop orbital shaker (Thermo
drugs lay in the weak electrolyte category) and potentially open Fischer, India) at room temperature (25 ± 2 °C) for a period of
up a new methodology of entrapping hydrophilic drugs in 24 h. A fixed aliquot from the supernatant of each vial was diluted
polymeric matrices. Selection of PLGA as a delivery construct for appropriately using water and subjected to chromatographic
DOX was based on PLGA’s proven industrial and commercial quantification (detailed in Supporting Information). Solubility
utility (Zoladex, Lupron, Sandostatin, AtriDox, Buduperon, and experiments were conducted in triplicate.
so on). Further advantages of PLGA can be gauged from its Development of Biotinylated−PEG-PLGA via Click
biodegradability, reproducible fabricability, and sheer number of Chemistry. Synthesis of Propargyl PLGA Conjugate (3a). A
biomedical devices/formulations undergoing advanced stages of mixture of PLGA (2a) (1 g, 0.02 mM, 1 equiv), propargyl alcohol
clinical trials.6 PLGA is famed for producing ultrafine spherical (1a) (0.6 mM, 3eq ), and N,N-dimethylaminopyridine (DMAP)
particles7 and the bi product it generates enter carboxylic acid (0.06 mM 0.3 equiv) was dissolved in 50 mL dry DCM in a
cycle, thereby undergoing innocuous elimination or reutilization 100 mL round-bottom flask and stirred in ice bath for half an
in bodily metabolomics.8 hour. To this mixture, DCC (1 equiv) dissolved in 15 mL dry
Although annals of nanotechnology-assisted anticancer DCM was added, and the sample was stirred in an ice bath for 1 h.
delivery are heavily interspersed with studies where researchers The reaction mixture was further stirred for 24 h at room tem-
have exploited minute dimensions of PLGA nanoparticles to perature. Reaction was monitored by IR analysis. After comple-
passively target tumor cells, it is generally agreed that specific tion, reaction mixture was filtered. Filtrate was washed with water
targeting with limited/no off target effects as postulated by (2 × 40 mL) and brine (1 × 40 mL), dried over anhydrous
Ehlrich in “magic bullet” concept is only realizable via active Na2SO4, and evaporated. After removal of solvent under vacuum,
targeting.9 We consequently decorated PLGA with biotinylated- extract was washed with ether and recrystallized with ethyl
PEG by utilizing click chemistry to generate a cancer cell homing acetate and hexane (1:2) to get clickable propargyl PLGA (3a);
copolymer: biotinylated−PEG-PLGA. Presence of PEG in tem- 85% yield.
plate was expected to accord nanoparticles long circulation half- Synthesis of Biotinylated-PEG- PLGA via Click Reaction.
lives, which in turn would assist in their probability of reaching Propargyl PLGA conjugate 3a (0.500 mg, 009 mM, 1 equiv) was
tumor microenvironment10 for targeted delivery of potent cyto- dissolved in tBuOH−H2O (1:1, 10 mL) by stirring for 15 min.
toxic DOX in 4T1 cell induced allogenic breast cancer model.11 Sodium ascorbate (0.0029 mM, 0.3 equiv) was then added and
It is assumed that these nanoparticles, after accumulation at allowed to stir for 15 min followed by copper(II) sulfate (0.0027
intended destination owing to their size, would then be pre- mM 0.1 equiv). After 15 min biotin-PEG-azide (4a) (0.01 mM,
ferentially internalized by cancer cells due to specific interaction 1 equiv) was added to the mixture and it was stirred at room
of their biotin component with overexpressed biotin receptors on temperature in nitrogen for 24 h. The reaction conversion was
4T1 cells.12 In the direction of scheme depicted in Figure 2, confirmed by IR analysis. Upon completion, the mixture was
nanoparticles made of biotinylated−PEG-PLGA containing high evaporated under vacuum. The crude resultant was diluted with
2750 DOI: 10.1021/acs.molpharmaceut.7b00310
Mol. Pharmaceutics 2017, 14, 2749−2765
Molecular Pharmaceutics Article

Figure 2. Work in brief. (A) Reaction scheme for synthesis of biotinylated−PEG-PLGA using click chemistry. (B) Biotinylated−PEG-PLGA was
employed to generate nanoparticles of DOX via W/O/W emulsion method. Using a strong electrolyte like NH4Cl bearing a common ion to doxorubicin
hydrochloride in the external aqueous phase suppresses aqueous solubility of DOX and traps it in organic phase containing biotinylated−PEG-PLGA.
(D) Once polymer solvent vaporizes, biotin tagged PLGA nanoparticles with multifold higher DOX content are obtained which when tested in vivo are
expected to elicit specific cytotoxic action against biotin receptor overexpressive 4T1 breast cancer cells. Key: 1a, propargyl alcohol; 2a, PLGA; 3a,
clickable alkyne-ended PLGA; 4a, biotin-PEG-azide; 5a, biotinylated−PEG-PLGA; DCC, N,N′-dicyclohexylcarbodiimide; DMAP, 4-dimethyla-
minopyridine; tBuOH, tertiary butyl alcohol; r.t., room temperature.

water and extracted with DCM (2 × 20 mL). The extract was Preparation of Nanoparticles. Biotinylated−PEG-PLGA
washed with brine solution and dried over anhydrous sodium nanoparticles containing DOX (BPNP) were prepared by a mod-
sulfate (Na2SO4). After removal of the solvent under vacuum, ified W/O/W double emulsion solvent diffusion technique with
extract was washed with ether three times. It was then precip- minor changes for trapping hydrophilic molecule DOX.13 Briefly,
itated in DCM and excess ether at low temperature to yield final as per Table 2, a specified quantity of polymer (biotinylated−
compound with 80% yield. The final compound, 5a (biotiny- PEG−PLGA) and PF127 (3% w/v) were dissolved in 3 mL ethyl
lated−PEG-PLGA), was confirmed by 1H NMR. acetate. To this organic solution, 1 mL triple distilled water
2751 DOI: 10.1021/acs.molpharmaceut.7b00310
Mol. Pharmaceutics 2017, 14, 2749−2765
Molecular Pharmaceutics Article

Table 1. Aqueous Solubility of Doxorubicin Hydrochloride in the Presence of Different Chloride Salts
NaCl (Ki 0.0047) KCl (Ki 0.0059) NH4Cl (Ki 0.0092) ZnCl2 (Ki 0.0259)
NaCl DOX solubility (mg/mL) KCl DOX solubility (mg/mL) NH4Cl DOX solubility(mg/mL) in ZnCl2 DOX solubility (mg/mL)
(mg/mL) in presence of NaCl (mg/mL) in presence of KCl (mg/mL) presence of NH4Cl (mg/mL) in presence of ZnCl2
0 10 ± 0.5 0 10 ± 0.5 0 10 ± 0.5 0 10 ± 0.5
2 8.4 ± 0.3 2 8.1 ± 0.3 2 6.0 ± 0.2 2 3.9 ± 0.3
3 7.8 ± 0.2 3 7.5 ± 0.5 3 3.7 ± 0.3 3 3.1 ± 0.5
5 7.3 ± 0.3 5 7.4 ± 0.4 5 2.6 ± 0.6 5 2.7 ± 0.1

Table 2. Batch Details of BPNP with Selected Physicochemical Characters


batch polymer (mg) drug polymer ratio particle Size (nm) PDI ZP (mV) %EE
F1 10 ∼1:1 48 ± 4 0.18 ± 0.07 −6.1 ± 2.1 3.1 ± 1.3
F2 15 2:3 72 ± 3 0.22 ± 0.11 −7.8 ± 2.0 5.2 ± 1.5
F3 30 1:3 81 ± 3 0.18 ± 0.05 −3.9 ± 0.9 7.4 ± 2.8
F4 40 1:4 98 ± 4 0.14 ± 0.07 −3.7 ± 1.3 12.0 ± 1.6
F5 60 1:6 155 ± 5 0.23 ± 0.14 −8.4 ± 2.7 14.5 ± 2.2

containing known amount of drug was added. The mixture was electron microscopy (TEM). Briefly, diluted BPNP suspension
then sonicated over an ice bath using a probe sonicator (Sonics, was smeared onto a 300-mesh copper grid in form of a film. The
USA) at 40% amplitude for 3 min to form a W/O emulsion. The grid was subsequently coated with 2% w/v uranyl acetate. Excess
resulting primary emulsion was added to 4 mL aqueous NH4Cl solution was blotted off and the grid was dried using liquid nitro-
solution (5 mg/mL), and resonicated over an ice bath at 30% gen. Dried BPNP were observed at 80 kV by using TECHNAI G2
amplitude for 10 min to form a W/O/W double emulsion. Ethyl 20 S-TWIN (FEI Netherlands) instrument and photomicro-
acetate was eliminated by evaporation under reduced pressure, graphs of different fields at several appropriate magnifications
causing hardening of polymer around drug to form fully ripened were taken.
nanoparticles (BPNP). Nanoparticle suspension was dialyzed Dissolution study was conducted using a dialysis tube. BPNP,
against triple distilled water to remove free drug and dissolved DOX solution, and DPNP with equivalent DOX content (2 mg)
NH4Cl. Optimization studies were conducted to estimate effect were sealed in an activated dialysis bag and submerged in 50 mL
of formulation and processing variables on size and entrapment phosphate buffer saline (PBS pH 7.4). The dissolution medium
efficiency of BPNP. For comparative purposes, blank nano- was magnetically stirred at 100 rpm and temperature was main-
particles were similarly developed (PNP or NP) without any tained within 0.5 °C of 37 °C. Periodic sampling was done and
drug. The formulated nanoparticles were stored in cold refrig- amount of drug released was analyzed by reverse phase HPLC.
erated conditions until further use. The final coded formulations Obtained dissolution profile was fitted in different release models
were as follows: biotinylated−PEG-PLGA nanoparticles of DOX via linear regression method. This was done to excavate probable
(BPNP); PLGA nanoparticles of DOX (DPNP); blank/dummy mechanistics of drug release.
biotinylated−PEG-PLGA nanoparticles (NP); blank PLGA Quantitative and Qualitative Cell Uptake Study:
nanoparticles (PNP). Dependency on Biotin Receptors. Flow cytometry was
Physicochemical Characterization. Particle size and used to quantitatively assess cellular uptake of DOX-loaded
polydispersity index were measured by dynamic light scattering. nanoparticles (BPNP vs DPNP vs DOX solution). Briefly, 4T1
Zeta potential was evaluated by calculating mean electrophoretic cells were seeded in six well plates at a density of 0.5 × 105 cells
mobility of particles. Formulations were appropriately diluted per well for plate adhesion and priming. Subsequently selected
and analyzed at ambient temperature using Malvern zetasizer wells were incubated with media containing BPNP, DPNP, and
(Nano ZS, Malvern Instruments, UK).14 Entrapment efficiency DOX solution for 6 h followed by washing with PBS. Cells were
was determined by employing reverse phase liquid chromatog- then trypsinized, pelleted, resuspended in PBS, and analyzed for
raphy. Briefly, specified volume of formulation was dissolved in fluorescence produced by internalized DOX by using flow
1:1 mixture of DMSO and acetone, and vortexed for 5 min. The cytometer (FACS Calibur, software cell quest Pro). To establish
mixture was thereafter diluted with water and analyzed on a dependency of BPNP on biotin receptor mediated uptake, an
HPLC. Optimized batch was also subjected to storage stability additional well of cells was first treated with Biotin-Blocking Kit
studies at room temperature (25 ± 2 °C) and cold refrigerated (E-21390, Thermo-Fischer Scientific, India) as per manufac-
conditions (4 °C). Samples were drawn on fixed days and turers protocol to saturate the surface expressed biotin receptors
observed for increment/decrement in particle size and entrap- and subsequently subjected to BPNP for 6 h and analyzed as
ment efficiency.15 above by FACS.
DSC analysis was performed on synthesized copolymer For fluorescence microscopy, 4T1 cells were seeded in 6-well
biotinylated−PEG−PLGA, dummy nanoparticles made out of plates containing poly L-lysine coated coverslips for 24 h.
biotinylated−PEG−PLGA, doxorubicin hydrochloride, and Thereafter cells were treated with DOX solution, DPNP, and
BPNP using a differential scanning calorimeter (PerkinElmer, BPNP for 6 h. We also analyzed time dependent uptake of BPNP.
USA). Dried samples (lyophilized or otherwise) weighing approx- After staining, cells were rinsed repeatedly in PBS and rein-
imately 5 mg, were placed in hermetically sealed aluminum pans cubated with formaldehyde (4% v/v in PBS) at 37 °C for 15 min.
and heated at uniform rate of 10 °C/min up to 800 °C in an inert The fixed cells were then aspirated. The coverslip left behind,
environment maintained under dynamic nitrogen purging. Shape, containing treated cells, was mounted on slides using ProLong
size, and surface appearance of nanoparticles were analyzed by Gold Antifade Mountant with DAPI (Thermo Fisher Scientific,
2752 DOI: 10.1021/acs.molpharmaceut.7b00310
Mol. Pharmaceutics 2017, 14, 2749−2765
Molecular Pharmaceutics Article

India) and observed under a fluorescence microscope at 60× were divided into six groups of five each (n = 5). A dosing interval
(Nikon Eclipse TS100 with fluorescence attachment Nikon- of 72 h was adapted with DOX solution, BPNP, and DPNP
CHGFI). administered via intravenous entry into mice’s tail on day 0 (day
Cytotoxicity Study and in Vitro Hemolysis. MTT-based of treatment initiation), 3, 6, and 9. Another group was dosed
in vitro assay was performed to compare cytotoxic effects of similarly with PBS to form the control group. The study was
different groups on 4T1 cells. Briefly, cells (0.5 × 104 cells per ended on 27th day by sacrificing animals and harvesting
well) in 96-well culture plates were seeded overnight in a their tumors for subsequent processing [sliced into thin sections
controlled environment of 37 °C, 5% CO2. Cells were then (5−8 μm) and embedded in paraffin] which included hemotoxy-
treated with DOX solution, BPNP, DPNP, PNP, and NP dis- line and eosin staining, immune-staining for PCNA and detec-
persed in media equivalent to different concentrations of DOX. tion of apoptotic cells population through a dead end fluorom-
After 24 h, the treatment media from wells was aspirated off and etric TUNEL assay using biotin-dUTP (biotinylated deoxyur-
replaced with fresh media containing MTT solution (0.5 mg/mL) idine riphosphate) and avidin-FITC as an apoptotic marker
for 4 h. The formazan crystals so formed were dissolved by (TUNEL apoptosis detection kit, Millipore). The purported
adding 0.2 mL of DMSO to each well and absorbance of resulting efficacy of administered treatments was gauged by measuring
solution was measured at 570 nm using a multiwell scanning tumor dimensions during the study as well as on the day of
spectrophotometer (PowerWave XS, Biotek, VT, USA). Non- termination. A digital Vernier’s caliper was employed to do the
linear regression analysis was applied to determine inhibitory same.
concentration 50% (IC50) for each treatment group.16 Pharmacokinetics. Pharmacokinetic study of DOX and its
Hemo compatibility of nanoparticles was evaluated in formulations was carried out in 4T1 cell induced tumor bearing
erythrocytes isolated from blood of SD rats by centrifugation Balb/C mice. DOX solution, BPNP, and DPNP were admin-
(1000 × g for 10 min). The settled RBCs were suspended in PBS. istered intravenously to conscious mice (via the caudal vein) at a
Accurately measured 2 mL RBC suspension was mixed with dose of 4 mg/kg. Blood (approximately 100 μL) was collected by
aliquots of drug and formulation in triplicate to attain a final puncturing retero-orbital plexus with heparinised capillaries at
DOX concentration of 0.5 to 25 (μg/mL). RBCs dispersed in 0.25, 0.5, 0.75, 1, 1.5, 2, 4, 6, 8, 10, 24, 48, and 72 h post dosing.
PBS only and PBS containing 10% Triton- X were used as Sparse sampling was applied for blood collection and the total
negative and positive control, respectively. The RBC-drug volume of blood collected from each mouse was not more than
aliquots were incubated at 37 °C in an atmosphere of 5% CO2 3% of total body volume. Blood samples were centrifuged at
for 2 h. After incubation, samples were centrifuged at 800 × g for 5000 rpm for 10 min at 4 °C and plasma was separated into clean
10 min, and supernatant was collected in 96 well plates to be and neatly labeled tubes. All samples were stored at −80 °C until
analyzed spectrophotometrically at 540 nm. analysis. A different set of animals which had received the same
Cell Cycle Distribution. Propidium iodide (PI) was used as treatment as above were sacrificed at 2, 6, 10, 24, 30, and 72 h
a fluorescent marker for cell cycle analysis. 4T1 cells were grown post dosing and organs, such as spleen, kidney, and heart, and
in six well plates (106cells/well) and allowed to adhere. Free tumor were collected. Protein precipitation approach was
DOX, BPNP, and DPNP (each equivalent to 50 nM DOX, employed for sample cleanup. To the plasma sample or tissue
respectively) were added into different wells for a period of 24 h. homogenate (30 μL; blank, spiked or test), 170 μL acetonitrile
After stipulated time, cells were detached, rinsed in PBS, and containing 30% ammonium acetate buffer (pH 3.5, 0.01M) was
fixed in chilled ethanol (70% v/v). Thereafter cells were spun, added. The samples were vortex-mixed for 5 min followed by
pelleted, resuspended in 250 μL PBS, and treated with centrifugation for 10 min at 10,00 rpm. Clear supernatant
ribonuclease A (100 μg/mL) for 15 min followed by staining (80 μL) was transferred into HPLC vials and 20 μL was injected
with PI (50 μg/mL) for another 30 min. Fluorescence induced into LC-MS/MS system (refer to Supporting Information). Win
by association of PI and remnant genetic material of perme- Nonlin 6.0 (Pharsight Co., Mountain View, CA, USA) was
abilized cells was measured employing a flowcytometer (BD utilized to determine various pharmacokinetic parameters.
Biosciences, FACS Aria, Germany) and translated into the phase In Vivo Fluorescent Imaging. To complement pharmaco-
of cell cycle arrest induced by different treatments. kinetic studies, distribution and localization of DOX was
Apoptosis. To determine extent and type of apoptosis measured by using live in vivo imaging system IVIS Spectrum
induced by DOX solution, BPNP, and DPNP we utilized (Caliper Life Sciences/PerkinElmer). Noninvasive whole body
Annexin V and PI kit (Invitrogen, CA, USA). Protocol prescribed fluorescent images of DOX solution, BPNP, and DPNP (equiv-
in the kit was strictly adhered to and 4T1 cells (cultured as in cell alent to 4 mg/kg DOX) treated mice (bearing 4T cell inuced
uptake) were treated with different formulations (all equivalent tumor) were generated by subjecting them to frontal examina-
to 100 nM DOX) for 24 h. Subsequently, cells were trypsinized, tion at 535 nm. Subsequently generated emission was recorded at
spun, washed thrice with PBS, and dispersed in 500 μL binding 560 nm. Images were acquired for 1 min at pre fixed time points
buffer. Thereafter, cells were incubated for 30 min in dark after (5 min, 6 and 24 h after dosing) and emission emanating from
adding annexin V-FITC (5 μL) and PI (5 μL). Finally these cells mice was analyzed using Living Image Software 4.4. In order to
were subjected to flow cytometry so as to differentiate their facilitate accurate imaging, mice were preanaesthetized using
staining pattern. ketamine (20 mg/kg i.p.) before their placement inside imaging
In Vivo Antitumor Activity. Five to seven week old female chamber of instrument.
Balb/C mice weighing 15−25 g acted as model for breast cancer. Toxicity Study. Acute toxicity study was performed on
Murine mammary carcinoma cell line (4T1) was injected sub- six week old Balb/C mice (weighing ∼20 g) according to insti-
cutaneously into fat pad of mammary gland in lower right tutionally approved protocol. Mice were divided into four groups
quadrant of mice’s abdomen (1 × 106 cells/mice). After a waiting of five animals each. The first group acted as control and was
period of 10 days post inoculation (tumors became palpable from treated with PBS via tail vein whereas the second, third, and
7 days on), a careful screening was done to select animals with fourth group received DOX solution, DPNP, and BPNP, respec-
adequate tumor volume (100−150 mm3). The selected animals tively at a dose equivalent to 10 mg/kg DOX. This dosing cycle
2753 DOI: 10.1021/acs.molpharmaceut.7b00310
Mol. Pharmaceutics 2017, 14, 2749−2765
Molecular Pharmaceutics Article

was repeated thrice, with 72 h between each dose. After 14 days, N−CH2), 2.90−2.86 (m, 2H, CHaHbS), 2.74−2.72 (m,
all animals were sacrificed and major organs, such as liver, heart, CHaHbS) 2.60−2.59 (m, 2H, O−CH2), 2.19 (t, J= 7.45 Hz,
and kidney, were harvested. For histological examination excised 2H, CH2-CO) 1.72−1.55 (m, PLA- CH3+ 6H, 3 × CH2);
organs were fixed in formalin (10% in normal saline), rooted in 1.46−1.41 (m, 2H, CH2) (Figure S3).
paraffin blocks, sliced to very thin sections using automatic micro Exploring Common Ion Effect and Its Influence on DOX
tome (Leica, model- 2155), and stained with hemotoxyline and Solubility. In our earlier effort, we had tried to circumvent low
eosin. The sections were observed under an optical microscope entrapment of DOX by hyodrophobicizing DOX into its base
to grossly map their cellular architecture (Eclipse 50i, Nikon, form: this involved subjecting DOX to triethylamine treatment
Japan). For biomarker assay, heart of each animal was purged in followed by extraction with chloroform. However, extraction
liquid nitrogen at the moment of excision, thereafter cut into was never absolute and there was perpetual drug loss even
weighted pieces and homogenized in PBS (weight to volume before delivery system was fabricated.5a Trying to find alternate
ratio of 1:5) at 20000 rpm for 3 min using an ULTRA TURRAX methods of solubility suppression we came across a study where
T-10 basic (Ika, Germany). The tissue homogenate was used for Miyazaki et al. had suggested precaution in oral use of hydro-
estimation of catalase (CAT), malondialdehyde (MDA), and chloride salts (papaverine and demeclocycline hydrochlorides)
superoxide dismutase (SOD). All three enzymes were estimated as their solubility was heavily suppressed due to abundant
as per protocols prescribed in their respective estimation kits chloride ion (common ion) present in gastric secretions.18
(Sigma-Aldrich, St. Louis, MO, USA) Taking note we tried common ion effect to reduce aqueous
Statistical Evaluation. Statistical appraisal of stability batches solubility of DOX. We did so by tuning solubility of DOX in the
was carried out by applying one way analysis of variance presence of chloride containing strong electrolytes like NaCl,
(ANOVA) followed by Dunnett’s Multiple Comparison Test NH4Cl, ZnCl2, and KCl. All solubility experiments were
using readings of day 1 as control. Results of in vivo efficacy conducted at room temperature. Saturation solubility of DOX
studies were compared by two way ANOVA followed by Bon- in water as per HPLC was 10 ± 0.5 mg/mL and modulation
Feroni post-test. Point to point comparison required in experi- attained in its solubility in the presence of different salts is shown
ments, such as IC50, were done with unpaired t tests. p < 0.05, Table 1.
p < 0.01, and p < 0.001 acted as different significance levels. All There was almost 5-fold reduction in solubility of DOX when
calculations were carried out using Graph pad Instat software 5.0 it was codissolved with 5 mg/mL NH4Cl or ZnCl2. Exact order
(Graph Pad Software Inc., CA, USA). of solubility reduction was as follows: NH4Cl (2.6 ± 0.6 mg/mL)

■ RESULTS AND DISCUSSION


Synthesis of Biotinylated−PEG-PLGA Using Click
∼ ZnCl2 (2.7 ± 0.1 mg/mL) ≫ NaCl (7.3 ± 0.3 mg/mL) ∼ KCl
(7.4 ± 0.4 mg/mL). Obtained DOX concentration in the pre-
sence of different salts (Sb) is plotted as a function of salt con-
Chemistry. The first premise of our work was to synthesize a centration according to empirical Setschenow equation in
ligand tagged PLGA copolymer: biotinylated−PEG-PLGA (5a). Figure S4 of Supporting Information.19
We proceeded as per scheme in Figure 2A. In order to make log S0/S b = K i[chloride salt]
PLGA (1a) clickable, we introduced an alkyne group in its struc-
ture by exploiting its terminal carboxylate residue. To do so, an Where S0 is saturation solubility of DOX alone, Sb is solubility of
esterification reaction was carried out with hydroxyl group of DOX in the presence of chloride salt, and Ki is salting out
propargyl alcohol (2a) using DCC coupling. Thereafter, formed constant attributable to that common chloride ion which reduces
propargyl PLGA conjugate (3a) was subjected to click condensa- solubility of DOX. Consistent with results in Table 1, ZnCl2
tion with commercially available Biotin-PEG-azide (4a). The (0.0259 mM−1) and NH4Cl (0.0092 mM−1) had highest salting
reaction underwent completion at room temperature under out constants. A higher salting out constant can be interpreted as
appropriate catalysis (sodium ascorbate and copper sulfate).17 greater capacity of the selected salt to suppress solubility of DOX.
Refer to Supporting Information for IR spectrum (Figure S1). Based on solubility data and estimated salting-out constants,
Product formation was confirmed by 1H NMR. NH4Cl and ZnCl2 were selected for further investigation of
NMR Results. PEG-Biotin Azide (4a). Purchased from Sigma- common ion effect on drug entrapment. A diagrammatic repre-
Aldrich. White solid; 1H NMR (500 MHz, CDCl3) δ = 4.51−4.48 sentation of how common ion effect reduces aqueous solubility
(m, 1H, NHCHCH), 4.32−4.29 (m, 1H, NHCHCH2), of DOX when an electrolyte bearing a common counterion
3.68−3.61 (m, 8H, 4 × O-CH2), 3.57−3.55 (m, 2H, CH2-O), (chloride) is added to its solution is shown in Figure 2. Upon
3,45−3,38 (m, 1H, S-CH), 3.16−3.12 (m, 2H, CH2NH), solubilization DOX yields a positively charged doxorubicin ion
2.91−2.87 (m, 1H, CHaHbS), 2.72−2.71 (m, 1H, CHaHbS) 2.22 and a chloride ion. In accordance with Le Chatelier’s principle if
(t, J = 7.39 Hz, 2H, H2C − CO), 1.72−1.65 (m, 6H, 2 × CH2); concentration of any one of the ions is increased (chloride in our
1.47−1.41 (m, 2H, CH2). Refer to Supporting Information for case by addition of a chloride bearing strong electrolyte), ion
spectrum (Figure S2). product of dissolved ions will exceed solubility product of DOX,
Propargyl PLGA Conjugate (3a). White solid; 1H NMR shifting ionic equilibrium toward left which in turn forces DOX
(500 MHz, CDCl3) 5.30−5.17 (m, CH, PLG) 4.89−4.37 to precipitate out of solution by combining with some dissolved
(m, CH2-PGA+ CH2−O-CO) 2.46O−CH2 (t, J = 7.26), chloride ion.20 Precipitation of DOX will continue until ion
1.72−1.55 (m, PLA- CH3). Refer to Supporting Information for product is equal to solubility product and as a result overall
spectrum (Figure S3). solubility of DOX is reduced many folds. Practically, it would be
Biotinylated−PEG-PLGA (5a). White solid; 1H NMR (500 MHz, possible to reduce solubility of DOX even further by raising
CD3OD) δ = 7.36 (s, 1H triazole proton), 5.25−5.16 (m, CH, dissolved chloride ion concentration. However, increasing salt
PLA) 4.90−4.76 (m, CH2-PGA+ CH2−O-CO), 4.45−4.43 content beyond 5 mg/mL (for all the selected salts) led to
(m, 1H, NHCHCH), 4.29−4.26 (m, 1H, NHCHCH2), cracking of emulsion in downstream formulation step. Also,
3.60−3.64 (m, 8H, 4 × O-CH2) (m, 6H), 3.54−3.52 (m, 2H, formulation manufactured using ZnCl2 at any concentration
CH2-O), 3.42−3.39 (m, 1H, S-CH), 3.38−3.20 (m, 2H experienced phase separation and we consequently used NH4Cl
2754 DOI: 10.1021/acs.molpharmaceut.7b00310
Mol. Pharmaceutics 2017, 14, 2749−2765
Molecular Pharmaceutics Article

Figure 3. Physicochemical characteristics, stability profile, and in vitro release of BPNP. (A) Particle size, (B) PDI and zeta potential, and
(C) entrapment efficiency of different batches of BPNP. (D) Effect of NH4Cl concentration on entrapment efficiency of DOX. (E) Measurements of
particle size and entrapment efficiency at different storage conditions during the course of stability study of F4. (n = 3; * p < 0.05, ** p < 0.01, *** p < 0.001,
n.s. not significant). Refer to Table 2 for further batch details. (F) Characterization of biotinylated−PEG-PLGA, dummy nanoparticles, DOX and BPNP
using differential scanning calorimetry. (G) TEM images of BPNP. Scale bar represents 100 nm. (H) Comparative dissolution profile of DOX solution,
DPNP, and BPNP in PBS pH 7.4. (I) Controlled release behavior offered by BPNP kicks in straightaway as suggested by the inset of initial time points.

at its highest permissible concentration of 5 mg/mL to exert effect modified W/O/W double emulsion method to manu-
common ion effect. facture BPNP (Batches: F1 to F5). It involved emulsifying a
Formulation Dynamics in the Presence of a Common primary W/O emulsion with drug containing aqueous phase
Ion: Why Entrapment Is Favored. We utilized common ion in a secondary external aqueous phase containing dissolved
2755 DOI: 10.1021/acs.molpharmaceut.7b00310
Mol. Pharmaceutics 2017, 14, 2749−2765
Molecular Pharmaceutics Article

NH4Cl (5 mg/mL). The oil phase consisted of ethyl acetate and carboxylate charge like in studies conducted on PLGA nano-
polymer (biotinylated−PEG-PLGA), whereas surfactant, plur- particles elsewhere.26
onic 127 (PF 127) was expected to be at its most convenient site Particle size and entrapment efficiency were given highest
of residence, i.e., oil−water interface.14 If we comprehend an priority during formulation optimization. It was found that
infinitesimally small duration which follows introduction of drug increasing content of polymer [drug polymer ratio (DPR) from
containing primary W/O emulsion droplet into NH4Cl con- 1:1 to 1:6] had a direct influence on both of these parameters.
taining external aqueous phase, under ultrasonic stimulation, two Best formulation with compliant size was F4 (DPR 1:4; size
concurrent dynamic processes responsible for raised entrapment 98 ± 4.58 nm). It had 12.0 ± 1.6% drug entrapment which was
efficiency of BPNP can be visualized. significantly greater (p < 0.01) than F3 (DPR 1:3) with drug
(1) W/O droplet is disrupted into many smaller droplets due entrapment of 7.4 ± 2.8%. Entrapment did not increase any
to intense energy produced by implosion of cavitation further (p > 0.05) when DPR was increased to 1:6 in F5 (14.50 ±
bubbles created by ultrasonic probe.21 This raises inter- 2.2%), but there was substantial size increase (from 98 ± 4.5 nm
facial energy of W/O/W system exponentially destabiliz- for F4 to 155 ± 5 nm for F5). Keeping size and entrapment in
ing the emulsion system despite it having simultaneously mind we did not iterate drug polymer combinations any further
attained increment in overall entropy.22 The only plausible and selected F4 as the optimum batch. Once size and loading
way to counter this instability is recoalescence of O/W protocols were optimized for BPNP, entrapment was maximized
droplets to reduce surface area. However, steric hindrance by manufacturing F4 in the presence of varying concentration of
offered by interfacially located PF 127 implies that distance NH4Cl.
of closest approach between two emulsified droplets is Effect of Ammonium Chloride on Drug Entrapment:
substantially increased.23 O/W droplets are therefore in a Storage Stability of Formulation. As suggested by Figure 3D,
state of suspended animation inside W/O/W emulsion, entrapment efficiency of BPNP increased in direct correlation
whereby they are constantly trying to reattach. to NH4Cl concentration in external aqueous phase of W/O/W
emulsion. Maximum entrapment (90 ± 3.2%) was obtained
(2) Both internal (DOX at 10 mg/mL) and external phase
when 5 mg/mL NH4Cl was used which is more than 8-fold
(NH4Cl at 5 mg/mL) consist of dissolved solutes. How-
greater than what was obtained in batches manufactured in
ever, for fixed 1 mL internal and 4 mL external phase
absence of NH4Cl. This ability to vary entrapment efficiency
volumes, the osmotic pressure exerted by NH4Cl in
favorably establishes the second premise of our work, i.e., to
external phase (2.22 atm) is roughly 5 fold greater than
adequately capture a hydrophilic salt like DOX in an optimally
that exerted by DOX in the internal phase (0.41 atm).
sized carrier system derived from a robust and safe polymer like
Osmotic pressure was calculated by employing van’t Hoff’s PLGA. Batch F4 was taken forward for further in vitro and in vivo
law; molar concentrations of solute in the internal and exter- testing. BPNP were tested for storage stability at room tem-
nal phases, respectively, at room temperature, i.e., 298 K perature (25 ± 2 °C) and in a refrigerator (4 °C). Particle size
with universal gas constant of 0.08206 L atm mol−1 K−1. and entrapment efficiency of BPNP after 30 days at 4 °C did
This osmotic pressure gradient effectively drags out water not register any significant alterations. However, particle size
from innermost phase toward the bulk.24 Intuitively it at 25 ± 2 °C increased rapidly to 135 ± 4.3 nm after 30 days
would seem that DOX will also be carried out with this (Figure 3E). One way ANOVA followed by comparative post-
hydrostatic drag to solubilize freely in bulk aqueous phase, test against readings of day 1 revealed significantly greater incre-
but existence of common chloride ion in external phase ment (p < 0.001) in particle size when BPNP were stored beyond
due to dissolved NH4Cl massively reduces doxorubicin 4 days at room temperature. Drug entrapment was also substan-
hydrochloride’s water solubility and constrains it in the tially reduced (75.6 ± 3.5%) at the end of 30 day testing period
oil phase away from aqueous bulk. Once ethyl acetate is (p < 0.001). Storage at room temperature increases kinetic
evaporated under vacuum, dissolved polymer congeals in energy of BPNP suspension, promoting collisions, which escalates
shape of its ancestral oil droplet to form regularly shaped chances of particle aggregation and subsequent growth.13 To sum
spherical nanoparticles trapping DOX which in actuality up it was found that BPNP were stable enough in refrigerator
had nowhere else to go due to influence of common ion conditions for a period of one month negating chances of any false
effect in the external phase. positives or negatives creeping into the study due to usage of a
Preliminary Optimization of BPNP. Size plays a pivotal stored formulation during that one month window.
role in passive targeting of cancerous site due to EPR effect. DSC. An overlay of DSC curves for BPNP, biotinylated−PEG-
Retrospective analysis reveals that in order to capitalize on EPR PLGA, dummy nanoparticles made from biotinylated−PEG-
effect, a carrier must be sized between 10 and 100 nm. Entities PLGA (NP) and DOX is provided in Figure 3F. DOX showed an
smaller than 10 nm are either washed away by glomerular endothermic peak at 241.76 °C which was in close agreement to
filtration or are extravasated at unwanted sites and those larger its reported melting point of 230 °C.27 An endothermic peak at
(∼100 nm upward) are trapped by reticuloendothelial system.25 375.37 °C indicative of thermal degradation was observed for
Therefore, one of our objectives was to keep BPNP within biotinylated−PEG-PLGA. DSC thermogram of blank nano-
touching distance of above prescribed size range (10−100 nm), particles of biotinylated−PEG-PLGA and BPNP had almost
so as to maximize any advantage offered by biotinylation of coincident endothermic peaks between 310 and 315 °C. How-
PLGA. BPNP were subjected to routine physicochemical char- ever, both of these peaks were less sharply defined than their
acterization like zeta potential, PDI, particle size, and entrap- parent copolymer i.e. biotinylated−PEG-PLGA. Shifting of
ment efficiency as shown in Figure 3A, B, and C. Particle size of thermal degradation peak from 375.37 °C to 310−315 °C
different batches was found in range of 40 to 150 nm, whereas suggests: biotinylated−PEG-PLGA when formulated into nano-
PDI was below 0.3 suggesting particle uniformity. Zeta potential particles degrades faster due to exposition of larger surface area
as expected was slightly negative between −1 to −8 mV due to and/or it has been reduced to a comparatively more amorphous
biotinylated−PEG-PLGA backbone which might have residual state.28
2756 DOI: 10.1021/acs.molpharmaceut.7b00310
Mol. Pharmaceutics 2017, 14, 2749−2765
Molecular Pharmaceutics Article

Figure 4. In vitro cell culture assays. (A) Quantitative cellular uptake of BPNP in 4T1 cells. 4T1 Cells were exposed to media containing (i) blank
nanoparticles, (ii) DOX solution, (iii) DPNP, and (v) BPNP at a concentration equivalent to 100 nm DOX for 6 h.(iv) A seeded well presaturated with
biotin was also used to investigate whether blockade of biotin receptor induces variation in BPNP uptake. Standard deviation in
MFI values were calculated for cells from 3 tests. (C) Results from MTT assay. BPNP (91.5 ± 13.7 nM) exhibit 2 fold reduction in IC50 of DOX
solution (179.5 ± 11.8 nM) whereas NP and PNP were inert. (C) BPNP cause lysis of less than 10% incubated erythrocytes at 10 μg/mL. Hemolysis was
nonexistent for NP or PNP. (D) Apoptosis and (E) cell cycle analysis was conducted at concentration equivalent to 100 nM and 50 nM DOX,
respectively, for a treatment period of 24 h. BPNP cause maximum early and late apoptosis (59% cells) and G2-M phase arrest (93.44%).

An amorphous substance has less ordered structure and between BPNP and DOX solution at all-time points, suggesting
consequently requires lesser energy to undergo thermal degrada- modulation in drug release was induced by usage of biotinylated−
tion. Since BPNP (265 mJ) requires lesser energy compared to PEG-PLGA. Obtained dissolution data for BPNP was fitted into
blank biotinylated−PEG-PLGA nanoparticles (316 mJ) and release kinetic models like Hixson-Crowell (r2 = .95), Korsmeyer
biotinylated−PEG-PLGA (395 mJ) it might have greater amor- peppas (r2 = .96, n = 0.70), Higuchi (r2=.98), zero order (r2 = .92),
phous character. Dissolution of polymer in an organic solvent and first order (r2 = .95), to gain an insight into mechanism of
followed by its co-precipitation with DOX into aqueous phase drug release. Since r2 values did not differ much and most of the
during formulation processing might be responsible for such a models had comparable fit we could not select any mechanism
shift in phase transition. Also disappearance/merging of endo- which out rightly dominated drug release. Literary sources also
thermic peak of DOX with the main endothermic peak of BPNP lend weight to our findings as they propose multiple com-
suggest that it might have been rendered completely amorphous binatorial mechanisms (like diffusion through particle matrix
and is uniformly dissolved in BPNP.29 either independently or following matrix disintegration, desorp-
In Vitro Drug Release. Comparative in vitro dissolution tion of surface-adhered drug, diffusion through polymer wall,
study for DOX solution, DOX loaded regular PLGA nano- surface and bulk degradation of polymer exposing housed drug to
particles (DPNP), and BPNP was conducted in PBS pH 7.4 and dissolution media) responsible for drug release in case of PLGA-
the results were plotted versus time in Figure 3H,I. Equilibration derived nanoparticles.30 Therefore, it will not be an over exag-
of DOX solution with dissolution media was seamless with geration to assume BPNP, which are made up of a derivatized
46.0 ± 2.6% drug content available within first 3 h followed by form of PLGA, also behave similarly.
net release of 86.3 ± 1.0% in 24 h. In contrast, BPNP and DPNP Quantitative Cell Uptake. DOX is an inherently florescent
released only 30.00 ± 2.9% and 32.8 ± 3.2% of their respective molecule which enables direct analysis of its uptake by 4T1 cells
drug contents in first 12 h of release study. Two-way ANOVA using flow cytometry (Figure 4A). Results revealed that BPNP
revealed significant difference (p < 0.05) in drug release pattern had adequate uptake in 4T1 cells. Median florescence intensity
2757 DOI: 10.1021/acs.molpharmaceut.7b00310
Mol. Pharmaceutics 2017, 14, 2749−2765
Molecular Pharmaceutics Article

Figure 5. Pictorial evidence of time dependent BPNP uptake in 4T1 cells. The drug seems to localize completely in nuclear region at 6 h after BPNP
treatment. However, comparative fluorescence from DPNP and DOX solution treated cells at 6 h is much lesser than that produced by BPNP. Scale bar
represents 30 μm.
(MFI) counted in 10000 cells after BPNP treatment (214 ± 19) dimension for an entity to undergo endocytosis (a specialized
was 20-fold higher (p < 0.001) than untreated control cells uptake mechanism existent in almost all cell types,) should be
(9.8 ± 2.1), 7-fold higher than DOX solution (36.9 ± 3.0), and below 100 nm.31 Since average particle size of DPNP and BPNP
2-fold greater than DPNP (113.4 ± 10.4) treated cells. Usually fall in this range, expected cell uptake should have been the same.
2758 DOI: 10.1021/acs.molpharmaceut.7b00310
Mol. Pharmaceutics 2017, 14, 2749−2765
Molecular Pharmaceutics Article

But surface decoration of PLGA with biotin, lends BPNP a they proceed with damaged DNA leading to cell death (mostly
recognizable epitope which causes its greater internalization by by apoptosis) after division.34
4T1 cells due to receptor mediated facilitatory uptake. We Since BPNP cause overt G-2 M phase arrest there is high
suspect biotinylation of PLGA leads to advertent targeting and probability its DOX content has profusely damaged DNA of
rapid cell uptake of carrier.32 Relatively high uptake of BPNP, in almost all the cells which necessitates activation of G-2 M
comparison to cells treated with DOX solution and DPNP, is checkpoint for DNA repair. BPNP, DPNP, and DOX solution
expected to be of substantial significance, as it would prefer- were also quantitatively evaluated for their apoptotic potential
entially ensure higher payload delivery inside cell improving by using dual annexin V and propidium iodide staining. Early
expected anticancer activity. To showcase involvement of apoptotic cells were captured in the lower right quadrant of dot
biotin receptors on uptake of BPNP we conducted a differential plot, while the late apoptotic cells were seen in the upper right
uptake experiment where 4T1 cells were pretreated with biotin quadrants (Figure 4D). BPNP enhanced early and late apopto-
(Figure 4A (iv). It was observed that uptake of BPNP and DPNP sis (59.63%) compared to DPNP (38%) and DOX solution
in biotin-treated cells was indistinguishable. Biotin treatment (36.44%). These results were substantiated qualitatively by fluo-
here acted as a competitive blocker of biotin receptors expressed rescence microscopy described next.
on 4T1 cell surface and thereby precluded biotin receptor Time Dependent Cell Uptake of BPNP by Fluorescence
mediated endocytic uptake of BPNP, reinforcing our earlier Microscopy. Encouraged by results of quantitative uptake and
statement. in vitro cytotoxicity experiments we attempted to visually map
Cytotoxicity Assay and Hemolysis. MTT assay was fate of BPNP both with respect to time and in comparison to
performed in order to ascertain in vitro cytotoxic activity of DPNP and DOX solution (Figure 5). 4T1 were cells cultured in
BPNP. Inhibitory concentration which induced cell death in 50% 6 well plates and incubated with BPNP dispersion equivalent to
population (IC50) was calculated using nonlinear regression 100 nM DOX. At prefixed times of 1, 3, and 6 h following BPNP
(Figure 4B). BPNP was 1.44-fold more cytotoxic than DPNP treatment; cells were counterstained with nuclear dye DAPI and
[P < 0.05 (n = 3)]. IC50 values of DOX solution, DPNP, and BPNP visualized. At 1 h it was found that doxorubicin signal was
were 179.5 ± 11.8 nM, 131.8 ± 17.2 nM, and 91.5 ± 13.7 nM, observable (indicated by arrows) specifically in cytoplasmic
respectively, after 24 h. BPNP inducing a 2-fold reduction in IC50 domain only, away from an independently discernible and
[P < 0.05 (n = 3)] compared to DOX solution might be a trickle- regularly shaped nuclear region. This suggests successful
down effect of enhanced biotin receptor mediated intracellular transmigration of drug across cellular membrane via BPNP and
drug uptake. Once inside the cell, whatever drug is released by its rapid intracellular availability. At 3 h, intensity of doxorubicin
BPNP is available in vicinity of its action site thereby exerting signal was clearly enhanced signifying increase in its concen-
greater cell death. Dummy nanoparticles (NP and PNP) were tration causing co-localization with DAPI, due to DNA inter-
found to be inert and did not cause any unwanted cell death calating ability of drug. It can be assumed that DOX contained
suggesting excipients employed in formulation did not have any inside BPNP has moved to its natural site of action, i.e., DNA
unwarranted cytotoxic activity of their own. housed in nuclear region. At 6 h nuclear migration of payload
Hemolysis study was performed on resuspended erythrocytes. seems complete as BPNP produces selective co-localization with
It was observed that both BPNP and DPNP were safe on DAPI. However, at same time point, intracellular levels of DOX
hemolytic front (Figure 4C). They caused lysis in 7.8 ± 2.4% and after incubation of 4T1 cells with DPNP and DOX solution are
6.9 ± 2.1% erythrocytes at concentration equivalent to 10 μg/mL significantly lesser, as suggested by low intensity of nuclear
DOX. However, at same concentration, pure drug solution fluorescence, and it becomes easier to comprehend why BPNP
caused lysis in 16.8 ± 2.6% cells (p < 0.01). Lowering of DOX’s is much more cytotoxic. This study further substantiates the
inherent hemolytic potential by BPNP might be due to altered observations made by flow cytometry based assays and proves
drug release offered by the carrier system, which reduces direct that decoration of PLGA template with biotin leads to both rapid
exposition of erythrocytes to drug. Low hemolytic potential of and quantitatively greater intracellular delivery of DOX than
BPNP will bode well for its amicability toward intravenous what was possible with native drug solution or a simpler carrier
administration. loaded with drug.
Cell Cycle and Apoptosis. Free DOX solution, BPNP, and Since cytotoxicity experiments had already shown that dummy
DPNP were incubated with 4T1 cells for 24 h and a propidium nanoparticles constructed out of biotinylated−PEG-PLGA or
iodide-based assay was performed to assess phase in which PLGA did not produce any activity of their own, it was concurred
treatments induce cell arrest, if any (Figure 4E). Results that dynamics of cellular killing were dictated by DOX alone, and
suggested that BPNP caused massive cell arrest (93.44%) in any advantage which had been obtained thus far by BPNP must
G-2 M phase which was greater than DPNP (89.42%), DOX be a manifestation of altered intracellular DOX presentation
solution (76.49%), and untreated control cells (13.19%). This vindicating our purported hypothesis of decorating PLGA with
incremental trend can be attributed to rapid and selective uptake biotin at least on the in vitro scale.
of BPNP, which causes sustained doxorubicin delivery at In Vivo Pharmacokinetics. DOX levels attained in plasma
intracellular locations. However, significance of above statistic and in different tissues after intravenous administration of DOX
becomes apparent only if we ponder into DOX’s mechanism of solution, DPNP, and BPNP (4 mg/kg) in tumor bearing Balb/C
action. Doxorubicin hydrochloride has high affinity toward DNA mice were monitored using LC-MS. Noncompartmental ana-
causing its intercalation and subsequent distortion. Doxorubicin lysis was applied on obtained measurements to estimate pharma-
specifically inhibits progression of topoisomerase II, which cokinetic parameters for formulations in plasma and differ-
ultimately results in deformed DNA during S-phase or synthetic ing tissues, namely tumor, heart, kidney, spleen, and lung
phase of cell cycle.33 Each cell has a DNA damage check point in (Figure 6A). DOX solution in correspondence to its dissolution
G2-M phase which ensures that cells do not initiate mitosis pattern attained Cmax of 758.84 ± 57.3 ng/mL within 5 min of
before they have had a chance to repair damaged DNA syn- dosing. Since DOX has high aqueous amicability, intravenously
thesized in S-phase. If cells have a defective G2-M checkpoint applied bolus dose of DOX solution undergoes instantaneous
2759 DOI: 10.1021/acs.molpharmaceut.7b00310
Mol. Pharmaceutics 2017, 14, 2749−2765
Molecular Pharmaceutics Article

Figure 6. Pharmacokinetics and tissue distribution. (A) Comparative pharmacokinetic profile attained by DOX solution, DPNP and BPNP in plasma
and different organs after intravenous dose equivalent to 4 mg/kg in tumor bearing Balb/C mice. All plasma values have been plotted as mean ± s.d. for
three animals each. Tissue distribution has been plotted using mean values only for sake of clarity. (B) In vivo images of 4T1 tumor bearing Balb/C mice
treated intravenously with DOX solution, DPNP, and BPNP, respectively, at different time points.

equilibration with blood and yields high systemic concentration. of half-life (35.18 ± 0.36 h) compared to DOX solution and
However, rapid disposition takes hold simultaneously and drug 2-fold prolongation in comparison to DPNP (17.71 ± 1.27 h).
levels thereafter start falling with a terminal half-life of 5.39 ± Half-life extension for BPNP might be a resultant of two
0.76 h. Drug levels attributable to BPNP and DPNP had an almost overlapping phenomena: (1) avoidance of phagocytic uptake due
5-fold lower Cmax of 154.3 ± 21.4 ng/mL and 162 ± 11.69 ng/mL, to PEG component of BPNP prolonging systemic circulation of
respectively. This stemmed from control exerted by polymeric intact nanoparticles providing sustained drug levels35 and/or (2)
carriers, i.e., biotinylated−PEG-PLGA and PLGA. For BPNP, accumulation of nanoparticles in large perforated organs (like
elimination changes drastically, resulting in 7-fold prolongation spleen, liver) juxtaposed with secondary redistribution of drug
2760 DOI: 10.1021/acs.molpharmaceut.7b00310
Mol. Pharmaceutics 2017, 14, 2749−2765
Molecular Pharmaceutics Article

Figure 7. BPNP show higher antiproliferative activity. (A) Photographs were taken to visually demonstrate discernible reduction in tumor caused by
BPNP in comparison to DPNP and DOX solution. (B) BPNP produced highly significant (p < 0.001) reduction in tumor volume when compared to
DOX solution and DPNP treated tumors. (C) Tumor weight was measured at the end of study and expectedly BPNP treated mice exhibited lower
tumor burden. Tumor section of BPNP treated animal had several areas of gross cellular degeneration and matrix abnormalities which were rare in DOX
solution treated animals. Number of PCNA carrying cells (appear brown) was substantially more in DOX solution treated tumor than in BPNP treated
tumor. Green fluorescence produced by apoptotic cells in TUNEL assay was also higher in BPNP treated group. These results cumulatively suggest
multifactorial improvement in in vivo efficacy of doxorubicin hydrochloride when it is administered as BPNP (n = 5; *p < 0.05, **p < 0.01, ***p < 0.001,
n.s. = not significant). Scale bar in tumor sections, PCNA stained sections, and TUNEL assay sections are equal to 100 μm.

2761 DOI: 10.1021/acs.molpharmaceut.7b00310


Mol. Pharmaceutics 2017, 14, 2749−2765
Molecular Pharmaceutics Article

from these temporary reservoirs back into plasma.36 Long circula- produce detectable signal in most regions apart from cardiac
tion times raises probability of BPNP reaching their intended site tissue whereas DPNP seems sparsely distributed and it is difficult
of action (i.e., tumor). Lower clearance (1505 ± 38.2 mL/h/kg), to draw any conclusion. Contrarily BPNP at same time point is
extended MRT (22.56 ± 3.8 h), and higher AUC0‑∞ (2657.2 ± housed exclusively in and around tumor (encircled). BPNP’s
180.4 h*ng/mL) of BPNP when compared to DPNP and distribution pattern suggests that drug carried by it is not only
DOX solution further substantiate consequences of above targeted specifically toward tumor due to active targeting, but
description. It was also noted that BPNP elicited significantly chances of it producing any unwanted off target effects like
greater (p < 0.01) Cmax (49.4 ± 8.2 ng/mg) and AUC0‑∞ (1588 ± cardiotoxicity in comparison to DOX solution are also minimal.
218.4 h*ng/mg) than DOX solution (Cmax 11.45 ± 4.7 ng/mg, In Vivo Antitumor Efficacy. Balb/C mice carrying 4T1
AUC0‑∞ 367 ± 58.4 h*ng/mg) and DPNP (Cmax 22.45 ± breast cancer were treated intravenously with DOX solution,
6.9 ng/mg, AUC0‑∞ 763 ± 80.4 h*ng/mg) in tumor tissue. This BPNP, and DPNP at a dose equivalent to 4 mg/kg. A separate
was a strong evidence showcasing effectiveness of active group of cancerous mice was treated intravenously with PBS to
targeting. Raised drug concentration in tumor vicinity will elicit act as negative control. Results are summarized in Figure 7.
a more potent response. Further complementing favorable tumor Relative efficacy of treatments was assessed by (1) measuring
accumulation of BPNP were its low levels in heart (AUC0‑∞ tumor volume during and after the study, (2) documenting
2255 ± 35.6 ng/g*h, Cmax 36.7 ± 8.3 ng/g). Contrastingly if tumor burden, (3) examining tumor histology, (4) staining for
we note statistically greater accumulation of drug in cardiac proliferating cell nuclear antigen (PCNA), and (5) counting
tissue (AUC0‑∞ 3961 ± 43.8 ng/g*h, Cmax 149.4 ± 14.6 ng/g) apoptotic cells in tumor sections via TUNEL assay. Photographs
following administration of DOX solution, it becomes clear as to in Figure 7A visually demonstrate discernible reduction in tumor
why doxorubicin hydrohloride elicits a cardiotoxic response. caused by BPNP in comparison to DPNP and DOX solution.
Several favorable distinctions exist at critical junctures for Two way ANOVA followed by Bon Feroni post-test (Figure 7B)
BPNP which are attributable to its specific size as well as mate- revealed that BPNP produced significantly greater (p < 0.001)
rial of construction, i.e., biotinylated−PEG-PLGA. Exploiting tumor volume reduction (from 99.36 ± 4.23 to 29.74 ±
(1) size (below 100 nm), which has been promulgated to be 9.17 mm3) when compared to DOX solution (from 100.43 ±
suited toward infiltrating leaky vasculature in and around tumor, 5.20 to 76.38 ± 6.24 mm3) and DPNP (from 102.72 ± 6.80 to
and (2) biotin component which facilitates rapid uptake inside 68.43 ± 8.42 mm3). BPNP also caused maximum reduction in
4T1 cancer cells, BPNP possesses capability to deliver drug speci- tumor burden (123.5 ± 20 mg). It approximately caused 10-fold
fically to tumor site away from cardiac tissue which might tilt risk- and 3-fold reduction in tumor burden when compared to PBS
benefit ratio of doxorubicin hydrochloride favorably. Please refer and DOX solution (Figure 7C). Interestingly there was insig-
to Supporting Information sheet for detailed listing of pharma- nificant difference in antitumor efficacy of DOX solution and
cokinetic parameters. Drug was not detected in brain. DPNP. If we look at the pointed portions in H and E stained
In Vivo Imaging. Current work professes delivering doxoru- tumor sections of BPNP, there are several regions where cellular
bicin hydrochloride in vicinity of tumor for eliciting maximum matrix has undergone major degeneration. Spindle shaped
possible therapeutic response, without burdening body with regular 4T1 cells which are most prominent in PBS treated group
undue toxicity. To further strengthen this hypothesis, we sub- and to some extent in DOX solution and DPNP treated tumors
stantiated quantitative pharmacokinetic data with live in vivo seem to have given way to constricted dots which might be a
images to accurately portray exact fate of BPNP (Figure 6B). We manifestation of pyknosis associated with apoptotic cells.
did so by employing above-described pharmacokinetic model, in Considering the DNA specific mechanism of action of DOX,
which three distinct mice were treated intravenously with DOX we also gauged PCNA expression in tumor sections. It was found
solution, DPNP, and BPNP via tail vein. Intensity of signal prod- that BPNP treated tumor section had much less PCNA expres-
uced was related to amount of fluorescent moiety, i.e., doxoru- sion (seen as brown spots in Figure 7) in comparison to DOX
bicin hydrochloride. solution treated tumor. PCNA is a DNA immobilizer and acts as
At 5 min post dosing, fluorescence emanating from doxoru- a functional co factor for DNA polymerase activity in replication.
bicin hydrochloride was visible in tail region of all animals. One PCNA recruits proteins involved in DNA repair and chromatin
might argue isolated fluorescence in tail region contradicts remodelling.37 Doxorubicin causes covalent cross-linking of
plasma pharmacokinetic data (an indicator of systemic drug PCNA trimers which raises probability of DNA damage.38
presence) where Cmax was recorded for all groups at 5 min; PCNA’s down regulation after treatment with BPNP is an indi-
however (1) superficial nature of tail vein, (2) time required for cator that cellular proliferation is stalled more effectively by it in
drug to undergo adequate distribution, and (3) lack of comparison to DOX solution or DPNP. TUNEL assay showed
surrounding connective tissue in mice’s tail might be responsible that BPNP induced greater apoptotic activity compared to DOX
for this selective signal. At 6 h owing to drug distribution, solution and DPNP against 4T1 cells (as suggested by number of
fluorescence signal was observable throughout animal body for green fluorescent spots in Figure 7). These results cumulatively
DOX solution, DPNP, and BPNP; however spatial disposition corroborate improved efficacy of BPNP, and strengthen the basis
for each was different. DOX’s inherent affinity toward cardiac of lending drug loaded PLGA nanotemplate an active targeting
tissue dictated DOX solution and produced little localization in ligand biotin so as to increase in vivo antitumor efficacy of DOX.
tumor associated regions. Since PLGA controls drug release, Toxicity Study. Results of toxicity study are summarized in
cardiac sequestration of DPNP was slightly lesser than DOX Figure 8. At the forefront it was observed that BPNP treated
solution at 6 h, though it was still more than what was seen with animals did not exhibit any behavioral modulation like irritancy,
BPNP at same point. In case of BPNP, we saw selective localiza- hair fall, necrosis at the site of injection, or morbidity. It was also
tion of drug in tumor region. Armed with its sub 100 nm size and found that BPNP induced roughly 2-fold greater (p < 0.05)
surface decorated biotin moiety, BPNP spontaneously gravitates catalase activity (1.24 ± 0.20 μmol/mL/min/mg) than that of
toward tumor, releasing substantial drug at required site of PBS treated control mice (0.65 ± 0.16 μmol/mL/min/mg),
action. At 24 h DOX solution owing to its elimination does not whereas DOX solution caused more than 3-fold (p < 0.001)
2762 DOI: 10.1021/acs.molpharmaceut.7b00310
Mol. Pharmaceutics 2017, 14, 2749−2765
Molecular Pharmaceutics Article

Figure 8. Acute toxicity study. DOX solution, BPNP, and DPNP were administered at a dose of 10 mg/kg via tail vein of healthy BALB/C mice and
toxicity was evaluated 14 days post injection by analyzing (A) tissue histology and measuring relative (B) catalase activity, (C) MDA content, and
(D) SOD levels in heart. Saline treated animals displayed regular tissue architecture. DOX solution caused massive cardiac degeneration. The striations
which typify cardiac tissue seem completely obliterated. Additionally there was substantial parenchymal thinning of liver and distortion of glomerulus
(pointed). Animals treated with DPNP and BPNP however conserved cardiac architecture but there were foci of tissue breakdown. Mild portal
congestion and tubular infiltration was also visible. (n = 3; * p < 0.05, ** p < 0.01, *** p < 0.001, n.s. not significant).

catalase activation (1.92 ± 0.29 μmol/mL/min/mg). SOD levels DOX is infamous for its cardiotoxic potential. Several studies
in treated mice varied as DOX solution (42.13 ± 4.18 U/mg have shown that microsome present in organs, including heart,
cardiac tissue) > DPNP (32.89 ± 2.10 U/mg cardiac tissue) > convert DOX into highly reactive semiquinone intermediate
BPNP (29.11 ± 2.23 U/mg cardiac tissue) > PBS (21.65 ± which facilitates generation of superoxide anion (O2− and
1.89 U/mg cardiac tissue). MDA levels also followed a similar H2O2−) and causes lipid peroxidation and cell damage.39 There-
trend: DOX solution (3.82 ± 0.10 nM/mg cardiac tissue) > DPNP fore, lower levels of above-described pathological markers
(3.45 ± 0.2 nM/mg cardiac tissue) > BPNP (3.01 ± 0.23 nM/mg [catalase, superoxide dismutase (SOD), lipid peroxidation
cardiac tissue) > PBS (2.2 ± 0.05 nM/mg cardiac tissue). (MDA)] in heart of BPNP treated animals in comparison to
2763 DOI: 10.1021/acs.molpharmaceut.7b00310
Mol. Pharmaceutics 2017, 14, 2749−2765
Molecular Pharmaceutics Article

DOX solution and DPNP are indicative of relative safety of Notes


BPNP. Histological analysis was conducted to map toxic The authors declare no competing financial interest.
implication of treatments in vital organs (liver, kidney, and
heart) using an optical microscope. It was seen in photomicro-
graphs that DOX solution produced areas of severe parenchymal
■ ACKNOWLEDGMENTS
We are thankful to DBT grant BT/PR14769/NNT/28/996/
damage, obliteration of myofibrils, and infiltration of inflam- 2015 for providing financial assistance in smooth conduction of
matory cells in cardiac tissue. There was also gross hepatic this study. We appreciate facilitation of flowcytometry experi-
parenchymal degeneration and dilatation of intertubular sinus- ments by Dr. A.L. Vishwakarma and Mrs. Madhu who are
oidal spaces and glomerular degeneration. Contrarily BPNP and members of SAIF Division, CDRI. We are also thankful to
DPNP produced only mild interfibrillar thinning and moderate Dr. Chauhan from CSIR-IITR, Lucknow, for providing TEM
portal vein congestion along with traces of intertubular infiltra- facility. CSIR CDRI Communication 9507.
tion. Tissue sections of kidney, heart, and liver displayed regular
architecture when animals were treated with PBS. In light of
toxicity results it would be appropriate to assume that BPNP
■ REFERENCES
(1) Gradishar, W. J.; Anderson, B. O.; Balassanian, R.; Blair, S. L.;
is safer than DOX solution and DPNP in terms of cardio- Burstein, H. J.; Cyr, A.; Elias, A. D.; Farrar, W. B.; Forero, A.; Giordano,
compatibility and overall effect on morphology of vital organs. S. H. Invasive breast cancer version 1.2016, NCCN clinical practice
BPNP’s altered biodistribution and control exerted in drug guidelines in oncology. J Ntl Compr Canc Netw 2016, 14 (3), 324−354.
release by polymeric vehicle might be responsible for this (2) Thorn, C. F.; Oshiro, C.; Marsh, S.; Hernandez-Boussard, T.;
favorable safety profile when compared to drug alone. McLeod, H.; Klein, T. E.; Altman, R. B. Doxorubicin pathways:


pharmacodynamics and adverse effects. Pharmacogenet. Genomics 2011,
21 (7), 440−446.
CONCLUSIONS (3) Iwamoto, T. Clinical application of drug delivery systems in cancer
BPNP displayed substantial merit in enhancing anticancer chemotherapy: review of the efficacy and side effects of approved drugs.
activity of DOX while simultaneously mitigating its toxic Biol. Pharm. Bull. 2013, 36 (5), 715−8.
potential due to altered spatial and temporal presentation of (4) Sparreboom, A.; Planting, A. S.; Jewell, R. C.; van der Burg, M. E.;
drug. Since the nanoparticle template possesses capability to van der Gaast, A.; de Bruijn, P.; Loos, W. J.; Nooter, K.; Chandler, L. H.;
undergo click reactions, we assume that in future it can be Paul, E. M.; Wissel, P. S.; Verweij, J. Clinical pharmacokinetics of
decorated with a variety of counter click ligands which in turn will doxorubicin in combination with GF120918, a potent inhibitor of
MDR1 P-glycoprotein. Anti-Cancer Drugs 1999, 10 (8), 719−28.
allow clickable drug loaded template to be theoretically used for (5) (a) Singh, P. K.; Sah, P.; Meher, J. G.; Joshi, S.; Pawar, V. K.; Raval,
targeting different cancer cells expressing different receptors. We K.; Singh, Y.; Sharma, K.; Kumar, A.; Dube, A.; Chourasia, M. K.
have already applied the principle of common ion effect to tailor Macrophage-targeted chitosan anchored PLGA nanoparticles bearing
entrapment of another hydrophilic drug diethyl cabamazine doxorubicin and amphotericin B against visceral leishmaniasis. RSC Adv.
citrate in conventional PLGA nanoparticles and believe this 2016, 6 (75), 71705−71718. (b) Kansal, S.; Tandon, R.; Dwivedi, P.;
technique is ripe for others to exploit. In future endeavors we Misra, P.; Verma, P. R. P.; Dube, A.; Mishra, P. R. Development of
wish to come up with a one-step fabrication technique, where in, nanocapsules bearing doxorubicin for macrophage targeting through the
an optimally synthesized and highly drug-loaded nanoparticle phosphatidylserine ligand: a system for intervention in visceral
template is decorated with a ligand in situ via an easy to carry out leishmaniasis. J. Antimicrob. Chemother. 2012, 67 (11), 2650−2660.
click reaction after all the pharmaceutical characterization tools (6) Danhier, F.; Ansorena, E.; Silva, J. M.; Coco, R.; Le Breton, A.;
Préat, V. PLGA-based nanoparticles: An overview of biomedical
have been applied.


applications. J. Controlled Release 2012, 161 (2), 505−522.
(7) Astete, C. E.; Dolliver, D.; Whaley, M.; Khachatryan, L.; Sabliov, C.
ASSOCIATED CONTENT M. Antioxidant Poly(lactic-co-glycolic) Acid Nanoparticles Made with
*
S Supporting Information α-Tocopherol−Ascorbic Acid Surfactant. ACS Nano 2011, 5 (12),
The Supporting Information is available free of charge on the ACS 9313−9325.
Publications website at DOI: 10.1021/acs.molpharmaceut.7b00310. (8) Murphy, W. L.; Peters, M. C.; Kohn, D. H.; Mooney, D. J.
Sustained release of vascular endothelial growth factor from mineralized
HPLC method, LC-MS method, FTIR spectra with inter- poly (lactide-co-glycolide) scaffolds for tissue engineering. Biomaterials
pretation, NMR spectra, macrophage evasion study via 2000, 21 (24), 2521−2527.
flow cytometry, differential uptake studies of BPNP in (9) Acharya, S.; Sahoo, S. K. PLGA nanoparticles containing various
4T1 cells, effect of BPNP on apoptotic and antiapoptotic anticancer agents and tumour delivery by EPR effect. Adv. Drug Delivery
proteins via Western blotting, induction of mitochondrial Rev. 2011, 63 (3), 170−183.
membrane potential disturbance (qualitative and quanti- (10) Jokerst, J. V.; Lobovkina, T.; Zare, R. N.; Gambhir, S. S.
Nanoparticle PEGylation for imaging and therapy. Nanomedicine
tative) and tables detailing pharmacokinetic parameters (London, U. K.) 2011, 6 (4), 715−728.
(PDF) (11) Pawar, V. K.; Singh, Y.; Sharma, K.; Shrivastav, A.; Sharma, A.;

■ AUTHOR INFORMATION
Corresponding Author
Singh, A.; Meher, J. G.; Singh, P.; Raval, K.; Bora, H. K.; Datta, D.; Lal, J.;
Chourasia, M. K. Doxorubicin Hydrochloride Loaded Zymosan-
Polyethylenimine Biopolymeric Nanoparticles for Dual ‘Chemo-
immunotherapeutic’ Intervention in Breast Cancer. Pharm. Res. 2017,
*Telephone: +91 522-2772450, 2772550; Fax: +91 522 1−15.
2623405; E-mail: manish_chourasia@cdri.res.in. (12) Chen, S.; Zhao, X.; Chen, J.; Chen, J.; Kuznetsova, L.; Wong, S. S.;
ORCID Ojima, I. Mechanism-Based Tumor-Targeting Drug Delivery System.
Manish K. Chourasia: 0000-0002-8318-9325 Validation of Efficient Vitamin Receptor-Mediated Endocytosis and
Drug Release. Bioconjugate Chem. 2010, 21 (5), 979−987.
Present Address (13) Singh, Y.; Srinivas, A.; Gangwar, M.; Meher, J. G.; Misra-
#
Faculty of Pharmaceutical Sciences, University of British Bhattacharya, S.; Chourasia, M. K. Subcutaneously Administered
Columbia, Vancouver, British Columbia V6T 1Z3, Canada. Ultrafine PLGA Nanoparticles Containing Doxycycline Hydrochloride

2764 DOI: 10.1021/acs.molpharmaceut.7b00310


Mol. Pharmaceutics 2017, 14, 2749−2765
Molecular Pharmaceutics Article

Target Lymphatic Filarial Parasites. Mol. Pharmaceutics 2016, 13 (6), (29) (a) Mainardes, R. M.; Gremião, M. P. D.; Evangelista, R. C.
2084−2094. Thermoanalytical study of praziquantel-loaded PLGA nanoparticles.
(14) Singh, Y.; Ojha, P.; Srivastava, M.; Chourasia, M. K. Rev. Bras. Cienc. Farm. 2006, 42 (4), 523−530. (b) Singh, Y.; Singh, M.;
Reinvestigating nanoprecipitation via Box−Behnken design: a system- Meher, J. G.; Pawar, V. K.; Chourasia, M. K. Trichotomous gastric
atic approach. J. Microencapsulation 2015, 32 (1), 75−85. retention of amorphous capecitabine: An attempt to overcome
(15) Singh, Y.; Vuddanda, P. R.; Jain, A.; Parihar, S.; Chaturvedi, T. P.; pharmacokinetic gap. Int. J. Pharm. 2015, 478 (2), 811−821.
Singh, S. Mucoadhesive gel containing immunotherapeutic nano- (30) (a) Hines, D. J.; Kaplan, D. L. Poly (lactic-co-glycolic acid)
particulate satranidazole for treatment of periodontitis: development controlled release systems: experimental and modeling insights. Crit.
and its clinical implications. RSC Adv. 2015, 5 (59), 47659−47670. Rev. Ther. Drug Carrier Syst. 2013, 30 (3), 257−276. (b) Körber, M.
(16) Pawar, V. K.; Panchal, S. B.; Singh, Y.; Meher, J. G.; Sharma, K.; PLGA Erosion: Solubility- or Diffusion-Controlled? Pharm. Res. 2010,
Singh, P.; Bora, H. K.; Singh, A.; Datta, D.; Chourasia, M. K. 27 (11), 2414−2420.
Immunotherapeutic vitamin E nanoemulsion synergies the antiprolifer- (31) (a) Sahay, G.; Alakhova, D. Y.; Kabanov, A. V. Endocytosis of
ative activity of paclitaxel in breast cancer cells via modulating Th1 and Nanomedicines. J. Controlled Release 2010, 145 (3), 182−195.
Th2 immune response. J. Controlled Release 2014, 196, 295−306. (b) Singh, Y.; Chandrashekhar, A.; Meher, J. G.; Durga Rao
(17) Hein, C. D.; Liu, X.-M.; Wang, D. Click Chemistry, a Powerful Viswanadham, K. K.; Pawar, V. K.; Raval, K.; Sharma, K.; Singh, P. K.;
Tool for Pharmaceutical Sciences. Pharm. Res. 2008, 25 (10), 2216− Kumar, A.; Chourasia, M. K. Nanosized complexation assemblies
2230. housed inside reverse micelles churn out monocytic delivery cores for
(18) Miyazaki, S.; Oshiba, M.; Nadai, T., Precaution on Use of bendamustine hydrochloride. Eur. J. Pharm. Biopharm. 2017, 113, 198−
Hydrochloride Salts in Pharmaceutical Formulation. J. Pharm. Sci. 210.
198170 (6), 594−596.10.1002/jps.2600700604 (32) Nateghian, N.; Goodarzi, N.; Amini, M.; Atyabi, F.;
(19) Larsen, S. W.; Ostergaard, J.; Poulsen, S. V.; Schulz, B.; Larsen, C. Khorramizadeh, M. R.; Dinarvand, R. Biotin/Folate-decorated Human
Serum Albumin Nanoparticles of Docetaxel: Comparison of Chemically
Diflunisal salts of bupivacaine, lidocaine and morphine. Use of the
Conjugated Nanostructures and Physically Loaded Nanoparticles for
common ion effect for prolonging the release of bupivacaine from mixed
Targeting of Breast Cancer. Chem. Biol. Drug Des. 2016, 87 (1), 69−82.
salt suspensions in an in vitro dialysis model. Eur. J. Pharm. Sci. 2007, 31
(33) Järvinen, T. A. H.; Tanner, M.; Rantanen, V.; Bärlund, M.; Borg,
(3−4), 172−9. Å.; Grénman, S.; Isola, J., Amplification and Deletion of Topoisomerase
(20) Mendham, J. Vogels textbook of quantitative chemical analysis; IIα Associate with ErbB-2 Amplification and Affect Sensitivity to
Pearson Education: India, 2006. Topoisomerase II Inhibitor Doxorubicin in Breast Cancer. Am. J. Pathol.
(21) Hielscher, T. Ultrasonic production of nano-size dispersions and 2000156 (3), 839−847.10.1016/S0002-9440(10)64952-8
emulsions. OAI 2007, 1. (34) Cuddihy, A. R.; O’Connell, M. J. Cell-cycle responses to DNA
(22) Singh, Y.; Meher, J. G.; Raval, K.; Khan, F. A.; Chaurasia, M.; Jain, damage in G2. Int. Rev. Cytol. 2003, 222, 99−140.
N. K.; Chourasia, M. K. Nanoemulsion: Concepts, development and (35) Perry, J. L.; Reuter, K. G.; Kai, M. P.; Herlihy, K. P.; Jones, S. W.;
applications in drug delivery. J. Controlled Release 2017, 252, 28−49. Luft, J. C.; Napier, M.; Bear, J. E.; DeSimone, J. M. PEGylated PRINT
(23) Torcello-Gomez, A.; Maldonado-Valderrama, J.; Jodar-Reyes, A. Nanoparticles: The Impact of PEG Density on Protein Binding,
B.; Foster, T. J. Interactions between Pluronics (F127 and F68) and bile Macrophage Association, Biodistribution, and Pharmacokinetics. Nano
salts (NaTDC) in the aqueous phase and the interface of oil-in-water Lett. 2012, 12 (10), 5304−5310.
emulsions. Langmuir 2013, 29 (8), 2520−9. (36) Pawar, V. K.; Gupta, S.; Singh, Y.; Meher, J. G.; Sharma, K.; Singh,
(24) Kalra, A.; Garde, S.; Hummer, G. Osmotic water transport P.; Gupta, A.; Bora, H. K.; Chaurasia, M.; Chourasia, M. K. Pluronic F-
through carbon nanotube membranes. Proc. Natl. Acad. Sci. U. S. A. 127 Stabilised Docetaxel Nanocrystals Improve Apoptosis by
2003, 100 (18), 10175−10180. Mitochondrial Depolarization in Breast Cancer Cells: Pharmacokinetics
(25) (a) Singh, Y.; Pawar, V. K.; Meher, J. G.; Raval, K.; Kumar, A.; and Toxicity Assessment. J. Biomed. Nanotechnol. 2015, 11 (10), 1747−
Shrivastava, R.; Bhadauria, S.; Chourasia, M. K. Targeting tumor 63.
associated macrophages (TAMs) via nanocarriers. J. Controlled Release (37) Moldovan, G.-L.; Pfander, B.; Jentsch, S. PCNA, the Maestro of
2017, 254, 92−106. (b) Singh, Y.; Tomar, S.; Khan, S.; Meher, J. G.; the Replication Fork. Cell 2007, 129 (4), 665−679.
Pawar, V. K.; Raval, K.; Sharma, K.; Singh, P. K.; Chaurasia, M.; Surendar (38) Bae, S. I.; Zhao, R.; Snapka, R. M. PCNA Damage Caused by Anti-
Reddy, B.; Chourasia, M. K. Bridging small interfering RNA with giant Neoplastic Drugs. Biochem. Pharmacol. 2008, 76 (12), 1653−1668.
therapeutic outcomes using nanometric liposomes. J. Controlled Release (39) (a) D’Alessandro, N.; Candiloro, V.; Crescimanno, M.; Flandina,
2015, 220, 368−387. (c) Pawar, V. K.; Singh, Y.; Meher, J. G.; Gupta, S.; C.; Dusonchet, L.; Crosta, L.; Rausa, L. Effects of multiple doxorubicin
Chourasia, M. K. Engineered nanocrystal technology: In-vivo fate, doses on mouse cardiac and hepatic catalase. Pharmacol. Res. Commun.
targeting and applications in drug delivery. J. Controlled Release 2014, 1984, 16 (2), 145−51. (b) Doroshow, J. H.; Locker, G. Y.; Myers, C. E.
183, 51−66. (d) Petros, R. A.; DeSimone, J. M. Strategies in the design Enzymatic defenses of the mouse heart against reactive oxygen
of nanoparticles for therapeutic applications. Nat. Rev. Drug Discovery metabolites: alterations produced by doxorubicin. J. Clin. Invest. 1980,
2010, 9 (8), 615−627. 65 (1), 128−35.
(26) Fonseca, C.; Simoes, S.; Gaspar, R. Paclitaxel-loaded PLGA
nanoparticles: preparation, physicochemical characterization and in
vitro anti-tumoral activity. J. Controlled Release 2002, 83 (2), 273−286.
(27) Doxorubicin. In Drug Bank, 17/8/2016 ed.; Canada, 2016.
(28) (a) Mainardes, R. M.; Gremião, M. P. D.; Evangelista, R. C.
Thermoanalytical study of praziquantel-loaded PLGA nanoparticles.
Rev. Bras. Cienc. Farm. 2006, 42, 523−530. (b) Lee, T.-M.; Uang, R.-H.;
Chiou, K.-C.; Wang, Y.-M.; Cheng, Y.-T., Method for manufacturing a
substrate with surface structure by employing photothermal effect.
Google Patents: 2012. (c) Danafar, H.; Davaran, S.; Rostamizadeh, K.;
Valizadeh, H.; Hamidi, M. Biodegradable m-PEG/PCL Core-Shell
Micelles: Preparation and Characterization as a Sustained Release
Formulation for Curcumin. Advanced Pharmaceutical Bulletin 2014, 4
(2), 501−510. (d) Antoniammal, P.; Arivuoli, D. Size and Shape
Dependence on Melting Temperature of Gallium Nitride Nano-
particles. J. Nanomater. 2012, 2012, 11.

2765 DOI: 10.1021/acs.molpharmaceut.7b00310


Mol. Pharmaceutics 2017, 14, 2749−2765

S-ar putea să vă placă și