You are on page 1of 18

Biomedicine & Pharmacotherapy 144 (2021) 112286

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


journal homepage: www.elsevier.com/locate/biopha

Development and optimization of paclitaxel loaded Eudragit/PLGA


nanoparticles by simplex lattice mixture design: Exploration of improved
hemocompatibility and in vivo kinetics
Gunjan Jeswani a, b, Lipika Chablani c, **, Umesh Gupta d, Rakesh K. Sahoo d, Kartik T. Nakhate e,
Ajazuddin f, *
a
Rungta College of Pharmaceutical Sciences and Research, Kohka-Kurud Road, Bhilai, Chhattisgarh 490024, India
b
Faculty of Pharmaceutical Sciences, Shri Shankaracharya Technical Campus, Bhilai, Chhattisgarh 490020, India
c
Department of Pharmaceutical Sciences, Wegmans School of Pharmacy, St. John Fisher College, Rochester, NY 14618, USA
d
Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan 305817, India
e
Department of Pharmacology, Shri Vile Parle Kelavani Mandal’s Institute of Pharmacy, Dhule, Maharashtra 424001, India
f
School of Pharmacy and Technology Management, SVKM’s NMIMS, Shirpur, Maharashtra 425405, India

A R T I C L E I N F O A B S T R A C T

Keywords: Anemia is the most common hematological abnormality of chemotherapy, which is responsible for poor clinical
Paclitaxel outcomes. To overcome this complication, the present study was aimed for developing a Eudragit/polylactic-co-
Nanoparticle(s) glycolic acid (PLGA) based nanoparticulate system for a model drug paclitaxel (PTX). The study was planned
Eudragit RLPO
using a simplex lattice mixture design. PTX nanoparticles (PTXNp) were evaluated in vitro for physicochemical
Eudragit RSPO
PLGA
properties, hemolytic effects and cytotoxic effects. Further, the nanoparticles were subjected to in vivo screening
Design of experiments (DOE) using rats for hemocompatibility, pharmacokinetic profile, and biodistribution to the vital organs. The PTXNps
Simplex lattice were 65.77–214.73 nm in size, showed more than 60% sustained drug release in 360 h and caused less than 8%
Hemocompatibility hemolysis. The parameters like red blood cell count, activated partial thromboplastin time (aPTT), prothrombin
time (PT) and C3 complement were similar to the negative control. Cytotoxicity results suggested that all the
PTXNp demonstrated drug concentration-dependent cytotoxicity. The in vivo pharmacokinetic study concluded
that PTXNp formulations had significantly higher blood AUC (93.194.55–163,071.15 h*ng/mL), longer half-lives
(5.80–6.35 h) and extended mean residence times (6.05–8.54 h) in comparison to PTX solution (p < 0.05).
Overall, the study provides a nanoparticulate drug delivery system to deliver PTX safely and effectively along
with reducing the associated hematological adverse effects.

1. Introduction 6.8 million deaths (6.9% of all deaths due to cancer) were reported due
to breast cancer in 2020. Further, it is projected that the number of cases
Breast cancer is one of the most common cancers in women. Ac­ will rise from 2.26 to 2.96 million by 2040 [1].
cording to the Global Cancer Observatory and the International Agency Currently, chemotherapy is the most utilized treatment for breast
for Research on Cancer (IARC), about 2.26 million new breast cancer cancer. However, it leads to several clinical adverse effects. These
cases (11.7% of all new cancer cases) were registered in 2020, and about adverse effects can range from but are not limited to hair loss, fatigue,

Abbreviations: AUC, area under curve in ng hr/mL; C0, initial concentration; Cl, Clearance; Clast, concentration corresponding to time of last measurable observed
concentration; Cmax, maximum observed concentration occurring at time Tmax; DLS, dynamic light scattering; EPR, enhanced permeation and retention; FDA, Food
and Drug Administration; Hb, Hemoglobin; HCT, haematocrit percentage; IARC, International Agency for Research on Cancer; MCH, mean corpuscular Hb, and
MCHC, mean corpuscular Hb concentration; MCV, mean corpuscular volume; PBS, phosphate buffered saline; PLGA, polylactic-co-glycolic acid; PLT, platelet count;
PT, prothrombin time; PTT, activated partial thromboplastin time; PTX, paclitaxel; PTXNp, PTX nanoparticle; RBC, red blood cells; T1/2, terminal half life; Vd,
volume of distribution based on the terminal phase; WBC, white blood cells.
* Corresponding author.
** Correspondence to: Department of Pharmaceutical Sciences, St. John Fisher College, 3690 East Ave, Rochester, NY 14618, USA.
E-mail addresses: lchablani@sjfc.edu (L. Chablani), ajazuddin@nmims.edu (Ajazuddin).

https://doi.org/10.1016/j.biopha.2021.112286
Received 31 August 2021; Received in revised form 24 September 2021; Accepted 5 October 2021
Available online 13 October 2021
0753-3322/© 2021 The Authors. Published by Elsevier Masson SAS. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
G. Jeswani et al. Biomedicine & Pharmacotherapy 144 (2021) 112286

vomiting, nausea, and clinically remarkable anemia [2]. Hemolytic evaluate the hemocompatibility, pharmacokinetic profile, and bio­
anemia predisposes patients to cardiac and neurological diseases. distribution of the drug from the optimized formulation.
Approximately 30–90% of patients receiving chemotherapeutics expe­
rience hemolytic anemia. The nadir is reached between 15 and 20 days. 2. Materials
It is a major challenge and is responsible for higher morbidity rates; such
adverse effects often lead to chemotherapeutic dose restrictions [3]. Paclitaxel (Benzenepropanoic acid, purity ≥103.3%) was received as
Chemotherapy also can cause antineoplastic multiple drug resistance, a generous gift from Intas Pharmaceuticals Ltd. (Batch No.
which is a common threat in relapsed tumors [4]. Due to these limita­ 684802806004; Exp. Dt. 06/2022). Eudragit RSPO (ERSPO) and
tions, chemotherapy needs to be re-evaluated to overcome these chal­ Eudragit RLPO (ERLPO) and Poly D, L Lactide-co-Glycolide 50:50
lenges to provide the intended therapeutic outcomes. (PLGA, Resomer® RG 502H, CAS # 26780–50–70) were obtained as a
The U.S. Food and Drug Administration (FDA) authorized paclitaxel gift from Evonik India Pvt. Ltd. Phosphate Buffered Saline, pH 7.4 1X
(PTX) as a first/second-line intervention for various metastatic malig­ (CAT # TL1101–500 mL), Fetal bovine serum (CAT #RM1112–500 mL),
nancies, including breast, ovarian, and non-small cell lung cancer [5]. Streptomycin-penicillin solution (CAT #A007–100 mL) and Dulbecco’s
Taxol® is the majorly prescribed formulation of PTX and has been Modified Eagle Medium, DMEM (CAT #AL007A-500 mL) were bought
effective for several patients with metastatic cancer. In 2005, FDA from HiMedia, Mumbai, India. Dimethyl sulfoxide (CAT #515505) was
approved an albumin-bound PTX nanoparticle, Abraxane®, showing procured from CDH, New Delhi, India. MCF-7 cell line was procured
clinical superiority over PTX alone [6]. Currently, 106 interventional from National Centre for Cell Science, Pune, India. Polyvinyl alcohol,
clinical studies are ongoing based on PTX therapies for breast cancer. acetone, and deionized water (HPLC grade) were bought from Sigma
Recently, nanotechnology has enabled targeted delivery of chemo­ Aldrich (Mumbai, India). Uniplastin and Liquicellin was the product of
therapeutics because of the enhanced permeation and retention (EPR) Tulip Diagnostics (P) Ltd, Goa, India. All other chemicals were of
effect observed in solid tumors. Such applications also limit drug-related analytical grade.
toxicity to healthy cells. Nanoparticles are among the most extensively
researched nanotechnology-based drug delivery systems [7]. The 3. Method
composition, particle size, and drug release profile of nanoparticles
regulate the efficacy of these drug delivery systems. Often generally 3.1. Experimental design and preparation of nanoparticle
regarded as safe (GRAS), polymers are chosen for the composition of
such nanoparticles. Resomer® RG 502 H, polylactic-co-glycolic acid 3.1.1. Experimental design
(PLGA) is a biodegradable, safe polymer with lactic acid and glycolic A three-factor, simplex lattice mixture design was constructed to
acid as the base monomers. It is completely metabolized via the Krebs evaluate the optimum amounts of individual polymers required to yield
cycle. However, the main drawbacks of the polymer include low circu­ a formulation with the lowest hemolysis (less than 10%), smallest par­
lation time, poor cell permeation, and rapid in vivo clearance [8]. The ticle size (less than 220 nm) and the desired drug release (more than
combination of PLGA with other ingredients like polyvinyl alcohol, 70%). Statistical software, JMP® version 15.2.1 (SAS Institute, Cary,
dextran, d-α-tocopheryl PEG 1000 succinate, chitosan, or hyaluronic NC), was used to create and evaluate the experimental design. PLGA,
acid, reduces the drawbacks mentioned above. ERSPO, and ERLPO concentrations were included as continuous factors
Eudragit RSPO (ERSPO) and Eudragit RLPO (ERLPO) are referred as to develop a mixture design (Table 1).
copolymers of ethyl acrylate and methyl acrylate.The average molecular The design aimed to determine the impact of the concentration of
weight of ERSPO/ERLPO is 32,000 g/mol. Physically, both appear as polymers on the formulation quality. The quality characteristics of the
white, fine powders and exhibit typical amine like smell. ERLPO con­ formulation included particle size, percent hemolysis, and drug release.
tains 10% of functional quaternary ammonium groups whereas ERSPO Polymer concentrations of all three polymers (PLGA, ERSPO, and
contains 5% functional quaternary ammonium groups. They have poor ERLPO) were varied in the range of 0–100% or 0–1 (as coded) (Table 1).
aqueous solubility but dissolve freely in acetone and alcohols. Because of The concentration of PTX was held at 20 mg, such that the drug-polymer
the difference in ratio of quaternary ammonium groups, the water ratio remained constant at 1:4.
permeability of ERLPO matrix is higher than ERSPO. These properties
make them an ideal candidate for preparing sustained release dosage 3.1.2. Preparation of nanoparticle
forms [9]. They have been used alone or in combination for preparing The nanoprecipitation method was used to formulate polymeric
customized matrix structures for drug delivery. However, they have not nanoparticles [15]. Briefly, PTX (20 mg) was dissolved in 4 mL ethanol.
been evaluated with PLGA to deliver chemotherapeutic drugs to the The solution was stirred on a magnetic stirrer (Remi, India) for 5 min at
specific tumor site so far. 100 rpm in a covered beaker to get a 5 mg/mL solution. For individual
A combination of different grades of Eudragit and PLGA has been formulations, a varying amount of PLGA/ERSPO/ERLPO was taken as
used previously for the formulation of the nanoparticles of heparin [10], mentioned in Table 1. The organic phase constituted the mixture of the
ciprofloxacin [11], diclofenac sodium [12], enteric coating of eluxado­ required amount of PLGA in 2 mL acetone. ERSPO, ERLPO, or both were
line PLGA nanoparticles for sustained release [13], and also for intra­ dissolved in 2 mL ethanol and the drug solution was also prepared in
muscular delivery of interleukin-10 [14]. The biocompatibility of PLGA ethanol separately. Both polymer solutions were mixed with constant
and Eudragit E nanoparticles has been previously established [14]. stirring at 100 rpm on a magnetic stirrer for 2 min. Next, 4 mL of drug
Considering the efficacy and broad application of PTX as a chemo­ solution was combined with the prepared polymer solution to constitute
therapeutic agent, this study aims to formulate an optimized nano­ the organic phase. The organic phase was subsequently added to 40 mL
particulate drug delivery system that limits the hematotoxicity of aqueous phase containing phosphate buffer saline (PBS), pH 7.4% and
associated with PTX. We hypothesized that PTX nanoparticles devel­ 1% v/v PVA. The addition was performed at a constant rate of 0.5
oped with a blend of polymers would improve hemocompatibility and mL/min using BD Veo™ insulin syringes with BD Ultra-Fine™ 31 G
enhance antitumor activity. The work utilizes a statistical design of needle. The addition continued with uninterrupted stirring at 1000 rpm
experiment to formulate and optimize PTX nanoparticle (abbreviated as on a magnetic stirrer at room temperature, 25 ◦ C. Lastly, the stirring was
PTXNp hereafter) formulation using three polymers (PLGA, Eudragit done at 200 rpm for 12 h in a rotary shaker maintained at 30 ◦ C to
RSPO, and Eudragit RLPO). The optimized formulations were then evaporate the organic phase. The resulting PTXNp suspension was
evaluated in vitro to determine physicochemical properties such as the centrifuged at 12,298 xg at 4 ◦ C for 15 min using Eppendorf Centrifuge
size, zeta potential, morphology, drug release profile, and hemolysis. 5424R (Eppendorf AG, Hamburg, Germany) to get a pellet and further
Further, the in vivo studies were performed in a Wistar rat model to washed twice with 5 mL of deionized water.

2
G. Jeswani et al. Biomedicine & Pharmacotherapy 144 (2021) 112286

Table 1
Simplex lattice mixture design of nanoparticle formulation showing different proportions of polymers used in 21 runs and resulting responses. ERSPO, ERLPO, PLGA
are used in proportion, [0,1]. Responses are represented as mean ± SD (n = 3).
Formulation Code Composition Hemolysis (%) Particle size (nm) Cumulative Release in PBS pH 7.4 (%)

ERSPO (mg) ERLPO (mg) PLGA (mg)

PTXNp1 0 [0] 32 [1] 0 [0] 23.23 ± 1.00 65.77 ± 1.93 82.02 ± 1.74
PTXNp2 6.4 [0.2] 25.6 [0.8] 0 [0] 19.76 ± 2.00 67.74 ± 1.85 80.23 ± 3.01
PTXNp3 6.4 [0.2] 0 [0] 25.6 [0.8] 07.43 ± 1.00 210.73 ± 1.85 66.92 ± 3.01
PTXNp4 0 [0] 19.2 [0.6] 12.8 [0.4] 15.89 ± 2.91 200.92 ± 2.64 72.66 ± 1.47
PTXNp5 12.8 [0.4] 0 [0] 19.2 [0.6] 09.00 ± 1.86 203.67 ± 2.17 68.34 ± 1.72
PTXNp6 12.8 [0.4] 6.4 [0.2] 12.8 [0.4] 11.33 ± 2.65 198.42 ± 3.79 65.58 ± 2.48
PTXNp7 12.8 [0.4] 19.2 [0.6] 0 [0] 16.89 ± 2.86 71.90 ± 1.36 78.28 ± 1.49
PTXNp8 25.6 [0.8] 6.4 [0.2] 0 [0] 18.98 ± 0.92 92.77 ± 0.89 72.65 ± 1.18
PTXNp9 0 [0] 6.4 [0.2] 25.6 [0.8] 07.89 ± 0.98 209.43 ± 1.87 67.32 ± 0.71
PTXNp10 0 [0] 25.6 [0.8] 6.4 [0.2] 21.58 ± 2.36 74.77 ± 1.55 78.51 ± 2.21
PTXNp11 6.4 [0.2] 6.4 [0.2] 19.2 [0.6] 10.21 ± 2.46 206.22 ± 1.22 66.05 ± 1.97
PTXNp12 6.4 [0.2] 12.8 [0.4] 12.8 [0.4] 13.58 ± 1.73 198.27 ± 1.24 65.70 ± 1.96
PTXNp13 19.2 [0.6] 0 [0] 12.8 [0.4] 12.98 ± 1.32 203.13 ± 1.91 69.20 ± 3.38
PTXNp14 0 [0] 12.8 [0.4] 19.2 [0.6] 12.07 ± 1.25 198.36 ± 1.24 69.80 ± 0.72
PTXNp15 0 [0] 0 [0] 32 [1] 02.01 ± 1.27 214.73 ± 4.48 65.00 ± 2.13
PTXNp16 32 [1] 0 [0] 0 [0] 21.00 ± 1.23 111.83 ± 1.77 70.02 ± 1.76
PTXNp17 19.2 [0.6] 12.8 [0.4] 0 [0] 20.89 ± 3.82 88.77 ± 1.68 74.30 ± 1.78
PTXNp18 6.4 [0.2] 19.2 [0.6] 6.4 [0.2] 14.23 ± 3.41 197.27 ± 1.98 78.64 ± 1.35
PTXNp19 19.2 [0.6] 6.4 [0.2] 6.4 [0.2] 13.67 ± 2.00 195.37 ± 1.53 68.51 ± 2.10
PTXNp20 25.6 [0.8] 0 [0] 6.4 [0.2] 18.25 ± 1.91 116.82 ± 3.24 69.84 ± 1.35
PTXNp21 12.8 [0.4] 12.8 [0.4] 6.4 [0.2] 17.36 ± 1.04 125.97 ± 2.58 70.27 ± 2.05
PTX solution in ethanol (200 µg /mL) – – – 100 – –

3.1.3. Freeze-dried nanoparticles India.


The nanoparticle pellet was reconstituted with PBS of pH 7.4 (10 mL)
and 5% w/v mannitol (cryo-preservative). The suspension was main­ 3.2.2.1. Hemolysis study. The whole blood was centrifuged (10 min,
tained at − 40 ◦ C for two days in a freezer. Subsequently, PTXNp sus­ 867 xg, Eppendorf Centrifuge 5424 R) to obtain a RBC pellet for the
pension was transferred to Heto PowerDry® LL 3000 freeze dryer hemolysis experiment [17]. The RBC pellets were washed twice with
(Thermo Fisher Scientific, United States) pre-chilled to − 40 ◦ C. Lyoph­ 0.9% saline. A 3% v/v RBC stock suspension in PBS (pH 7.4) was pre­
ilization was performed for five days under vacuum (− 20 ◦ C, 100 pared using 3 mL of packed cell volume. To prepare the sample, 1 mL of
mTorr). At the end of the process, free-flowing freeze-dried PTXNps PTXNp suspension in PBS (containing 200 micrograms PTX) was mixed
were collected in a 10 mL glass vial and were dessicated at ambient with 1 mL of 3% v/v RBC suspension. The mixture was incubated for 4 h
temperature. Freeze-dried samples were used for all further evaluations. at 37 ◦ C [16]. Distilled water (1 mL) was used for 100% hemolysis
Collocating with our aim, percent release, hemolysis, and particle size (positive control), and 1 mL of PBS was used for 0% hemolysis (negative
were used as response variables for the study. control) [18]. After full incubation time, the suspension was centrifuged
for 10 min (867 xg using Eppendorf Centrifuge 5424 R). The supernatant
3.2. In vitro characterization was examine using UV spectrophotometer (Double beam UV Vis Spec­
trophotometer LT 2800, Labtronics, India) for released hemoglobin (Hb)
3.2.1. Particle size at 540 nm [18]. The hemolysis percent was calculated as per Eq. (1).
The mean particle diameter and size distribution of PTXNp suspen­

Absorbance ofblood incubated with PTXNp − Absorbance of negative control


Hemolysis %= ∗ 100 (1)
Absorbanceof positive control − Absorbanceofnegative control

sion was determined by DLS technique on a Nano ZS (Zeta Sizer, Mal­


vern, UK) at 25 ◦ C with an equilibration time of 60 s and 90◦ angle of
detection using DTS0012 transparent disposable cuvette. One milligram 3.2.3. In vitro PTX release profile
of lyophilized PTXNp formulations were dispersed in 10 mL HPLC grade The dialysis bag diffusion technique was used for the in vitro release
water and sonicated for 1 min before measurements. Measurements of studies, as described by Dong et al. [19]. Lyophilized PTXNp (100 mg)
each sample were done in triplicates and reported with standard were incubated with 2 mL of PBS (pH 7.4, receptor fluid to mimic
deviations. physiological blood-plasma pH) in a dialysis membrane bag (MW cut off
14000 D). The dialysis bag was secured and placed into a beaker con­
3.2.2. Hemocompatibility studies taining 40 mL of PBS with 0.1% Tween 80, maintained at 37 ◦ C in a
Human blood (10 mL/volunteer) from healthy volunteers (18–50 shaking water bath. At regular time points (2, 4, 6, 8, 10, 28,50, 74, 168,
years) was collected after a written consent. The samples were pooled 360 h) until 15 days, 2 mL of receptor fluid was withdrawn and diluted
and stored in EDTA anti-coagulant blood storage vials at − 6 ± 2◦ C until to 10 mL with HPLC mobile phase to determine PTX concentration via
used for all the experiments of hemocompatibility [16]. The study HPLC analysis (details provided in the supplementary file, Table S1). In
protocol was permitted by the Institutional Research Committee of vitro release data of all PTXNp suspensions were fitted to relevant kinetic
Rungta College of Pharmaceutical Science and Research, Bhilai, CG, models, and the best fit model was chosen based on the highest

3
G. Jeswani et al. Biomedicine & Pharmacotherapy 144 (2021) 112286

correlation coefficient. incubated for a specific time as indicated in the individual tests with
1 mg/mL of all the optimized and placebo nanoparticles for different
3.3. In vitro analyses of optimized formulations hemocompatibility tests in individual EDTA (hemolysis test) or sodium
citrate tubes (coagulation test). The treated sample was used for further
After evaluating the PTXNp for particle size, hemolysis, and in vitro analyses. A 1 mg/mL concentration of the particle was used to achieve
release, additional in vitro studies were carried out on selected optimized 0.77 ± 1.14 mg/mL PTX concentration in vitro. The concentration is
formulations only. These formulations included PTXNp3, PTXNp9, and much higher than the concentration used in animal models
PTXNp15. (0.06–0.12 mg/mL). Blood incompatibility issues are reported to be
concentration-dependent; hence we selected much higher theoretical
3.3.1. Surface morphology concentration than used for in vivo studies to predict the safety of the
Electron transmission microscopy (TEM) (Tecnai, G 20, FEI), with formulation at a higher dose [21].
image processing software iTEM, Olympus Soft Imaging System, Ger­
many, was used to study the surface morphology of PTXNps. Before grid 3.3.5.1. Whole blood clotting time. The whole blood clotting time assay
preparation, 1 mL of nanoparticle suspension was diluted with 5 mL of was executed as per the Lee White method (17). After 30 s of incubation
saline solution and sonicated for 5 min 20 µL of the sample was posi­ at 37 ◦ C, the tubes were tilted to 30º to check the blood clotting. After
tioned onto the non-copper grid and negatively stained with a 0.01% w/ every 30 s, the tubes were tilted further until they stopped flowing at
v phosphotungstic acid solution for 50 s [20]. The grid was allowed to 90º, and at that instance, the clotting time was recorded. Blood from
air dry for 5 min and then imaged under the microscope supported with blank EDTA test tubes (without formulation) served as the negative
a CCD camera. TEM analysis was performed at All India Institute of control, and non-EDTA glass tubes were a positive control. All mea­
Medical Sciences (AIIMS), New Delhi, India. Furthermore, for surface surements were performed in triplicates.
topography, the PTXNps were mounted onto a stub using conductive
carbon adhesive tape and then sputter-coated with gold. The samples 3.3.5.2. Blood counts. To determine the blood counts, 1 mL of blood
were imaged using a ZEISS EVO series Scanning Electron Microscope was incubated with 1 mg of formulation (placebo and PTXNp) for
(SEM; Model EVO 18). 30 min at 37 ◦ C. A double capillary blood cell counter (Model Sysmex
XP-300, Automated hematology analyzer) was used to determine com­
3.3.2. Zeta potential plete blood count, including RBC and WBC count, total Hb concentra­
Electrophoretic light scattering technique was used for determining tion, hematocrit (HCT) percentage, platelet (PLT) count, and differential
electrophoretic mobility (zeta potential) using Nano ZS Zetasizer (Zeta counts (lymphocytes, monocytes, and granulocytes). We also deter­
Sizer, Malvern, UK) at 25 ◦ C. The test was carried using a zeta cell, mined the blood indexes [mean corpuscular volume (MCV), mean
DTS1060C, with 30 s equilibration time. One milligram of PTXNp for­ corpuscular Hb (MCH), and mean corpuscular Hb concentration
mulations were dispersed in 10 mL of HPLC grade water and sonicated (MCHC)] [16].
for 1 min before measurements.
3.3.5.3. Activated partial thromboplastin time (aPTT) and prothrombin
3.3.3. FTIR and XRD time (PT) for coagulation. A 1 mL of sodium citrate blood sample was
The potassium bromide disc method using infrared (IR) spectroscopy mixed with 1 mg of the formulation (placebo and PTXNp) to prepare the
(Shimadzu IR Affinity 1 DRS8000) over wavelength range sample for aPTT determination. Then, 50 µL of the sample was mixed
4000–400 cm− 1 was used to determine any drug-polymer interaction. with 50 µL of aPTT reagent (LiquicellinE, Tulip Diagnostics) and incu­
For X-ray diffraction (XRD) studies approximately, 2 mg of the drug, bated with a steel ball for 20 min at 37 ◦ C. After the incubation, 50 µL
polymers, and PTXNp were analyzed by high-resolution X-ray diffrac­ calcium chloride was added, and results were recorded with Stago Art4
tometer (PANalytical 3 kW X′ pert Powder – Multifunctional) in a range Coagulomete Diagnostica, France. Similarly, for PT measurements,
of 5–80◦ (2θ), using Cu-Kα radiation (λ = 1.544 Å) generated at 40 kV 50 µL of plasma sample with steel ball were incubated with Calcium
and 30 mA, at a scan step size of 0.026◦ . Thromboplastin Reagent (Uniplastin, Tulip Diagnostics), and values
were recorded [22]. Measurements were performed in triplicates.
3.3.4. Loading and entrapment efficiency
Briefly, 5 mg of each lyophilized PTXNp formulation was suspended 3.3.5.4. C3 complement. Immunoturbidimetry method using the Com­
in HPLC grade water (10 mL) and stirred for 15 min on a magnetic plement protein C3 kit (Biobase, China) was used to determine com­
stirrer (Remi, India) at 100 rpm and then sonicated for 5 min (400 W). plement activation by optimized PTXNps and respective placebo
The prepared suspensions were centrifuged (Eppendorf Centrifuge nanoparticles [23]. The assay was conducted as per the manufacturer’s
5424R) twice at 1491 xg for 10 min to determine the amount of unin­ protocol. Briefly, 1 mL of human serum sample was mixed with 1 mg of
corporated drug in the supernatant using UV Spectroscopy (Shimadzu formulation (placebo and PTXNp) and incubated for 1 h at 37 ◦ C.
UV-1800, Labomed USA). The concentration of the PTX was estimated Absorbance was measured thereafter. All measurements were performed
using the calibration curve. Entrapment efficiency and drug loading in triplicates [24].
were calculated using the Eqs. (2) and (3) resp.
Total PTX added- PTX in the supernatant
Entrapment Efficiency(%) = 3.4. In vitro cytotoxicity assay
Total PTX added
∗ 100
Cytotoxicity assay of PTX solution, PTXNp3, PTXNp9, and PTXNp15
(2) was performed against MCF-7 breast cancer cells using the referenced
MTT assay protocol [25]. Cytotoxic effects of placebo particles corre­
Mass of PTX in the nanoparticle
Drug loading(%) = ∗ 100 (3) sponding to PTXNp3, PTXNp9, and PTXNp15were also determined.
The total mass of PTXNp
MCF-7 cells were grown in a T-75 flask using complete Dulbecco’s
All the determinations were performed in triplicates, and results are modified eagle medium (DMEM) containing fetal bovine serum (10%
reported as mean ± standard deviation. v/v) and streptomycin-penicillin (1% v/v). Confluent cells were coun­
ted, and 10,000 cells were seeded into each well of a 96-well plate fol­
3.3.5. Blood compatibility lowed by incubation in a CO2 incubator (Thermo-Fisher Heracell150i).
Pooled human blood (1 mL) as described in Section 3.2.2 was Serial dilutions of PTX (1, 2.5, 5, 7.5, 10, 20, 35, and 50 µg/mL) were

4
G. Jeswani et al. Biomedicine & Pharmacotherapy 144 (2021) 112286

formulated in complete DMEM media. Similarly, corresponding to the ± standard deviation (SD) of the three replicates. Sample means were
PTX concentrations based on the entrapment efficiency, serial dilutions compared by t-test, one-way or two-way ANOVA, whichever applicable,
of PTX loaded and placebo nanoparticles were prepared in the same followed by a Tukey post hoc test. A p-value of < 0.05 was considered
media (70, 350, 525, 700, 1400, 2100, 3500, 4900 µg/mL). Further, the significant.
cancer cells were treated with the known concentrations of PTXNp or
PTX to be investigated and incubated for 24 h. Complete DMEM media 5. Result and discussion
served as the negative control. After the incubation period, each well
was washed with pH 7.4 PBS and 50 µL of MTT solution was added. Recently, advancements in drug delivery techniques have allowed
Subsequently, the cells were kept for 4 h of incubation. To dissolve the clinicians to deliver chemotherapeutic agents to cancer patients with
formazan crystals produced due to viable cells, 150 µL dimethyl sulf­ potentially minimum adverse effects. Our research focuses in the same
oxide was added into each well [26]. The absorbance was analyzed (λmax direction as we utilize a statistical design of experiment to formulate a
= 570 nm) using a microplate reader (Omega Fluster, BMG Labtech) and PTXNp with the desired physicochemical properties. A mixture design
the IC50 values of PTX and nanoparticles were calculated. was prepared with varying Eudragit RSPO/RLPO and PLGA concentra­
tions to study the response variables, including the particle size, percent
3.5. In vivo pharmacokinetic studies hemolysis, and drug release. The overall goal of the study is to benefit
from a small particle size formulation to achieve desirable EPR effect,
Pharmacokinetic parameters were determined in male Wistar rats minimize hemolysis, and obtain efficient drug release.
(~250 g) aged 10–12 weeks. The study protocol was approved by the
Institutional Animal Ethics Committee, Rungta College of Pharmaceu­ 5.1. Mixture design and preparation of nanoparticles
tical Sciences, Bhilai, CG, India (1189/PO/Re/s/08/CPCSEA/2018/6).
Animals were housed in a temperature and relative humidity controlled PTX is a Biopharmaceutical Classification System class IV drug. It has
area (25ºC and 50% RH) for 12 h light/dark cycle. The rats were an aqueous solubility of 0.25 µg/mL (log D of ~4.95), and the log P
randomly divided into five groups, P1, P2, P3, P4, and P5 (n = 3/group). value is 3.66 [29]. Moreover, the major clinical challenge of PTX is
Group P1, P2, and P3 received PTX nanoparticle formulations, PTXNp3, drug-induced hemolytic toxicity. Low solubility, low permeability, and
PTXNp9, and PTXNp15, respectively. The formulations were adminis­ drug-induced toxicity are the critical limitations of PTX. Nanoparticles
tered via the tail vein as a single dose of 5 mg/kg body weight [27]. increase the solubility of poorly soluble drugs, enhance permeability,
Group P4 received pure PTX solution in ethanol (5 mg/mL), and P5 and help in achieving targeted drug release. Nanoprecipitation method
served as the control. Post-treatment, 0.2 mL of blood samples were is the most convenient and feasible formulation technique to prepare
collected at the predetermined intervals (0.5, 1, 2, 4, 8, 12, and 24 h) in nanoparticles that can help to overcome the above-discussed challenges
heparinized capillaries from retro-orbital plexus. The collected samples [30]. It was hypothesized that composition variables of nanoparticles
were immediately centrifuged at − 6 ◦ C and 31,304 xg for 5 min significantly affect the major formulation characteristics of PTX (particle
(Eppendorf Centrifuge 5424 R). The plasma was collected in a labeled size, drug release, and hemolysis). Hence, modulation of composition
microcentrifuge tube and stored at − 20 ºC until further use. Before the variables can enhance drug delivery, sustain drug release, and minimize
analysis, 200 µL of plasma was mixed with 1 mL acetonitrile to extract hematotoxicity.
PTX, [28]. The extracted PTX solutions were centrifuged, the superna­ To validate the hypothesis, JMP® version 15.2.1 (SAS Institute, Cary,
tant was collected and filtered using a 0.25 µm membrane filter, fol­ NC) statistical software was used to generate a mixture design-based
lowed by analysis through HPLC. Pharmacokinetic parameters, terminal design of experiment. It generated experimental runs to predict a set
half-life (T1/2), maximum plasma concentration (Cmax), area under of formulations that will be least hemolytic and have the desired particle
curve (AUC), volume of distribution based on the terminal phase (Vd), size along with the adequate sustained release. Further, the optimum
clearance (Cl), and mean residence time (MRT) were calculated using combination of Eudragit RSPO/RLPO and PLGA was carefully chosen
Phoenix 64 WinNonlin® version 8.3.3.33 (Pharsight Corporation, with the help of a simplex lattice mixture design, wherein the pro­
MountainView, CA, USA). portions of constituents were varied from none to whole without the
application of any constraints. In the present investigation, PTX loaded
3.6. Biodistribution and hematological studies PLGA, ERSPO, ERLPO, or both nanoparticles were prepared using
nanoprecipitation technique using ethanol and acetone as the organic
Post-treatment, at 24 h, rats were euthanized. Blood was collected by phase. Although all the polymers are readily soluble in acetone, ethanol
cardiac puncture, and samples were analyzed for a complete blood was used for better tolerability [31]. By varying the proportion of
count. Further, the liver, spleen, lungs, kidneys, and heart were isolated. polymers from 0 to 1 at five different levels (0, 0.2, 0.4, 0.6, 0.8, and 1),
HPLC grade acetonitrile (10 mL) was used to precipitate out the plasma the study attempts to balance the toxicity and the therapeutic efficacy of
proteins and extract the drug from the collected organs. Each organ was PTX with an optimum concentration of biodegradable, hemocompatible
weighed separately and homogenised with a homogenizer (10 min, polymer (PLGA), and sustained release polymers (ERSPO/ERLPO).
10,000 rpm, Ultra-Turrax®, T-25, IKA, Germany). The homogenate was Table 1 lists all of the 21 experimental runs.
then centrifuged for 10 min at 2772 xg to extract the drug for its Percent hemolysis, cumulative release in PBS (pH 7.4), and particle
quantitative analysis. The supernatant was transferred to sample vials, size were measured as three responses. The determination of particle
reconstituted with 1 mL of mobile phase, and vortexed for 2 min. Once size correlates with the effect of formulation variables, giving a clear
again, the vials were centrifuged for 15 min at 4928 xg. The supernatant understanding of the internal phase viscosity containing polymers due to
was subsequently collected and filtered using a 0.25 µm membrane fil­ their concentration, type, and proportion. Drug release is one of the
ter, followed by HPLC analysis. critical parameters that predict the mechanism of the drug release from
the particle matrix in physiological conditions. A low release may be due
4. Data analysis and statistics to differences in particle size or the degradation of the polymer. The
conditions of drug release also depends on the tumor environment. The
JMP® version 15.2.1 (SAS Institute, Cary, NC) was used to create the third and most important variable that was analyzed was the percent
design of experiment. All the statistical analyses were done using hemolysis. Hemolysis is one of the most challenging drawbacks of the
GraphPad Prism v 8.0. Phoenix 64 WinNonlin® version 8.3.3.33 chemotherapeutics as the RBC depletion occurs within a few weeks
(Pharsight Corporation, MountainView, CA, USA) was used for non- following the treatment leading to an increased risk and requiring RBC
compartment pharmacokinetic analysis. Results are expressed as mean transfusion. Hence, hemolysis was evaluated as one of the vital variables

5
G. Jeswani et al. Biomedicine & Pharmacotherapy 144 (2021) 112286

Fig. 1. Mixture profiler with contour grids of


polymeric nanoparticles for particle size less
than 200 nm, cumulative drug release at 15
days greater than 70%, and percent hemolysis
less than 10%. The three contour lines represent
the trend in following response variables:
green=particle size; red: drug release; and
blue=hemolysis. The uncolored white region
denotes the operational design space. Five
nanoparticle formulations: PTXNp3, PTXNp5,
PTXNp9, PTXNp11, and PTXNp15 met the set
criteria for the response variables and appeared
in the uncolored region (PTXNp1-PTXNp21 are
represented as 1–21 in the figure).

in the study. Summary of fit (R2 coefficient of determination, and owing to easy penetration through leaky vasculature, impaired
adjusted R2) and analysis of variance (ANOVA) were used to evaluate all lymphatic drainage (EPR effect) and lack of renal clearance [34,35].
the variables. R2 shows that whether there is a good fit between the ANOVA indicated significant influence of all formulation components
results and the model obtained. Higher is the value, the more variation is on particle size (p < 0.0001; R2 = 0.83; adjusted R2 = 0.783). All the
explained, and the better is the model; however, on adding more vari­ polymers impacted the particle size significantly individually ERSPO
ables, adjusted R2 is used to judge the goodness of the model as it only (p = 0.0006), ERLPO (p = 0.0462), and PLGA (<0.0001) and their
counts the significantly affected variables. ANOVA shows whether there two-factor interactions ERSPO*PLGA (p = 0.0317) and ERLPO*PLGA
is any significant difference between the runs or not [32]. Table 1 lists (0.0068). A prediction equation calculated through the best fit model for
the results from all 21 runs. As depicted in Fig. 1, a prediction profiler particle size is given in Eq. (4).

95.14 ∗ ERSPO + 47.78 ∗ ERLPO + 202.88 ∗ PLGA + ERSPO(ERLPO ∗ 89.626) + ERSPO(PLGA ∗ 217.09) + ERLPO(PLGA ∗ 287.43) (4)

shows how different formulation variables influence the responses.


The statistical analysis suggests that the DOE factors including the
composition of the formulation significantly influences the studied re­ According to the equation, all the polymer concentrations exhibited
sponses. An in-depth analysis of all three factors was further done to a positive correlation with particle size. PLGA terms (individual or
define the optimal composition of the formulation. interaction terms) had more prominent constants than ERSPO and
ERLPO terms. Thus PLGA, as a polymer, had a significant impact on
5.2. In vitro characterization to determine particle size, hemolysis, and particle size than other polymers. PLGA has contributed towards an
drug release increase in the overall particle size. This was evident from the particle
size of PTXNp15 (only PLGA matrix) being 214.73 nm. Whereas, the
5.2.1. Particle size mean particle size of PTXNp3 (ERSPO-PLGA combination matrix) was
The particle size was determined to observe the impact of polymer 210.73 nm, and particles devoid of PLGA such as PTXNp1 (ERLPO) and
type and its concentration used in the formulation. The mean particle PTXNp16 (ERSPO) had further smaller sizes of 65.77 nm and 111.83 nm
size of PTXNps ranged between 65.77 ± 1.93 nm to 214.73 ± 4.48 nm respectively. Based on the Eq. (4), the numeric constants for PLGA,
with a relative standard deviation of ≤ 5%, as shown in Table 1. How­ PLGA*ERSPO, PLGA*ERLPO are 202.88, 217.09 and 287.43 respec­
ever, the average particle size of most of the formulations was within tively. This confirms the impact of PLGA and its related terms on overall
250 nm, ideal for accumulation at the tumor site [33]. Particles in the particle size. Comparable findings were also reported by Hasan et al. and
range of 50–300 nm in size are reported to accumulate at the tumor site Cetin et al. [36,37]. As noted elsewhere, an increase in the PLGA

6
G. Jeswani et al. Biomedicine & Pharmacotherapy 144 (2021) 112286

Table 2 hemocompatibility [11,16,42]. Most published studies on PLGA nano­


Effect Summary table for 21 observation. particles have reported hemocompatibility of PLGA nanoparticles
Response R2 R2 Root Mean Mean of p value similar to our results [16,17].
adjusted Square Response Taxanes generally cause anemia due to myelosuppressive effects or
Error drug-induced hemolytic anemia [43]. In a study, PTX caused complete
Particle Size 0.837 0.783 27.428 154.881 <0.0001* hemolysis at the concentration of 200 µg, while polymeric blend nano­
Percent 0.942 0.922 1.526 14.670 <0.0001* particles inhibited the drug-induced hemolysis [44]. According to the
Hemolysis hemolysis reports by Namgung et al., Taxol containing 40 µg of PTX
Release in 0.870 0.827 2.179 71.380
exhibited 100% hemolysis (erythrocytic toxicity) at 20 ppm concentra­
<0.0001*
PBS pH 7.4
tion [45]. A much higher concentration was employed in our study to
determine the effect of nanoparticles on hemolysis.
concentration possibly will increase viscosity and diffusion coefficient,
Our study observed that the percent of hemolysis was reduced to
leading to the formation of larger particles [38].
2.01% (PTXNp15). The results of the study indicated the protective ef­
Nanoparticles prepared using ERLPO showed a smaller mean size
fect of nanoparticles on RBCs. The reported reduction in hemolysis can
compared to ERSPO (PTXNp1 Vs. PTXNp16). Similar findings were re­
be credited to the slow-sustained release of PTX from the polymer ma­
ported by Pignatello et al. (2002), Y. Y. Jiao et al.(2001), and Hasan
trix. Comparatively, the mixture of polymers reduced the hemolysis up
et al.(2012) when they encapsulated flurbiprofen, heparin, and dorzo­
to ~20%, which is five times lesser than the pure drug. Placebo nano­
lamide hydrochloride, respectively. They related this effect to higher
particles exhibited a negligible effect on the RBCs.
quaternary ammonium groups in ERLPO [39–41].
The equation related to hemolysis and varying proportion of the
polymers is given below:
5.2.2. Hemolysis study
Table 1 summarizes the percentage of hemolysis on incubation of

21.77 ∗ ERSPO + 23.34 ∗ ERLPO + 2.99 ∗ PLGA + ERSPO(ERLPO ∗ − 18.77) + ERSPO(PLGA ∗ − 5.18) + ERLPO(PLGA ∗ 3.54) (5)

nanoparticle formulation with RBCs. Hemocompatibility of the formu­


lations was the primary aim of the study, and it was evaluated by This equation depicts that the PLGA has a minor cytotoxic/hemolytic
determining the hemolytic toxicity. Hemolysis is considered a vital effect on the RBCs as represented by its numeric constant, 2.99. PLGA is
screening test to perform according to the ISO10993–4 parameters. It a negatively charged polymer due to carboxyl groups, so it does not form
measures the fragility of the RBC membrane when in contact with a complex with negatively charged RBCs [46]. In contrast, ERSPO and
foreign materials. The hemolysis of all the formulations ranged from 2% ERLPO are positively charged polymers due to quaternary ammonium
to 23%. Lower values were obtained for PTXNP15<PTXNp3 groups present in the molecule, contributing to the formation of the drug
<PTXNp9<PTXNp11<PTXNp6 ranging from ~2–11%. Hemolysis was RBC complex [37,47]. Most hemolysis was observed with formulations
significantly impacted by the presence of ERSPO and ERLPO in the containing only ERLPO (constant value of 23.34) and ERSPO (constant
formulation (p < 0.0001) along with their cross term (p = 0.0022) value of 21.77). In contrast, minimum hemolysis was observed with the
(Table 2). However, the impact of PLGA was comparatively less signif­ PLGA nanoparticles. The hemolysis values of PTXNp1 (ERLPO) was
icant on hemolysis (p < 0.0274). Increasing the proportion of PLGA in 23.23 ± 1.00, PTXNp 16 (ERSPO) was 21 ± 1.23, and PTXNp15 (PLGA)
nanoparticles led to lower hemolysis as PLGA had improved was 2.01 ± 1.27. Thus, PLGA as a polymer has a protective impact on

Table 3
Kinetic modeling of PTX release from different nanoparticle formulations. The quantity of PTX released from PTXNp was calculated by means of validated HPLC
method, using a calibration curve (available in supplementary file, Table S1).
Formulation Code R2 Release exponent

Zero Order First order Higuchi Hixon Korsmeyer- Peppas

PTXNp1 0.849 0.961 0.974 0.931 0.976 0.366


PTXNp2 0.852 0.956 0.977 0.926 0.995 0.276
PTXNp3 0.831 0.938 0.958 0.9 0.948 0.344
PTXNp4 0.798 0.904 0.953 0.871 0.992 0.253
PTXNp5 0.834 0.923 0.97 0.896 0.994 0.263
PTXNp6 0.782 0.871 0.947 0.842 0.994 0.289
PTXNp7 0.831 0.952 0.978 0.923 0.994 0.235
PTXNp8 0.61 0.885 0.948 0.99 0.855 0.256
PTXNp9 0.86 0.91 0.96 0.882 0.988 0.255
PTXNp10 0.816 0.896 0.938 0.858 0.986 0.227
PTXNp11 0.829 0.871 0.947 0.846 0.988 0.235
PTXNp12 0.74 0.871 0.944 0.843 0.985 0.254
PTXNp13 0.798 0.89 0.949 0.86 0.991 0.251
PTXNp14 0.682 0.901 0.958 0.873 0.991 0.259
PTXNp15 0.855 0.842 0.931 0.815 0.989 0.257
PTXNp16 0.845 0.86 0.938 0.831 0.989 0.260
PTXNp17 0.628 0.906 0.921 0.868 0.932 0.262
PTXNp18 0.705 0.887 0.919 0.845 0.98 0.213
PTXNp19 0.872 0.881 0.949 0.851 0.991 0.272
PTXNp20 0.808 0.851 0.923 0.817 0.988 0.250
PTXNp21 0.778 0.807 0.905 0.777 0.981 0.256

7
G. Jeswani et al. Biomedicine & Pharmacotherapy 144 (2021) 112286

RBCs compared to the other polymers. blends and their varying degradation and release mechanisms. There­
fore, the best fit model was selected based on the goodness of fit (R2
5.2.3. In-vitro drug release kinetics values). For each plot, slope and release rate constants were determined.
Table 1 presents the percent cumulative release of 21 PTXNp for­ The same procedure was adopted for each model. The release rate
mulations. The release studies were executed to establish the influence mechanism was also inferred from the same. The correlation coefficient
of the composition of the nanoparticles on the release kinetics and their (R2) was calculated using different release model equations.
in vivo performance. Release data were processed and mapped according Table 3 presents the release profiles of the drug from 21 PTXNp
to different release kinetic equations accompanied by regression ana­ formulations. About 9–20% of drug release was detected in the first 2 h.
lyses to explain the drug release mechanism of from various formula­ The burst release may be caused by two reasons: firstly, the fraction of
tions. Additionally, the in-vitro performance indicates the in-vivo the unincorporated drug weakly bound to the vicinity of the nano­
therapeutic efficacy of the formulation. Defining the exact mechanism, particle matrix or its surface, and secondly, owing to the existence of
order of release for formulations is challenging due to different polymer quaternary ammonium functional group present in the Eudragit

Fig. 2. (A) Transmission electron microscopic (TEM) images of. Scale bar = 1 µm (B) Scanning electron microscopic images (SEM) Scale bar = 2 µm.

8
G. Jeswani et al. Biomedicine & Pharmacotherapy 144 (2021) 112286

molecule, which interacts with water molecules leading to swelling of desirable values were set as following: particle size less than 220 nm,
the polymeric matrix. These results also corroborate with the diffusion- cumulative drug release greater than 50% at 15 days, and percent he­
based drug release pattern, as discussed further in this section. In the molysis less than 10%. Five nanoparticle formulations that met these
nanoparticles, some drugs may be dispersed in the amorphous state in criteria were PTXNp3, PTXNp5, PTXNp9, PTXNp11, and PTXNp15.
the polymer matrix, as reported in FTIR studies. These results are similar In the second step, out of the five formulations, three formulations
to those reported by Dillen et.al regarding the drug being in amorphous/ (PTXNp3, PTXNp9, PTXNp15) were selected based on their low hemo­
semicrystalline state in the nanoparticles [11]. As the amorphous form lysis values and clinical significance of hemolysis. The hemolysis values
has better solubility, it can contribute to the burst release of drug mol­ of these three formulations ranged between 2% and 8%.
ecules from the nanoparticle. As observed in the release study, beyond
2 h, the release rates were meager. The required time to release more
than 65% of the drug from PTXNp is too long (360 h). ANOVA showed 5.4. In vitro and in vivo characterization of selected nanoparticle
that the proportion of polymers significantly impacted the drug release formulations: PTXNp3, PTXNp9, and PTXNp15
(p < 0.0001, Table 2). The R2 obtained for the model was 0.870, and the
R2 adjusted was 0.827. The equation modeling related to drug release 5.4.1. Physical appearance and morphology
and a different proportion of ERSPO, ERLPO, and PLGA is given in Eq. Nanoparticles obtained through the use of nanoprecipitation tech­
(6): nique displayed a uniform appearance. The particles appeared as
distinct entities as seen on transmission electron microscopy images and

70.83 ∗ ERSPO + 84.13 ∗ ERLPO + 66 ∗ PLGA + ERSPO(ERLPO ∗ − 8.928) + ERSPO(PLGA ∗ − 5.610) + ERLPO(PLGA ∗ − 19.568) (6)

also supported by polydispersity index (Fig. 2A) (data available in


supplementary file Table S2). However, as seen in SEM images, the
This equation indicates that a negative impact on drug release is particles had a rough beaded/spiked surface (Fig. 2B). This potentially
exerted by combining ERLPO and PLGA as indicated by the lowest supports them being polydispersed and avoid the adherence of the
interaction coefficient (− 19.568). This trend was followed by the particles to each other. Uneven surfaces have been reported to sustain
negative impact of ERSPO and PLGA combination (coefficient = the drug release. Niu Y et al., 2015 reported improved sustained release
− 5.610). The highest release was obtained from PTXNp1 containing profile from irregular surfaced silica nanoparticles [50]. Similarly, Sun
only ERLPO. This data was corroborated by the highest coefficient of et al. reported prolonged blood circulation time for the rough-surfaced
84.13 associated with ERLPO in Eq. (6). ERSPO/ERLPO polymers have a nanoparticles compared to smooth doxorubicin nanoparticles [51].
quaternary ammonium functional group that leads to quick disintegra­
tion of ERSPO/ERLPO containing nanoparticles. The disintegration of 5.4.2. Zeta (ζ) potential
the particles facilitates the drug release through diffusion, whereas The negative surface charge of the pure PLGA nanoparticles was
PLGA being less hydrophilic, limits the drug release. Consequently, drug provided by the carboxyl groups present on the polymer chain endings,
release from nanoparticles happens due to diffusion and surface/bulk while the positive zeta potential of Eudragit nanoparticles was due to
erosion of PLGA matrix. The difference in the release profile may also be quaternary ammonium groups of Eudragit RSPO and RLPO. For inject­
credited to the variation in particle size of PTXNp. An increase in the ables, positive surface charge on particles increases the cellular uptake
particle size results in an increased length of the diffusional pathways by interacting with the negatively charged mucin, sialic acid, and
and reduced total exposed surface area. Notably, the mixture of the phospholipid present on the cell surface. In contrast, a negative charge is
polymers decreases the drug release [48]. Overall, an increase in the appropriate for the EPR effect [52]. However, in the present studies, a
quantity of ERLPO influenced the dissolution profile due to more blend of Eudragit RSPO, RLPO, and PLGA have provided more neutral
permeability than ERSPO and PLGA. particles due to the shielding effect of the carboxyl group of PLGA on the
The average in vitro release data was processed for zero order, first quaternary ammonium groups of Eudragit. Moreover, some studies
order, Higuchi, Hixon, and Korsmeyer-Peppas release kinetics. Higuchi report that neutral particles are best suited for matrix transport as they
kinetics showed the best fit compared to others (Table 3). These findings do not undergo any repulsive interactions with biomacromolecule fibers
are consistent with the literature where Eudragit and PLGA nano­ present in the tumor’s extracellular matrix [53]. Data presented in
particles were used [49]. It was observed that the Korsmeyer-Peppas Fig. 4C and supplementary data file, Table S2.
model also depicted the data fit for drug release profiles (as seen in
Table 3). The Higuchi model described that the drug on the surface is 5.4.3. Excipients compatibility by Fourier transform infrared and XRD
released at the beginning, followed by that in the core. The drug in the FTIR and XRD analyses were performed for free PTX, PLGA, ERSPO/
center needs to diffuse first to the surface and then solubilize from there. ERLPO, physical mixture, and the prepared nanoparticle (PTXNp18)
A decrease in drug diffusion results in a proportionately lower release (Fig. 3 A). The major infrared peaks assigned to PLGA were observed at
rate. Korsmeyer-Peppas allows exponential analyses of release rate. It 2995 cm− 1 (C–H), 1775 cm− 1 (C− O), 1420 cm− 1 (–CH2), and
was also observed that all the PTXNps followed Fickian diffusion (n less 1253 cm− 1 (CH3). The PTX spectrum showed characteristic peaks at
than 0.5). Conclusively, the release rate of PTX was influenced by the 2954 cm− 1 (C–H), 1649 and 1726 cm− 1 (CO carbonyl ketone group),
drug diffusion rate, leading to a sustained drug release. 1649 cm− 1 (amide group), 1254 cm− 1 (C–O–O bending vibrations),
1085 cm− 1 (C–O stretch vibration), and 704 cm− 1 (aromatic C–H).
ERSPO peaks were observed at 2980 cm− 1 (C–H), 1729 cm− 1 (CO),
5.3. Screening of best formulation from mixture profiler 1461 cm− 1 (–CH2), and 1374 cm− 1 CH(CH3) (Peak intensity was 29.8).
ERLPO peaks were observed at 2980 cm− 1 (C–H), 1728.29 cm− 1 (C-O),
A two tier screening approach was followed to select the optimized 1463 cm− 1 (–CH2), and 1374 cm− 1 CH(CH3) (Peak intensity was 36.84
formulation(s) with minimal toxicity. In the first step, five formulations because of higher electronegativity due to 10% of functional quaternary
were selected from the design. The white/unshaded region in Fig. 1 ammonium group compared to 5% in ERSPO). A rise in peak intensity
portrays the operational design space for the selected formulations. The

9
G. Jeswani et al. Biomedicine & Pharmacotherapy 144 (2021) 112286

Fig. 3. (A) Excipient compatibility by Fourier transform infrared (acronym PM represents PTXNp18). Fig. 3. (B) XRD patterns of (a) PLGA, Eudragit RSPO, and
Eudragit RLPO (b) PTXNp3, PTXNp9, PTXNp15, and PTX. The graph of FTIR is between %Transmittance (Y axis) and wavelength (X axis) and XRD between In­
tensity, counts (Y Axis) and 2 Theta (◦ ), (X Axis).

signifies an increase in the number of the functional group associated decreased, which might be attributed to some PTX molecules present in
with the molecular bond (per unit volume). Noteworthy, all the char­ the amorphous state. The amorphous form allows easy solubility, which
acteristic peaks of drug and polymers were noticeable in the physical allowed burst release from nanoparticles in vitro release studies. The
mixture spectra, confirming the identity. Some shifts and broadening diminished peak for PTX was observed in PTXNp15 formulation as
bands due to overlapping peaks of similar functional groups of both drug compared to PTXNp3 and PTXNp9. Therefore, despite the bigger par­
and polymer were observed in the nanoparticle. No new peaks were ticle size of the PTXNp15 nanoparticle, the results were comparable to
observed, indicating that no chemical change or interaction has taken PTXNp3 and PTXNp9 having smaller particle sizes. Furthermore, the
place and the drug has been physically encapsulated in the polymeric decrease in the crystallinity of polymers assists in the release of drugs
nanoparticles. from polymeric matrices due to its degradation. These findings are
XRD analysis was used to investigate the crystallinity changes within similar to those of Zhang et al., corresponding to PTX loaded PLGA
the drug molecule during nanoparticle formulation. As depicted in microspheres [54].
Fig. 3B, PTX lyophilized powders exhibited a few distinctive diffraction
peaks at 8.9, 12.6, 16.8, and 34.1 scattered angles (2θ), indicating the 5.4.4. Loading and entrapment efficiency
crystalline structure of molecules. The broad peak pattern of ERSPO, All the optimized lyophilized nanoparticle formulations were
ERLPO, and PLGA indicate an amorphous state of polymers (Fig. 3B,a). assessed for entrapment efficiency and drug loading. The entrapment
Meanwhile, in the nanoparticle diffractogram, the characteristic peaks efficiency (%w/w) for all the formulations was > 70%w/w, and loading
of PTX were present at the same location as that in PTX, representing was in the range 8–11%w/w (Fig. 4 A and B). Among the optimized
that PTX in the mixture still exists in crystalline form predominantly formulations, the entrapment efficiency was in the following order
(Fig. 3B,b). Nevertheless, the intensity of PTX diffraction peaks PTXNp15 (78.56 ± 5.25) >PTXNp3 (77.05 ± 4.07) >PTXNp9

10
G. Jeswani et al. Biomedicine & Pharmacotherapy 144 (2021) 112286

Fig. 3. (continued).

(76.23 ± 5.33). Although the ANOVA for entrapment efficiency was required to prevent any hemorrhagic phenomenon. Furthermore, the
insignificant (p = 0.85), pure PLGA nanoparticles (PTXNp15) had the deleterious acts of cytotoxic drugs on RBCs’ membrane results in their
highest entrapment efficiency because of their significant size as destruction causing hemolytic anemia. The activation of the comple­
compared to other nanoparticles (PTXNp3 and PTXNp9); therefore, ment system is responsible for cell inflammation and hemolysis.
entrapment was enhanced. However, an attraction between carboxyl Thereby, an assessment of the bio and hemocompatibility of placebo and
groups (negative charge) on PTX and the quaternary ammonium group loaded nanoparticle helps to establish the safety and efficacy of the
(positive charge) on ERSPO/ERLPO resulted in substantial entrapment nanoparticles. The results presented in Table 4 describe the effects of
efficiency. Minor differences between the PTXNp3 and PTXNp9 can be nanoparticle composition on different hematological parameters.
explained based on the particle size. These findings are consistent with
those reported by Cetin et al., who related the difference with the mo­ 5.4.5.1. Blood clotting time. Lee white method, as described in Section
lecular weight and available charge on the polymer [12]. Similarly, 3.3.5.1, was used to assess polymer ratio impact on the whole blood
there was an insignificant difference in the drug loading for different clotting time [56]. The mean clotting time (in min) for different for­
nanoparticle formulations (p = 0.94) (Fig. 4A). mulations were 5.33 ± 0.29 (PTXNp3); 5.76 ± 0.21 (PTXNp9);
5.67 ± 0.34 (PTXNp15); 5.06 ± 0.19 (PTXNp3 Placebo); 5.70 ± 0.18
5.4.5. Blood compatibility (PTXNp9 Placebo); 5.80 ± 0.15 (PTXNp15 Placebo). The difference
Histological challenges are the major limitation of the chemo­ between clotting time for PTXNp3, PTXNp9, and PTXNp15 was insig­
therapy; hence it is vital to evaluate the hemocompatibility of the nificant (p = 0.16) and differed from the positive control by 266 s
optimized formulation. The shape and size of the nanoparticles (significant; p = 0.05). The mean clotting time for positive control was
contribute to reduced blood compatibility [55]. The characteristic fea­ 1.4 ± 0.32 min (Fig. 5). The clotting time of the nanoparticle treated
tures responsible for hemocompatibility challenges are reactive groups samples was similar to the negative control (p = 0.07). The normal
on the particle surface and a greater surface/volume ratio [55]. For value of clotting time is 5–10 min and our study results complied with
adequate hemocompatibility, the nanoparticles should not activate this range [57]. This study indicates that the nanoparticles did not
platelet cells or the coagulation pathway. The formulation should avoid interfere with the blood clotting mechanism.
any damage to the endothelial cells, resulting in thrombus formation. It
should maintain the normal level of the plasma factors for the coagu­ 5.4.5.2. Blood counts and hemolysis. Table 4 summarizes the values of
lation pathway without creating any imbalance. These ideal features are blood counts obtained with 1 mg of PTX loaded and corresponding

11
G. Jeswani et al. Biomedicine & Pharmacotherapy 144 (2021) 112286

Fig. 4. Graphical representation of drug loading and entrapment efficiency of PTXNp3, PTXNp9, and PTXNp15. (A) Drug loading; (B) Drug entrapment efficiency;
and (C) Zeta(ζ) potential values (mean ± SD; n = 3). (**p < 0.05).

Table 4
The effect of PTXNp on hematological parameters. Each value represents the mean of three determinations ± standard deviation, n = 3. Values in parenthesis indicate
normal value range. PLT (150–400 × 103 /µL); WBC (4–11 × 103 / µL); RBC (4.7–6.1 × 106 / µL); Hb (12–17 g/dL), HCT (37–52%); MCV (77–100 fL); MCH
(27–33 pg); MCHC (33.4–35.5 g/dL). Negative control: phosphate buffer saline.
Formulation Code PLT (103 /µL) WBCs (103 /µL) RBC (106 /µL) Hb (g/dL) HCT (%) MCV (fL) MCH (pg) MCHC (g/dL)
(ERSPO:ERLPO: PLGA)

PTXNp3 (0.2:0:0.8) 337.33 ± 10.01 12.18 ± 1.22 5.72 ± 0.92 11.39 ± 0. 93 37.51 ± 2.25 54.89 ± 3.74 18.67 ± 0.90 37.53 ± 0.73
PTXNp3Placebo 332.90 ± 9.25 10.02 ± 2.08 5.98 ± 0.79 11.68 ± 0.96 37.82 ± 1.10 56.66 ± 3.23 19.23 ± 0.62 36.25 ± 0.50
PTXNp 9(0:0.2:0.8) 357.66 ± 21.78 10.55 ± 1.24 5.54 ± 0.70 11.42 ± 1.25 37.82 ± 1.04 55.16 ± 1.97 18.3 ± 0.65 37.08 ± 0.70
PTXNp 9 Placebo 341.33 ± 8.02 10.34 ± 1.26 5.65 ± 0.61 13.88 ± 1.62 37.12 ± 1.59 55.16 ± 1.55 18.6 ± 0.80 35.73 ± 1.04
PTXNP15 (0:0:1) 365.33 ± 12.66 11.62 ± 1.84 5.51 ± 0.83 12.97 ± 0.41 37.65 ± 0.71 55.6 ± 3.35 18.48 ± 1.05 35.1 ± 2.06
PTXNp15 Placebo 334.66 ± 5.54 12.02 ± 1.53 4.54 ± 0.56 11.75 ± 0.96 37.9 ± 1.43 52.1 ± 1.53 19.17 ± 0.81 35.06 ± 1.71
Negative control 344.33 ± 11.01 11.50 ± 1.07 5.8 ± 0.94 13.38 ± 1.47 38.19 ± 1.57 55.3 ± 0.25 19.4 ± 1.04 34.96 ± 1.09

placebo nanoparticles. When compared to the negative control, none of others surpassed the 5% limit recommended by ISO-10993. However,
the placebo or PTX-loaded nanoparticles had any effect on blood count the difference is marginal, and further improvements can be made to
(PLT, WBC, RBC, MCH, MCV, MCHC, HCT and Hb) (p > 0.05). reduce hemoncompatibility. The obtained value is less than the percent
Fig. 6 shows the hemolysis values of selected PTXNps and corre­ hemolysis caused by Taxol®, as reported by Ran Namgung et. al [45].
sponding placebo nanoparticles. All placebo nanoparticles did not show The group observed hemolysis greater than 40% for 10, 20, and 40 µg of
any significant hemolysis compared to the negative control (p > 0.001). PTX. The results suggest that PTX nanoparticles have better biosafety
The values obtained for PTXNp3, PTXNp9, and PTXNp15 were 7.43%, than Taxol®. Materials with more than 5% hemolysis are classified as
7.89%, and 2.01%, respectively. The results showed that pure PLGA hemolytic, those with 2–5% hemolysis as slightly hemolytic, and those
nanoparticles (PTXNp15) displayed the least hemolysis values while with less than 2% hemolysis as nonhemolytic [58]. Overall, blood count

12
G. Jeswani et al. Biomedicine & Pharmacotherapy 144 (2021) 112286

The clinical etiology of extended clotting times in PT values indicates


reduced levels of factors VII, X, V, prothrombin, or fibrinogen [60].
Although aPTT is considered a sensitive version of the PTT, many factors
influence the results of these tests, like coagulant type, test compounds,
method of sample storage, and the time difference between collection,
processing and analysis. Therefore, all aPTT tests were carried out
within 2 h of collection of a blood sample.
Nanoparticles can have an impact on coagulation time. Shorter pe­
riods of coagulation may lead to toxicity issues due to thrombus for­
mation that could partly or entirely occlude blood vessels, whereas
longer cycles of coagulation might cause bleeding. The incompatibility
of the nanoparticles with the coagulation system may be due to the
biocompatibility issues of the polymers and the negative surface charge
on the nanoparticles. The surface charge on nanoparticles plays an
important role in the aggregation of PLT and hence thrombus formation.
The association between oppositely charged particles and platelets re­
duces the surface charge of PLT and allows them to form aggregates
leading to thrombocytopenia that occurs after injection [61]. Therefore,
Fig. 5. Clotting Time of PTXNp3, PTXNp9, and PTXNp15 compared to their it can be concluded that the neutral particles are safer than the charged
respective placebos (mean ± SD; n = 3). particles in terms of hemocompatibility and transport to tumors [53].
Hence, the lower the surface charge of the particles better is the
hemocompatibility. The values of zeta potential of the nanoparticles
PTXNp3, PTXNp9, and PTXNp15 were − 5.56 ± 0.34, − 5.37 ± 0.47,
and − 7.44 ± 0.41 mV, respectively. The statistical analysis revealed a
significant difference between the zeta potential of the three formula­
tions (p = 0.0014). Pure PLGA particles are highly negatively charged
(− 7.44 ± 0.41 mV) and pure Eudragit RLPO/RSPO particles are highly
positively charged (11.15 ± 0.23/10.5 ± 0.4), the zeta potential of the
developed nanoparticles as a result of the combination of both polymers
is close to neutral.

5.4.5.4. C3 complement. Fig. 8 shows the concentration of C3 protein


complement after incubating the PTXNp and corresponding placebo
nanoparticles with 1 mL of human serum. The expression of comple­
ment C3 protein indicates the association with the coagulation cascade.
Hence, it also indicates the hemolytic nature of nanoparticles. In gen­
eral, nanoparticles form a protein corona upon contact with blood,
which is followed by conformational changes in protein C3 in the
presence of complement factors B and D, resulting in the induction of
enzyme C3 convertase and consecutively splitting of protein C3 into C3a
and C3b, thereby activating the alternative pathway. Hence, C3 deple­
tion in the blood indicates an indirect measurement of complement
activation. Nanoparticles of size less than 100 nm activate the comple­
ment system strongly [62]. However, as the size of the obtained nano­
Fig. 6. Hemolysis of PTXNp3, PTXNp9, and PTXNp15 compared to their
respective placebo particles (mean ± SD; n = 3) (****p < 0.0001; ns =
particles was greater than 100 nm, the values of the C3 complement
not significant). component after incubation for 1 h at 37 ◦ C (108–135 mg/dL)were
within the normal range. The normal range for a complement C3 blood
test is 80–178 mg/dL [63]. Also, the results of all the formulations were
studies suggested that the formulation constituents were biocompatible.
very close to negative control values (114 mg/dL), showing that the
The reason behind the biocompatibility cannot be clearly stated at this
formulations had no effect on complement activation at the particular
stage, other than the restricted exposure of blood components to the
concentration (1 mg) used in this experiment (p = 0.27).
drug and the use of nontoxic polymers.
Therefore, all formulations tested for hemocompatibility as per ISO-
10993 indicated strong hemo-tolerance to blood components. Addi­
5.4.5.3. Activated partial thromboplastin time (aPTT) and prothrombin
tionally, all the research methods used in this section did not require
time (PT) for coagulation. Fig. 7A shows the aPTT values for all nano­
animals as used for in vivo tests.
particle formulations lie within the range 31–36 s Fig. 7B shows the PT
values for all nanoparticle formulations ranging between 13 and 15 s.
5.4.6. In vitro cytotoxicity assay
The aPTT and PT measurement exhibits an effect of nanoparticles
Colorimetric MTT assay was performed against MCF-7 blood cancer
interaction with the coagulation cascade. Results showed that after 4 h
cells to evaluate the cytotoxicity of PTX and the developed nano­
of incubation with a defined nanoparticles concentration (1 mg), there
particles. The results obtained from the in vitro cytotoxicity assay were
was no substantial reduction in prothrombin activity relative to the
represented as a percent cell viability vs. concentration graph (Fig. 9).
negative control, PBS (p = 0.99). Basal aPTT values vary between
The outcomes revealed that all the placebo nanoparticles were non-
32− 36 s. At the same time, PT values range from 12 to 15 s for healthy
cytotoxic against MCF-7 breast cancer cells. PTX loaded nanoparticles
adults aged 18–50 years [59]. Mittal et al. [72] reported similar results
exhibited cell viability ranging between 14% and 99%. Results suggest
where the PLGA nanoparticles of PTX demonstrated hemocompatibility
that all the PTXNp demonstrated concentration-dependent cytotoxicity.
in terms of hemolysis, aPTT and PT measurement.

13
G. Jeswani et al. Biomedicine & Pharmacotherapy 144 (2021) 112286

Fig. 7. Interaction of nanoparticles with the coagulation cascade for selected formulation PTXNp3, PTXNp9, PTXNp15 compared with placebo (A) aPTT (B) PT
(mean ± SD; n = 3). Basal values of the aPTT range between 32 and 36 s while PT ranges between 12 and 15 s for healthy adults of age between 18 and 50 years.
Tests carried out within 2 h of blood sample collection.

Fig. 9. Percent cell viability vs concentration plot obtained from MTT assay of
PTX, PTXNp drug loaded and respective placebo particles against MCF-7 breast
Fig. 8. C3 protein complement activation for PTXNp3, PTXNp9, and PTXNp15
cancer cells after 24 h incubation (mean ± SD; n = 3).
compared to their respective placebo particles (mean ± SD; n = 3).

m2 via the IV route for a duration of three hours. The administered PTX
For the entire concentration range, the cell viability of the respective
dose for this study was chosen at a much lower concentration than the
placebo nanoparticles was significantly higher than the drug-loaded
human equivalent therapeutic dose. Pure PTX solution at the same dose
formulations (p < 0.0001). The lack of cytotoxicity of the placebo
was also administered to allow for pharmacokinetic comparisons. The
nanoparticles confirmed the biocompatibility of matrix polymers,
mortality rate was zero percent for the study.
Eudragit RSPO/RLPO, and PLGA. The IC50 value of PTX was
Fig. 10A depicts log mean plasma drug concentration versus time
40.26 ± 2.70 µg/mL (supplementary data in Fig. S1), and PTXNp3 and
curve for all the PTXNPs and pure PTX solution. In vivo pharmacoki­
PTXNp15 were 3453.30 ± 111.10 and 3382.30 ± 95.32 µg/mL,
netics of PTX for all the treatments was investigated using Wistar rats at
respectively. PTXNp9 exhibited the minimum IC50 value among all the
a dose of 5 mg/kg. A non-compartment analysis was performed to
formulations (1913.15 ± 64.12 µg/mL) (supplementary data in
determine the pharmacokinetic parameters of interest including area
Table S3). The reason behind the enhanced cytotoxicity could be due to
under the curve (AUC); terminal half-life, (T1/2); total body clearance
the presence of positively charged polymers, ERLPO and ERSPO [64].
(Cl), and volume of distribution based on the terminal phase (Vd)
(Table 5). The calculated R2 and R2 adjusted values for all the formu­
5.4.7. In-vivo pharmacokinetic studies
lations were close to 1, indicating a perfect fit to the model. The phar­
As established through the cytotoxicity study results, all three for­
macokinetic curves of the log PTX concentration in the plasma versus
mulations (evaluated within the concentration range of 70–4900 µg/
time are shown in Fig. 10A. The mean AUC of PTX upon IV adminis­
mL) were suitable for the in vivo pharmacokinetic and biodistribution
tration of PTXNp was significantly higher than free PTX (p < 0.0001).
study. All three formulations were individually evaluated using 10–12
For pure PTX solution, the drug was rapidly cleared from the plasma
week old male Wistar rats. The drug loaded formulations were admin­
after bolus administration. On the contrary, all the PTXNps exhibited
istered via the intravenous route at an equivalent PTX dose of 5 mg/kg.
delayed clearance/ higher Clast when compared to PTX solution
Clinically, PTX is generally administered at a dose of 135 and 175 mg/
(PTXNp9 (267 times) > PTXNp3 (213 times) > PTXNp15 (79 times)).

14
G. Jeswani et al. Biomedicine & Pharmacotherapy 144 (2021) 112286

Fig. 10. (A) log Plasma concentration (ng/mL) over time (h) after IV administration of pure drug solution, PTXNp 3, PTXNp9, and PTXNp15 at a dose of 5 mg/kg (B)
Bio-distribution in Wistar rat after 24 h at single dose of 5 mg/kg (C) In vivo hematological parameters observed aat 24 h after a single dose of 5 mg/kg (mean ± SD;
n = 3) (****p < 0.0001).

Table 5
Pharmacokinetic parameters from the in vivo study using pure drug solution, PTXNp3, PTXNp9, and PTXNp15 at a dose of 5 mg/kg (mean ± SD, n = 3). Multiple
comparisons were performed using one-way ANOVA and Tukey post hoc test.
Parametrs PTXNp3 PTXNp9 PTXNp15 Pure drug solution

Avg. SD Avg. SD Avg. SD Avg. SD

R2 0.97 0 0.98 0 1 0 1 0
R2 adjusted 0.96 0 0.97 0 1 0 1 0
Cmax ng/mL 20,482.04 9.97 24,228.96 4.55 22,381.57 7.62 18,317.91 1.52
C0 ng/mL 23,852.72 16.83 31,078.55 18.51 31,921 1.43 29,869.4 58.19
AUC h*ng/mL 141,260.24 128.39 163,071.15 86.68 93,194.67 52.03 39,589.31 238.23
Cl mL/h/kg 32.89 0.03 28.25 0.03 51.59 0.15 126.24 0.77
MRT h 8.29 0.02 8.54 0.02 6.05 0.09 2.04 0.05
Vd mL/kg 291.01 0.19 258.98 0.26 431.41 0.99 438.85 12.57
T1/2 h 6.13 0.36 6.35 0.01 5.88 0.14 2.41 0.08
Clast ng/mL 1216.60 2.60 1521.21 7.58 445.74 18.26 5.69 1.11

Rapid clearance of PTX in the solution group is also confirmed with the The terminal half-life (T1/2), was found to be highest for PTXNp9,
declining PTX concentration for that group. In 24 h, its concentration which is 2.64 times longer than for pure PTX solution (p < 0.0001). The
decreased from peak to below minimum therapeutic effective level volume of distribution is an essential clinical pharmacokinetic param­
(43 ng/mL) [65]. Further, the total area under the curve (AUC), which eter useful in discussing drug disposition processes. Smaller values imply
determines the total systemic exposure to the drug from PTXNps, was swift elimination of the drug from the blood. The PTXNps exhibited low
also higher than pure PTX solution {PTXNp9 (4.12 times) > PTXNp3 clearance (Cl), and volume of distribution (Vd) compared to pure PTX
(3.57 times) > PTXNp15 (2.35 times)}. The difference in the results can solution.
be attributed to the polymer characteristics. As both PLGA (50:50) and The volume of distribution was lowest for PTXNp9 (258.01 mL/kg)
Eudragit RSPO/RLPO are hydrophobic polymers, they resist the drug which was 0.59 times of 438.85 mL/kg for pure PTX solution
release from the polymer matrix. Consequently, the combination of (p < 0.0001). Similarly, PTXNp9 exhibited the lowest clearance
PLGA with Eudragit RSPO/RLPO further reduced the release of PTX (28.25 mL/h/kg) which was 0.22 times of 126.24 mL/h/kg for pure PTX
from the formulation of PTXNp9 and PTXNp3 as compared to PTXNp15 solution (p < 0.0001), which validates the half-life analysis result.
(only PLGA). Hence, the in vitro drug release results corroborate the in Reduced clearance leads to increased plasma half-life, as expected.
vivo pharmacokinetic study results. PTXNp9 increased the MRT of PTX in plasma by 4.19 times. Overall, the

15
G. Jeswani et al. Biomedicine & Pharmacotherapy 144 (2021) 112286

results confirmed improvement in pharmacokinetic profiles when PTX between MCV values for all formulations and the control (p > 0.05).
was formulated as a nanoparticle PTXNps when compared to PTX Higher MCV value indicate abnormal size of RBCs.
solution. The MCH value for the PTXNp3, PTXNp9, PTXNp15 and control
The in vivo results were corroborated by the in vitro drug release were 19.19 ± 1.57, 19.09 ± 2.55, 19.74 ± 1.72, 19.22 ± 3.56 respec­
studies where all the formulations released only upto 20% of the drug tively. All PTXNps showed MCH values < 25 pg with no significant
within 2 h. After 360 h, the cumulative drug released was in the range of differenece when compared with control (p > 0.05). Higher MCH values
65–82% w/w, confirming the slow, sustained release of the drug. indicate abnormal/large size of RBCs. Similarly, MCHC values of
Similarly, the in vivo results support the prolonged circulation times of PTXNp3, PTXNp9, PTXNp15 and control were 38.39 ± 1.67,
the nanoparticles in vivo along with a longer plasma half-life. Also, the in 38.01 ± 1.35, 40.24 ± 1.06, and 37.22 ± 2.37 g/dL respectively. All
vivo results confirmed the significant safety and efficacy of the nano­ PTXNps showed MCHC values < 40 g/dL with no significant differenece
particle formulations versus the pure PTX solution. Similarly, the safety when compared with control (p > 0.05). Low MCHC values indicate less
profile was also evident via the in vitro cytotoxicity studies mentioned amount of Hb in the RBCs. The hematological parameters were used to
earlier. Based on the in vitro and in vivo study results, PTXNp9 is the most diagnose the effect of nanoparticles on the RBCs. Since all the polymers
optimum formulation among the three optimized formulations were used below their LD50 values, no toxicity or adverse effects were
(PTXNp3, PTXNp9, and PTXNp15). With further modifications, it can observed. The values of all haematological tests [as shown in Fig. 10(C)]
further be effectively used to gain site-specific drug delivery. for all groups treated with nanoparticles were close to the control, thus
confirming the safety and biocompatibility of the formulations.
5.4.8. Biodistribution and hematological studies
A biodistribution study was conducted to quantify the amount of 6. Limitation
drug deposited in different organs. Five cardinal organs were selected
for the study: heart, spleen, kidney, lung, and liver. Fig. 10B shows that To get an idea of the off-target effects of the developed nanoparticles
the nanoparticulate formulations led to a substantially higher level of the present study has been limited to blood compatibility studies and
drug delivery to all the isolated organs than the pure drug solution biodistribution studies in detail. However, the microtubule stability is
correlating to the sustained drug release of PTX from the nanoparticles. also important activity to be noted as microtubules are important for cell
PTX was majorly distributed to the liver, spleen, and heart. division, specifically in the segregation of chromosomes.
A significantly higher amount of drug was observed in the liver in the
group receiving PXTNp formulations (p < 0.0001) as compared to the
7. Conclusion
group receiving pure PTX solution. Similar results have been reported by
De Jong et al., where the distribution of gold nanoparticles to the liver
The study utilizes a statistical design to formulate a nanoparticulate
occurred preferentially irrespective of the size of the nanoparticles [66].
sustained release drug delivery systems for PTX. This is the first study to
Li et al. also reported significant levels of PTX in the liver, spleen, kid­
evaluate the role of such formulations on hemocompatibility parameters
ney, heart, and lung [67]. The study reported an eight-fold higher PTX
as specified by the ISO-10993. The study evaluates the formulated
levels in the liver than the other organs. The reason for this selective
nanoparticles via both in vitro and in vivo analysis. The in vitro drug
accumulation is the fact that plasma proteins rapidly coat nanoparticles
release profiles corroborated the in vivo pharmacokinetic profiles of the
after an intravenous injection. Protein-coated nanoparticles easily
nanoparticles. Additionally, the biodistribution studies provided a
accumulate in the liver while those present in the bloodstream diffuse
unique understanding of drug distribution to five cardinal organs and
into the tumor upon circulation [68]. Overall, we infer that all three
the impact of PTX delivery to those sites. Overall, the formulation and
PTXNp formulations had a significant effect on the biodistribution of the
evaluation approaches offered in this study are promising to deliver PTX
drug, and the extent of biodistribution of PTX was organ-dependent with
in a safe, efficient, and reliable manner to enhance its clinical outcomes
the maximum accumulation in the liver.
and reduce the associated adverse effects.
The hematological analysis, Fig. 10C, showed that the concentration
of PLT in the group that received PTXNps was substantially higher
compared to the group treated with pure PTX solution (p < 0.0001). CRediT authorship contribution statement
Also, the RBC count for the PTX solution group was lower (3.81 × 106
/µL) compared to PTXNp9 treated group (5.54 × 106 /µL). RBCs are Gunjan Jeswani: Conceptualization, Investigation, Methodology,
vital for haemoglobin levels and to transport oxygen. Thus, lack of RBCs Formal analysis, Data curation, Writing − review & editing. Lipika
can be detrimental clinically. Reduced RBC counts indicate toxic effects Chablani: Conceptualization, Methodology, Formal analysis, Data
of the treatment [69,70]. The WBCs counts were 11.62 ± 1.84 × 103 curation, Writing − review & editing, Supervision. Umesh Gupta:
per µL for the PTXNp15 treated group, 10.55 ± 1.24 × 103 per µL for Investigation, Methodology, Data curation, Writing. Rakesh Kumar
the PTXNp9 treated group, 12.18 ± 1.22 × 103 per µL for the PTXNp3 Sahoo: Investigation, Methodology, Data curation, Writing, Formal
treatment, 3.47 ± 0.62 × 103 per µL for pure drug solution, and analysis. Kartik T. Nakhate: Investigation, Methodology. Ajazuddin:
11.50 ± 1.079 × 103 per µL for the control group. Hb levels were close Supervision, Conceptualization, Methodology.
to the values of control group (13.38 ± 1.4 g /dL), in PTXNp treated
groups (PTXNp3 11.39 ± 0.93 g /dL, PTXNp9 11.42 ± 1.2 and
PTXNp15 12.97 ± 0.41) as compared to the pure drug solution Conflict of interest statement
(7.95 ± 0.08 g/dL). The HCT value for the PTXNp3, PTXNp9 and
PTXNp15 were 27.39 ± 0.78, 29.69 ± 0.75, 30.44 ± 0.72, The authors declare that there are no conflicts of interest.
31.75 ± 1.74 respectively. All PTXNps showed HCT values < 45%
which is within the reference value and with no significant differenece Acknowledgment
when compared with control, 31.74 ± 1.72 (p > 0.05) [71]. HCT mea­
sures the percentage of red blood cells in the sample of blood. It is an All the research studies were done under the joint supervision of Dr.
indirect method of measuring Hb. Thus, the normal values obtained for Ajazuddin and Dr. Lipika Chablani. We acknowledge Intas Pharmaceu­
the PTXNp treated groups are indicative of healthy RBC counts and no ticals Ltd. for providing Paclitaxel as a gift and Evonik India Private Ltd.
blood disorder. The MCV value for the PTXNp3, PTXNp9, PTXNp15 and for providing ERSPO, ERLPO, and PLGA samples. Special thanks to Dr.
control were 55.48 ± 1.45, 55.05 ± 1.50, 55.44 ± 1.27, and Naveen Verma, M.D Pathology, for his support during the hemo­
51.00 ± 1.26 respectively. Moreover, there was no statistical difference compatibility studies.

16
G. Jeswani et al. Biomedicine & Pharmacotherapy 144 (2021) 112286

Appendix A. Supporting information [23] A. Mayer, M. Vadon, B. Rinner, A. Novak, R. Wintersteiger, E. Fröhlich, The role of
nanoparticle size in hemocompatibility, Toxicology 258 (2009) 139–147, https://
doi.org/10.1016/j.tox.2009.01.015.
Supplementary data associated with this article can be found in the [24] K.P. Kumar, W. Paul, C.P. Sharma, Green synthesis of silver nanoparticles with
online version at doi:10.1016/j.biopha.2021.112286. zingiber officinale extract and study of its blood compatibility, Bionanoscience 2
(2012) 144–152, https://doi.org/10.1007/s12668-012-0044-7.
[25] T. Mosmann, Rapid colorimetric assay for cellular growth and survival: application
References to proliferation and cytotoxicity assays, J. Immunol. Methods 65 (1983) 55–63,
https://doi.org/10.1016/0022-1759(83)90303-4.
[1] Global Cancer Observatory, n.d. 〈https://gco.iarc.fr/〉 (accessed 25 June 2021). [26] A.K. Sharma, L. Gupta, H. Sahu, A. Qayum, S.K. Singh, K.T. Nakhate, Ajazuddin,
[2] G. Jeswani, A. Alexander, S. Saraf, S. Saraf, A. Qureshi, Ajazuddin, Recent U. Gupta, Chitosan engineered PAMAM dendrimers as nanoconstructs for the
approaches for reducing hemolytic activity of chemotherapeutic agents, J. Control. enhanced anti-cancer potential and improved in vivo brain pharmacokinetics of
Release 211 (2015) 10–21, https://doi.org/10.1016/j.jconrel.2015.06.001. temozolomide, Pharm. Res. 35 (2018) 1–14, https://doi.org/10.1007/s11095-017-
[3] I. Takeuchi, K. Makino, Colloids and Surfaces B: biointerfaces Biocompatibility and 2324-y.
e ff ectiveness of paclitaxel-encapsulated micelle using phosphoester compounds as [27] R.K. Averineni, G.V. Shavi, A.K. Gurram, P.B. Deshpande, K. Arumugam,
a carrier for cancer treatment, Colloids Surf. B Biointerfaces 177 (2019) 356–361, N. Maliyakkal, S.R. Meka, U. Nayanabhirama, PLGA 50:50 nanoparticles of
https://doi.org/10.1016/j.colsurfb.2019.02.017. paclitaxel: development, in vitro anti-tumor activity in BT-549 cells and in vivo
[4] J. Kydd, R. Jadia, P. Velpurisiva, A. Gad, S. Paliwal, P. Rai, Targeting strategies for evaluation, Bull. Mater. Sci. 35 (2012) 319–326, https://doi.org/10.1007/s12034-
the combination treatment of cancer using drug delivery systems, Pharmaceutics 9 012-0313-7.
(2017), https://doi.org/10.3390/pharmaceutics9040046. [28] L. Wang, X. Zhao, F. Yang, W. Wu, M. Wu, Y. Li, X. Zhang, Loading paclitaxel into
[5] R.S. Faria, L.I. de Lima, R.S. Bonadio, J.P.F. Longo, M.C. Roque, J.N. de Matos porous starch in the form of nanoparticles to improve its dissolution and
Neto, S.E. Moya, M.C. de Oliveira, R.B. Azevedo, Liposomal paclitaxel induces bioavailability, Int. J. Biol. Macromol. 138 (2019) 207–214, https://doi.org/
apoptosis, cell death, inhibition of migration capacity and antitumoral activity in 10.1016/j.ijbiomac.2019.07.083.
ovarian cancer, Biomed. Pharmacother. 142 (2021), 112000, https://doi.org/ [29] A.K. Dash, Editorial: the dark side of paclitaxel, Oncol. Rev. 4 (2010) 71–72,
10.1016/J.BIOPHA.2021.112000. https://doi.org/10.1007/s12156-010-0052-1.
[6] F. Wang, M. Porter, A. Konstantopoulos, P. Zhang, H. Cui, Preclinical development [30] J. Mu, H. Zhong, H. Zou, T. Liu, N. Yu, X. Zhang, Z. Xu, Z. Chen, S. Guo, Acid-
of drug delivery systems for paclitaxel-based cancer chemotherapy, J. Control. sensitive PEGylated paclitaxel prodrug nanoparticles for cancer therapy: effect of
Release 267 (2017) 100–118, https://doi.org/10.1016/j.jconrel.2017.09.026. PEG length on antitumor efficacy, J. Control. Release 326 (2020) 265–275, https://
[7] W. Lu, J. Yao, X. Zhu, Y. Qi, Nanomedicines: redefining traditional medicine, doi.org/10.1016/j.jconrel.2020.07.022.
Biomed. Pharmacother. 134 (2021), 111103, https://doi.org/10.1016/j. [31] T. Aomori, H. Makino, M. Sekizuka, T. Hashita, T. Araki, K. Iizuka, T. Nakamura,
biopha.2020.111103. K. Yamamoto, Effect of ethanol in paclitaxel injections on the ethanol
[8] PLGA-Based Nanoparticles in Cancer Treatment Sima Rezvantalab.pdf, n.d. concentration in exhaled breath, Drugs R. D. 12 (2012) 165–170, https://doi.org/
[9] A. Gandhi, S. Jana, K.K. Sen, In-vitro release of acyclovir loaded Eudragit RLPO® 10.2165/11634690-000000000-00000.
nanoparticles for sustained drug delivery, Int. J. Biol. Macromol. 67 (2014) [32] H.I. Shahin, B.P. Vinjamuri, A.A. Mahmoud, R.N. Shamma, S.M. Mansour, H.
478–482, https://doi.org/10.1016/j.ijbiomac.2014.04.019. O. Ammar, M.M. Ghorab, M.B. Chougule, L. Chablani, Design and evaluation of
[10] Y. Jiao, N. Ubrich, M. Marchand-Arvier, C. Vigneron, M. Hoffman, T. Lecompte, novel inhalable sildenafil citrate spray-dried microparticles for pulmonary arterial
P. Maincent, In vitro and in vivo evaluation of oral heparin-loaded polymeric hypertension, J. Control. Release 302 (2019) 126–139, https://doi.org/10.1016/j.
nanoparticles in rabbits, Circulation 105 (2002) 230–235, https://doi.org/ jconrel.2019.03.029.
10.1161/hc0202.101988. [33] E. Russo, N. Gaglianone, S. Baldassari, B. Parodi, I. Croce, A.M. Bassi, S. Vernazza,
[11] K. Dillen, J. Vandervoort, G. Van den Mooter, A. Ludwig, Evaluation of G. Caviglioli, Chitosan-clodronate nanoparticles loaded in poloxamer gel for intra-
ciprofloxacin-loaded Eudragit® RS100 or RL100/PLGA nanoparticles, Int. J. articular administration, Colloids Surf. B Biointerfaces 143 (2016) 88–96, https://
Pharm. 314 (2006) 72–82, https://doi.org/10.1016/j.ijpharm.2006.01.041. doi.org/10.1016/j.colsurfb.2016.03.028.
[12] M. Cetin, A. Atila, Y. Kadioglu, Formulation and in vitro characterization of [34] H. Kobayashi, R. Watanabe, P.L. Choyke, Improving conventional enhanced
Eudragit® L100 and Eudragit® L100-PLGA nanoparticles containing diclofenac permeability and retention (EPR) effects; what is the appropriate target?
sodium, AAPS PharmSciTech 11 (2010) 1250–1256, https://doi.org/10.1208/ Theranostics 4 (2014) 81–89, https://doi.org/10.7150/thno.7193.
s12249-010-9489-6. [35] Y. Nakamura, A. Mochida, P.L. Choyke, H. Kobayashi, Nanodrug delivery: is the
[13] K. Anwer, R. Al-shdefat, E. Ezzeldin, S.M. Alshahrani, Preparation, Evaluation and enhanced permeability and retention effect sufficient for curing cancer? Bioconjug.
Bioavailability Studies of Eudragit Coated PLGA Nanoparticles for Sustained Chem. 27 (2016) 2225–2238, https://doi.org/10.1021/acs.bioconjchem.6b00437.
Release of Eluxadoline for the Treatment of Irritable Bowel Syndrome, 8, 2017: [36] A.A. Hasan, S.A. Sabry, M.H. Abdallah, D.A. El-damasy, Formulation and in vitro
1–11. https://doi.org/10.3389/fphar.2017.00844. characterization of poly(dl-lactide-co-glycolide)/Eudragit RLPO or RS30D
[14] A. Basarkar, J. Singh, Poly (lactide-co-glycolide)-polymethacrylate nanoparticles nanoparticles as an oral carrier of levofloxacin hemihydrate, Pharm. Dev. Technol.
for intramuscular delivery of plasmid encoding interleukin-10 to prevent 21 (2016) 655–663, https://doi.org/10.3109/10837450.2015.1041044.
autoimmune diabetes in mice, Pharm. Res. 26 (2009) 72–81, https://doi.org/ [37] M. Cetin, A. Atila, S. Sahin, I. Vural, Preparation and characterization of metformin
10.1007/s11095-008-9710-4. Preparation and characterization of metformin, 7450, 2013. https://doi.org/
[15] H. Fessi, F. Puisieux, J.P. Devissaguet, N. Ammoury, S. Benita, Nanocapsule 10.3109/10837450.2011.604783.
formation by interfacial polymer deposition following solvent displacement, Int. J. [38] W. Huang, C. Zhang, Tuning the size of poly (lactic-co-glycolic acid)(PLGA)
Pharm. 55 (1989) R1–R4, https://doi.org/10.1016/0378-5173(89)90281-0. nanoparticles fabricated by nanoprecipitation, Biotechnol. J. 13 (2018) 1–19,
[16] E. Cenni, D. Granchi, S. Avnet, C. Fotia, M. Salerno, D. Micieli, M.G. Sarpietro, https://doi.org/10.1002/biot.201700203.Tuning.
R. Pignatello, F. Castelli, N. Baldini, Biocompatibility of poly(d,l-lactide-co- [39] R. Pignatello, C. Bucolo, G. Puglisi, Ocular tolerability of Eudragit RS100® and
glycolide) nanoparticles conjugated with alendronate, Biomaterials 29 (2008) RL100® nanosuspensions as carriers for ophthalmic controlled drug delivery,
1400–1411, https://doi.org/10.1016/j.biomaterials.2007.12.022. J. Pharm. Sci. 91 (2002) 2636–2641, https://doi.org/10.1002/jps.10227.
[17] C. Fornaguera, G. Calderó, M. Mitjans, M.P. Vinardell, C. Solans, C. Vauthier, [40] Y. Jiao, N. Ubrich, M. Marchand-Arvier, C. Vigneron, M. Hoffman, T. Lecompte,
Interactions of PLGA nanoparticles with blood components: Protein adsorption, P. Maincent, In vitro and in vivo evaluation of oral heparin-loaded polymeric
coagulation, activation of the complement system and hemolysis studies, nanoparticles in rabbits, Circulation 105 (2002) 230–235, https://doi.org/
Nanoscale 7 (2015) 6045–6058, https://doi.org/10.1039/c5nr00733j. 10.1161/hc0202.101988.
[18] C. Yan, N. Liang, Q. Li, P. Yan, S. Sun, Biotin and arginine modified hydroxypropyl- [41] A.A. Hasan, Formulation and evaluation of dorzolamide hydrochloride-loaded
β-cyclodextrin nanoparticles as novel drug delivery systems for paclitaxel, nanoparticles as controlled release drug delivery system, 2012, 67–73. https://doi.
Carbohydr. Polym. 216 (2019) 129–139, https://doi.org/10.1016/j. org/10.4103/0973–8398.100148.
carbpol.2019.04.024. [42] R. Sankar, V. Ravikumar, Biocompatibility and biodistribution of suberoylanilide
[19] X. Dong, C.A. Mattingly, M. Tseng, M. Cho, V.R. Adams, R.J. Mumper, hydroxamic acid loaded poly (DL-lactide-co-glycolide) nanoparticles for targeted
Development of new lipid-based paclitaxel nanoparticles using sequential simplex drug delivery in cancer, Biomed. Pharmacother. 68 (2014) 865–871, https://doi.
optimization, Eur. J. Pharm. Biopharm. 72 (2009) 9–17, https://doi.org/10.1016/ org/10.1016/j.biopha.2014.07.015.
j.ejpb.2008.11.012. [43] R.M. Leger, S. Jain, T.A. Nester, H. Kaplan, Drug-induced immune hemolytic
[20] M. Huo, H. Wang, Y. Zhang, H. Cai, P. Zhang, L. Li, J. Zhou, T. Yin, Co-delivery of anemia associated with anti-carboplatin and the first example of anti-paclitaxel,
silybin and paclitaxel by dextran-based nanoparticles for effective anti-tumor Transfusion 55 (2015) 2949–2954, https://doi.org/10.1111/trf.13255.
treatment through chemotherapy sensitization and microenvironment modulation, [44] W. Gu, J. Chen, P. Patra, X. Yang, Q. Gu, L. Wei, J.P. Acker, B. Kong,
J. Control. Release 321 (2020) 198–210, https://doi.org/10.1016/j. Nanoformulated water-soluble paclitaxel to enhance drug efficacy and reduce
jconrel.2020.02.017. hemolysis side effect, J. Biomater. Appl. 32 (2017) 66–73, https://doi.org/
[21] N.R. Kuznetsova, C. Sevrin, D. Lespineux, N.V. Bovin, E.L. Vodovozova, 10.1177/0885328217708458.
T. Mészáros, J. Szebeni, C. Grandfils, Hemocompatibility of liposomes loaded with [45] R. Namgung, Y. Mi Lee, J. Kim, Y. Jang, B.H. Lee, I.S. Kim, P. Sokkar, Y.M. Rhee, A.
lipophilic prodrugs of methotrexate and melphalan in the lipid bilayer, J. Control. S. Hoffman, W.J. Kim, Poly-cyclodextrin and poly-paclitaxel nano-assembly for
Release 160 (2012) 394–400, https://doi.org/10.1016/j.jconrel.2011.12.010. anticancer therapy, Nat. Commun. 5 (2014) 1–12, https://doi.org/10.1038/
[22] P. Shobharani, P.M. Halami, N.M. Sachindra, Potential of marine lactic acid ncomms4702.
bacteria to ferment Sargassum sp. for enhanced anticoagulant and antioxidant [46] N.K. Al-Nemrawi, A.R. Okour, R.H. Dave, Surface modification of PLGA
properties, J. Appl. Microbiol. 114 (2013) 96–107, https://doi.org/10.1111/ nanoparticles using chitosan: effect of molecular weight, concentration, and degree
jam.12023.

17
G. Jeswani et al. Biomedicine & Pharmacotherapy 144 (2021) 112286

of deacetylation, Adv. Polym. Technol. 37 (2018) 3066–3075, https://doi.org/ coagulation tests performed using destiny plus, Iran. J. Pediatr. 26 (2016) 3–10,
10.1002/adv.22077. https://doi.org/10.5812/ijp.6177.
[47] R. Tomas, A. Shillingburg, Drug-induced immune hemolytic anemia associated [60] M. Roshal, Prothrombin time, in: Transfus. Med. Hemost. Clin. Lab. Asp., Second
with albumin-bound paclitaxel, J. Community Support. Oncol. 13 (2015) 298–299, ed.,, Elsevier Inc, 2013, pp. 799–803, https://doi.org/10.1016/B978-0-12-397164-
https://doi.org/10.12788/jcso.0160. 7.00124-5.
[48] V. Hoffart, N. Ubrich, C. Simonin, V. Babak, C. Vigneron, M. Hoffman, [61] M.A. Abdel-Wahhab, K.G. Abdel-Wahhab, F.A. Mannaa, N.S. Hassan, R. Safar,
T. Lecompte, P. Maincent, Low molecular weight heparin-loaded polymeric R. Diab, B. Foliguet, L. Ferrari, B.H. Rihn, Uptake of Eudragit Retard L (Eudragit®
nanoparticles: formulation, characterization, and release characteristics, Drug Dev. RL) nanoparticles by human THP-1 cell line and its effects on hematology and
Ind. Pharm. 28 (2002) 1091–1099, https://doi.org/10.1081/DDC-120014576. erythrocyte damage in rats, Mater. (Basel) 7 (2014) 1555–1572, https://doi.org/
[49] A.A. Hasan, S.A. Sabry, M.H. Abdallah, D.A. El-damasy, Formulation and in vitro 10.3390/ma7031555.
characterization of poly (DL -lactide-co-glycolide)/ Eudragit RLPO or RS30D [62] G.L. Szeto, E.B. Lavik, Materials design at the interface of nanoparticles and innate
nanoparticles as an oral carrier of levofloxacin hemihydrate, Pharm. Dev. Technol. immunity, J. Mater. Chem. B. 4 (2016) 1610–1618, https://doi.org/10.1039/
00 (2015) 1–9, https://doi.org/10.3109/10837450.2015.1041044. c5tb01825k.
[50] Y. Niu, M. Yu, A. Meka, Y. Liu, J. Zhang, Y. Yang, C. Yu, Understanding the [63] D.J. Birmingham, F. Irshaid, H.N. Nagaraja, X. Zou, B.P. Tsao, H. Wu, C.Y. Yu, L.A.
contribution of surface roughness and hydrophobic modification on silica Hebert, B.H. Rovin, The complex nature of serum C3 and C4 as biomarkers of lupus
nanoparticles for enhanced therapeutic protein delivery, J. Mater. Chem. B 4 renal flare, n.d. https://doi.org/10.1177/0961203310371154.
(2015) 212–219, https://doi.org/10.1039/C5TB01911G. [64] E. Fröhlich, The role of surface charge in cellular uptake and cytotoxicity of
[51] X. Sun, M. Li, Y. Yang, H. Jia, W. Liu, Carrier-free nanodrug-based virus-surface- medical nanoparticles, Int. J. Nanomed. 7 (2012) 5577–5591, https://doi.org/
mimicking nanosystems for efficient drug/gene co-delivery, Biomater. Sci. 6 10.2147/IJN.S36111.
(2018) 3300–3308, https://doi.org/10.1039/c8bm01033a. [65] Y. Dong, S.S. Feng, In vitro and in vivo evaluation of methoxy polyethylene glycol-
[52] S. Honary, F. Zahir, Effect of zeta potential on the properties of nano-drug delivery polylactide (MPEG-PLA) nanoparticles for small-molecule drug chemotherapy,
systems − a review (Part 2), Trop. J. Pharm. Res. 12 (2013) 265–273, https://doi. Biomaterials 28 (2007) 4154–4160, https://doi.org/10.1016/j.
org/10.4314/tjpr.v12i2.20. biomaterials.2007.05.026.
[53] H. Holback, Y. Yeo, Intratumoral drug delivery with nanoparticulate carriers, [66] W.H. De Jong, W.I. Hagens, P. Krystek, M.C. Burger, Particle size-dependent organ
Pharm. Res. 28 (2011) 1819–1830, https://doi.org/10.1007/s11095-010-0360-y. distribution of gold nanoparticles after intravenous administration, 29, 2008:
[54] Z. Zhang, X. Wang, B. Li, Y. Hou, Z. Cai, J. Yang, Y. Li, Paclitaxel-loaded PLGA 1912–1919. https://doi.org/10.1016/j.biomaterials.2007.12.037.
microspheres with a novel morphology to facilitate drug delivery and antitumor [67] R. Li, J.S. Eun, M.K. Lee, Pharmacokinetics and biodistribution of paclitaxel loaded
efficiency, RSC Adv. 8 (2018) 3274–3285, https://doi.org/10.1039/c7ra12683b. in pegylated solid lipid nanoparticles after intravenous administration, Arch.
[55] K.M. de la Harpe, P.P.D. Kondiah, Y.E. Choonara, T. Marimuthu, L.C. du Toit, Pharm. Res. 34 (2011) 331–337, https://doi.org/10.1007/s12272-011-0220-2.
V. Pillay, The hemocompatibility of nanoparticles: a review of cell-nanoparticle [68] D. Van Haute, J. o Berlin, Challenges in realitzin selectivitat for nanoparticle
interactions and hemostasis, Cells 8 (2019), https://doi.org/10.3390/ Biodistribution and clearance: lesions form Gold nanoparticles, Ther. Deliv. 8
cells8101209. (2016) 763–774.
[56] Z. He, Z. Shi, W. Sun, J. Ma, J. Xia, X. Zhang, W. Chen, J. Huang, [69] R.N. Pittman, Oxygen transport in the microcirculation and its regulation,
Hemocompatibility of folic-acid-conjugated amphiphilic PEG-PLGA copolymer Microcirculation 20 (2013) 117–137, https://doi.org/10.1111/micc.12017.
nanoparticles for co-delivery of cisplatin and paclitaxel: treatment effects for non- [70] V.V. Revin, N.V. Gromova, E.S. Revina, I.P. Grunyushkin, A.Y. Tychkov, A.
small-cell lung cancer, Tumor Biol. 37 (2016) 7809–7821, https://doi.org/ Y. Samonova, A.N. Kukina, A.A. Moskovkin, J.-C. Bourdon, N. Zhelev, The effect of
10.1007/s13277-015-4634-1. experimental hyperoxia on erythrocytes’ oxygen-transport function, Biotechnol.
[57] J.D. de Brito Sousa, J.A.G. Sachett, S.S. de Oliveira, I. Mendonça-da-Silva, H. Biotechnol. Equip. 32 (2018) 1236–1250, https://doi.org/10.1080/
O. Marques, M.V.G. de Lacerda, H.W. Fan, W.M. Monteiro, Accuracy of the 13102818.2017.1414633.
lee–white clotting time performed in the hospital routine to detect coagulopathy in [71] S.L. Delwatta, M. Gunatilake, V. Baumans, M.D. Seneviratne, M.L.B. Dissanayaka,
Bothrops atrox envenomation, Am. J. Trop. Med. Hyg. 98 (2018) 1547–1551, S.S. Batagoda, A.H. Udagedara, P.B. Walpola, Reference values for selected
https://doi.org/10.4269/ajtmh.17-0992. hematological, biochemical and physiological parameters of Sprague-Dawley rats
[58] M. Weber, H. Steinle, S. Golombek, L. Hann, C. Schlensak, H.P. Wendel, M. Avci- at the Animal House, Faculty of Medicine, University of Colombo, Sri Lanka, Anim.
Adali, Blood-contacting biomaterials: in vitro evaluation of the hemocompatibility, Model. Exp. Med. 1 (2018) 250–254, https://doi.org/10.1002/ame2.12041.
Front. Bioeng. Biotechnol. 6 (2018) 99, https://doi.org/10.3389/ [72] P. Mittal, H. Vardhan, G. Ajmal, G. Vasant Bonde, R. Kapoor, A. Mittal, B. Mishra,
fbioe.2018.00099. Formulation, optimization, hemocompatibility and pharmacokinetic evaluation of
[59] F.D. Arslan, M. Serdar, E.M. Ari, M.O. Oztan, S.H. Kozcu, H. Tarhan, O. Cakmak, PLGA nanoparticles containing paclitaxel, Drug Dev. Ind. Pharm. 45 (3) (2019)
M. Zeytinli, H.Y. Ellidag, Determination of age-dependent reference ranges for 365–378, https://doi.org/10.1080/03639045.2018.1542706.

18

You might also like