In Vivo Pulmonary Delivery and Magnetic ... - ACS Publications

Oct 25, 2017 - compared to all other types of cancer accounting for 26.5% of all cancer deaths.1−3 Conventional ..... using GraphPad Prism statistic...
2 downloads 12 Views 1MB Size
Subscriber access provided by READING UNIV

Brief Article

In vivo pulmonary delivery and magnetictargeting of dry powder nano-in-microparticles Dominique N. Price, Loreen R. Stromberg, Nitesh K Kunda, and Pavan Muttil Mol. Pharmaceutics, Just Accepted Manuscript • DOI: 10.1021/acs.molpharmaceut.7b00532 • Publication Date (Web): 25 Oct 2017 Downloaded from http://pubs.acs.org on October 30, 2017

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

Molecular Pharmaceutics is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 31

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

1 2 3

Molecular Pharmaceutics

In vivo pulmonary delivery and magnetictargeting of dry powder nano-inmicroparticles

4 5

Dominique N. Price1, Loreen R. Stromberg1,2, Nitesh K. Kunda1, and Pavan Muttil1,3*

6 7

1

8

Health Sciences Center, Albuquerque, New Mexico

9

2

Department of Mechanical Engineering, Iowa State University, Ames, Iowa

10

3

The University of New Mexico Cancer Center, Albuquerque, New Mexico

Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico

11 12 13

Conflicts of Interest: The authors have no conflicts of interest to declare.

14 15 16

ACS Paragon Plus Environment

1

Molecular Pharmaceutics

Page 2 of 31

1 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

2

Abstract

3

This brief communication evaluates the cytotoxicity and targeting capability of a dry powder

4

chemotherapeutic. Nano-in-microparticles (NIMs) are a dry powder drug delivery vehicle

5

containing superparamagnetic iron oxide nanoparticles (SPIONs) and either doxorubicin (w/w

6

solids) or fluorescent nanospheres (w/v during formulation; as a drug surrogate) in a lactose

7

matrix. In vitro cytotoxicity was evaluated in A549 adenocarcinoma cells using MTS and LDH

8

assays to assess viability and toxicity after 48 hours of NIMs exposure. In vivo magnetic-field-

9

dependent targeting of inhaled NIMs was evaluated in a healthy mouse model. Mice were

10

endotracheally administered fluorescently-labeled NIMs either as a dry powder or a liquid

11

aerosol in the presence of an external magnet placed over the left lung. Quantification of

12

fluorescence and iron showed a significant increase in both fluorescence intensity and iron

13

content to the left magnetized lung. In comparison, we observed decreased targeting of

14

fluorescent nanospheres to the left lung from an aerosolized liquid suspension, due to the

15

dissociation of SPIONs and nanoparticles during pulmonary administration. We conclude that

16

dry powder NIMs maintain the therapeutic cytotoxicity of doxorubicin and can be better

17

targeted to specific regions of the lung, in the presence of a magnetic field, compared to a liquid

18

suspension.

19 20

ACS Paragon Plus Environment

2

Page 3 of 31

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

1

Keywords: Targeted pulmonary delivery; nano-in-microparticles (NIMs); pulmonary

2

chemotherapeutic; aerosolized drug delivery; non-small cell lung cancer; superparamagnetic

3

iron oxide nanoparticles (SPIONs)

4 5 6

Introduction

7

Lung cancer is the second most commonly diagnosed cancer in both men and women, and it has

8

the highest mortality rate compared to all other types of cancer accounting for 26.5% of all

9

cancer deaths1–3. Conventional chemotherapy for lung cancer is administered intravenously and

10

does not distinguish between cancerous and healthy cells. The high concentrations of

11

chemotherapeutic agents that are required for systemic administration often lead to nonspecific

12

adverse effects, while lung tumor microenvironments that are distantly located from capillaries

13

receive sub-therapeutic drug concentrations.

14

Pulmonary drug delivery has been used to treat respiratory diseases such as asthma and

15

microbial infections, as well as systemic diseases4–9. The lungs are well-suited for drug delivery

16

due to their large surface area, thin alveolar epithelium, easily permeable membrane, and

17

extensive vasculature, which allow substantial and rapid drug absorption for increased local and

18

systemic efficacy9. Pulmonary delivery of chemotherapy has been evaluated in human trials for

19

the treatment of lung cancer10–15. A phase I/II study of inhaled doxorubicin combined with

20

systemic platinum-based therapy was administered to twenty-eight patients with advanced non-

21

small cell lung cancer11. However, while the results of this localized delivery were promising, a

22

few patients experienced dose-limiting pulmonary-toxicity. In a preclinical study, Dames and

23

colleagues attempted to minimize toxicity to the whole lung by targeting a therapeutic surrogate

24

to a specific lung lobe in mice using iron oxide nanoparticles in the presence of an external

25

magnetic field16. However, targeting to the magnetized lobe was minimal due to the separation

26

of the drug surrogate and iron oxide nanoparticles during pulmonary administration of the liquid ACS Paragon Plus Environment

3

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

1

Page 4 of 31

suspension.

2

The aim of this study was two-fold, 1) to determine if dry powder nano-in-microparticles

3

(NIMs)17 containing doxorubicin still display therapeutic cytotoxicity after the spray drying

4

process and 2) to evaluate the magnetic-field-dependent targeting of dry powder NIMs

5

administered endotracheally into the lungs of healthy mice (Fig. 1A). Our hypothesis was that

6

the incorporation of a chemotherapeutic agent and iron-oxide nanoparticles in a powder matrix

7

should prevent the separation of nanoparticles and therapeutic observed by Dames et al., while

8

still targeting the drug payload to the magnetized lung region (Fig. 1A)16. This inhalable drug-

9

delivery approach has the potential to reduce toxicity to healthy tissues by targeting drugs

10

directly to specific regions of the lung, thus achieving therapeutic concentration near solid

11

tumors (Fig. 1B)18,19.

12

Experimental Methods

13

Materials

14

Alpha-D-(+)-lactose monohydrate Respitose ML-001 was a gift from DMV-Fonterra

15

Excipients GmbH & Co. KG (Goch, Germany). FluidMAG-UC SPIONs with a hydrodynamic

16

diameter of 50 nm was purchased from Chemicell GmbH (Berlin, Germany). Fluorescent dye-

17

containing NIMs were formulated with Molecular Probes® FluoSpheres® Carboxylate-

18

Modified Nanospheres (F8783, 0.02µm, λex = 660nm and λem = 680nm, Molecular Probes®,

19

Life Technologies, Thermo Fisher Scientific, Inc. Waltham, MA, USA).

20

Formulation and characterization of the NIMs delivery vehicle

21

Spray drying of the NIMs delivery vehicle was performed as described previously17.

22

Briefly, a suspension containing approximately 20% (w/w) SPIONs and either 2.8% (w/w)

23

doxorubicin (Selleck Chemicals, LLC, Houston, TX, USA) (w/w) (for in vitro studies) or 10%

24

(w/w) fluorescent nanospheres (fluorescent drug surrogate for in vivo studies) was spray-dried

25

in MilliQ water and a lactose matrix (2.5% w/v total feed concentration) (Fig. 1A). A B-290 ACS Paragon Plus Environment

4

Page 5 of 31

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

1

mini-spray dryer with a standard two-fluid nozzle (0.7mm diameter) (Büchi Corporation,

2

Flawil, Switzerland), was used to spray dry the suspension with the following parameters: inlet

3

temperature 170 ± 2°C, outlet temperature 103 ± 2°C, aspirator rate 100%, and an atomization

4

air flow rate of 742NL/h. Spray-dried control microparticles were formulated containing a

5

higher concentration of 5% lactose only (w/v) in MilliQ water to keep particle sizing similar.

6 7

Figure 1: Magnetic field-dependent lung targeting. A) Pictorial representation of NIMs dry

8

powder showing lactose (gray), doxorubicin (red), and SPIONs (black)17. B) Scanning electron

9

microscope image of NIMs dry powder. C) Schematic of endotracheal delivery of magnetically-

10

targeted NIMs. D) Orientation of the permanent magnet in thoracotomized mice prior to

11

pulmonary delivery of NIMs. E) Orientation of the trachea, right lung, and left lung prior to

ACS Paragon Plus Environment

5

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 6 of 31

1

imaging. SPIONs are visible in the trachea and left lung (shown by arrows) demonstrating

2

magnetic targeting.

3

Dry powder NIMs particle sizing

4

NIMs geometric particle size was measured using a Mastersizer 3000 analyzer (Malvern

5

Instruments Ltd, Worcestershire, UK) attached to a dry-dispersion accessory device (Aero S,

6

Malvern Instruments). A pressure of 4 bars was used for the venturi dispenser with a feed rate of

7

90%. The refractive index of the sample was taken to be the average of the NIMs components

8

multiplied by the percent (w/w) contained in the formulation: 70% w/w lactose (1.35), 20% w/w

9

SPIONs (2.42), and 10% w/w fluorescent nanospheres (1.52) for a weighted average of 1.58.

10

The results were expressed in terms of Dv50 (volumetric median diameter). All samples were

11

analyzed in triplicate and are expressed as mean ± standard deviation. The aerodynamic

12

diameter of the NIMs was determined using a Next Generation Impactor (NGI) (model 170,

13

MSP Corporation, Shoreview, MN). NIMs were insufflated into the NGI using the DP4 dry

14

powder insufflator™ for rat (Penn Century, Inc., USA). The NGI was operated with a flow rate

15

of 30L/min for 10min. Particle deposition was determined by the gravimetric method.

16

Cell culture

17

Human lung adenocarcinoma A549 cells (ATCC® CCL-185™) (American Type Tissue

18

Culture, Manassas, Virginia, USA) were grown in Ham’s F12K (Kaighn’s) medium and

19

supplemented with fetal bovine serum (FBS), 5% L-glutamine, 3% antimycotic, and 3%

20

antibiotic (Life Technologies, Grand Island, NY, USA).

21

Cytotoxicity studies

22

Confluent cells were exposed to dry powder NIMs containing doxorubicin (D-NIMs)

23

and controls (doxorubicin solution, lactose solution, and SPIONs suspension). A549 cell

24

viability and toxicity were tested using the CytoTox96® Non-Radioactive (3-(4,5-

25

dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium

ACS Paragon Plus Environment

(MTS) 6

Page 7 of 31

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

1

cell proliferation assay and the CytoTox96® Non-Radioactive lactate dehydrogenase (LDH)

2

cytotoxicity assay, respectively. Briefly, D-NIMs were uniformly dispersed in F12K non-

3

supplemented media with FBS at three different doxorubicin concentrations: 0.16µg/mL (low

4

concentration), 1.6µg/mL (medium concentration), and 16µg/mL (high concentration) (referred

5

to henceforth as low, medium, and high dose). The same media was used to generate free

6

doxorubicin solutions with concentrations of 0.3µg/mL (low), 3µg/mL (medium), and 30µg/mL

7

(high). Controls consisting of lactose solution and SPIONs suspension were used at

8

concentrations similar to those present in NIMs, as ratios from the spray dried feed solution

9

were held constant relative to doxorubicin. Prior to exposure, A549 cells were washed with

10

fresh media and 100µL of the suspensions/solutions (D-NIMs; doxorubicin only; lactose;

11

SPIONs).

12

CellTiter 96® Aqueous One Solution Reagent MTS assay (Promega, Madison,

13

Wisconsin, USA) and CytoTox96® Non-Radioactive Assay LDH assay (Promega, Madison,

14

Wisconsin, USA) were performed according to the manufacturer’s instructions. In brief, at time

15

periods of 1, 8, 24, 48 hours post-exposure, 80µL of cell culture supernatant was aspirated from

16

each well and centrifuged at 10,000 rpm for 10 minutes to settle any SPIONs or cells. MTS

17

reagent was mixed with the cell culture supernatant at a ratio of 1:6 (MTS) or 1:2 (LDH),

18

aliquoted into a black optical bottom 96-well plate and incubated at 37°C and 5% CO2 for 3

19

hours (MTS) or 30 min (LDH). For LDH development, 50µL of stop solution was added to each

20

well. Absorbance was measured at a wavelength of 490nm. Results were plotted as absorbance

21

over time with respect to increasing amounts of doxorubicin present in the D-NIMs.

22

In vivo study design

23

Male Balb/c mice (6-8 weeks old; Jackson Laboratory, Sacramento, CA, USA) were

24

used for the in vivo targeting studies. All rodents were housed in a temperature and light cycle

25

controlled facility, and their care followed the guidelines of the National Institutes of Health and

ACS Paragon Plus Environment

7

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

1

an approved Institutional Animal Care and Use Committee protocol (IACUC protocol number

2

11-100747-HSC). Mice were randomly assigned to 5 different groups with n=3 per group: 1)

3

Untreated control mice; 2) Mice administered NIMs dry powders magnetically-targeted to the

4

left lung; 3) Mice administered NIMs dry powders in the absence of magnetic targeting; 4) Mice

5

administered NIMs liquid suspensions magnetically-targeted to the left lung; 5) Mice

6

administered NIMs liquid suspensions in the absence of magnetic targeting. For the in vivo

7

studies, doxorubicin in the NIMs was replaced with fluorescent nanoparticles as a drug

8

surrogate to facilitate imaging.

9

Mice were anesthetized with a standard dose of xylazine/ketamine. A dry powder

10

Insufflator™ for mouse (DP-4-M; Penn-Century Inc., Wyndmoor, PA, USA) was attached to a

11

1mL disposable plastic syringe that was used to endotracheally administer the dry powder

12

NIMs. Briefly, anesthetized mice were placed on their back on a intubation platform (Penn-

13

Century Inc., Wyndmoor, PA, USA) and the tracheal opening was visualized by inserting a

14

small animal laryngoscope (Model LS2; Penn-Century Inc.)20. The insufflator delivery tube was

15

inserted gently into the trachea of the animal, proximal to the carina, until the bend of the

16

delivery tube was positioned at the incisors. Skin over the left lung of the mouse was carefully

17

dissected without exposing the thoracic cavity

18

neodymium−iron−boron (NdFeB) permanent cylindrical magnet (grade N52, 22mm long ×

19

20mm in diameter; Applied Magnets, Plano, TX) was centered 1mm above the left lung to avoid

20

contact with the tissue, with the edge of the magnet perpendicular to the upper region of the left

21

lung (Fig. 1C). Skin removal was performed to decrease the distance between the lung and the

22

magnet, and maximize the magnetic targeting. The orientation of the magnet over the left lung

23

was critical for the NIMs to be sufficiently attracted to the magnetic pole that was created by the

24

externally-applied magnetic field. We had previously characterized the permanent magnet to

25

have a magnetic field of 0.58 Tesla17.

26

16

and a commercially available

Approximately 2.0mg of NIMs were loaded in the insufflator and administered using the 8 ACS Paragon Plus Environment

Page 8 of 31

Page 9 of 31

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

1

plastic syringe with an air volume of 500µL for a total of 10 puffs (10 actuations). Of the 2.0mg

2

loaded in the insufflator, 0.5mg was delivered into the mouse lung based on the gravimetric

3

analysis of the insufflator before and after dosing. A NIMs liquid suspension was administered

4

using the MicroSprayer® aerosolizer (1A-1C; Penn-Century) for mouse and was attached to a

5

hand-operated, high-pressure syringe (FMJ-250; Penn-Century)21. Mice were endotracheally

6

intubated and 50µL of saline containing 0.5mg of the NIMs dry powder (w/v solids; containing

7

equivalent amounts of the fluorescent dye and SPIONs) was administered directly into the

8

airways with the MicroSprayer®. Control mice were treated similarly using the dry powder

9

NIMs and liquid suspension in the absence of magnetic-field-dependent targeting. Mice were

10

sacrificed immediately after pulmonary delivery; lungs and trachea were removed en bloc,

11

separated (Fig. 1D), and fluorescence and iron deposition were quantified in the respective

12

tissues.

13

Fluorescence quantification

14

NIMs dry powder and NIMs suspension were administered endotracheally and targeted

15

to the left lung in the presence of a permanent magnet. Individual lung lobes and trachea were

16

excised and imaged immediately for fluorescence (λex = 660nm and λem = 680nm) with an

17

exposure time of 10 seconds using a Caliper IVIS Lumina II imaging system (Caliper Life

18

Sciences). A region of interest (ROI) was drawn around the trachea, left lung, right lung, and

19

background. The ROI area was kept constant at 2.5 cm2 and fluorescence was expressed as units

20

of average radiance efficiency (RE) (p/sec/cm2/sr)/(µW/cm2)22.

21

Equation 1. Fluorescence calculations

22

% Targeting =

23

Targeting Efficiency L:R Ratio = %

24 25

  

× 100 %   

Percent targeting was calculated by determining ratios of the fluorescence intensity (FI individual)

of ROIs from individual tissue (e.g. left lung) in the numerator, divided by the sum of ACS Paragon Plus Environment

9

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 10 of 31

1

fluorescence intensity (FI

2

denominator (Equation 1), and multiplied by 100. Targeting efficiency L:R ratio was calculated

3

using the percent targeting of the left lung in the numerator, divided by the percent targeting of

4

the right lung in the denominator.

sum)

of the three ROI areas (trachea, left lung, and right lung) in the

5 6

Iron quantification

7

Inductively coupled plasma optical emission spectroscopy (ICP-OES) was used to

8

measure ferrous iron content in the mouse lungs and trachea. Briefly, tissue was digested with

9

0.5mL nitric acid (HNO3) at 105 °C for 1 hour and vortexed until completely dissolved

10

(protocol modified from Niazi et al.23). After cooling, the samples were brought up to a known

11

volume with MilliQ water. Samples were analyzed with a PerkinElmer Optima 4300 DV ICP-

12

OES. The recommended wavelength for iron was used, Fe (II)-λ-259.939 nm. A five-point

13

calibration standard range was used that ranged from 0.1 ppm to 25 ppm (mg/L). Endogenous

14

iron was analyzed from the lungs of untreated mice (n = 3), and the average of these values was

15

subtracted from the treated mice. Calibration and instrument verification samples were

16

incorporated before and after analyzing the samples, as well as periodically throughout the

17

measurements.

18

Equation 2. Iron calculations

19

% Targeting =

20

Targeting Efficiency L:R Ratio = %[]

[] [] 

× 100 %[]   

21

Targeting efficiency ratio was calculated by determining ratios of the iron content from

22

individual tissue (e.g. left lung) in the numerator, divided by the sum of the total iron content for

23

all three tissues (trachea, left lung, and right lung) in the denominator (Equation 2), and

24

multiplied by 100. Targeting efficiency L:R ratio was calculated using the percent targeting of

ACS Paragon Plus Environment

10

Page 11 of 31

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

1

the left lung in the numerator, divided by the percent targeting of the right lung in the

2

denominator.

3

Dye and iron targeting corroboration

4

Equation 3. Fluorescence and iron differentials

5

Fluorescence differential = 

6

Iron differential = [)*+]

!" #$

!" #$

− &'$(" #$

− [)*+]&'$(" #$

7

Targeting of dye and iron was corroborated by calculating the differential between the

8

fluorescence (relative efficiency) or iron concentrations in left lung compared to the right

9

(Equation 3).

10 11

Statistical analysis

12

All statistical analysis was performed using GraphPad Prism statistical software

13

(GraphPad Software, San Diego, CA). Paired t-tests with Bonferroni correction were used to

14

compare two or more groups of independent data. The difference between variants was

15

considered significant if p < .05. For comparisons of more than two groups an ordinary one-way

16

ANOVA was used to determine significance, with Tukey’s multiple comparisons and a Brown-

17

Forsythe test used to compare variances. If more than two variables were taken into account, a

18

two-way ANOVA was employed with Sidak’s multiple comparison. A post hoc power

19

calculation was performed using the free program G*power (Dusseldorf, Germany)24,25. For in

20

vivo animal experiments with equivalent sample sizes and an effect size of n = 3, the power was

21

calculated to be > 0.95.

22

Results

23

Dry powder NIMs formulation and characterization

24

Dry powder NIMs for pulmonary administration were prepared as described previously

25

using lactose as the bulking agent17,26. NIMs were characterized for aerodynamic and geometric

ACS Paragon Plus Environment

11

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 12 of 31

1

diameters, surface charge of the SPIONs, morphology, magnetic behavior, iron content, and

2

drug loading (Table 1)17. For this study, doxorubicin, a common chemotherapeutic agent with

3

demonstrated activity against lung cancer, was used. NIMs with doxorubicin will be referred to

4

as D-NIMs. Table 1. Characterization of SPIONs and NIMs* SPIONs average radius density zeta potential

NIMs

56 ± 6nm 2.5 g/cm3 -49 mV

Concentration (w/w) 1.6% Doxorubicin 10.6% Fe3O4 Laser Diffraction 1.6µm Next generation Impaction MMAD‡ 3.27µm FPF‡ > 90% GSD‡ ± 1.69

*

For more details on the NIMs powder preparation and characterization consult McBride et al [17] For NIMs dry powder ‡ Mass Median Aerodynamic Diameter (MMAD), Fine Particle Fraction (FPF), Geometric Standard Deviation (GSD) †

5 6

In vitro evaluation of chemotherapeutic efficacy

7

To assess whether the doxorubicin in D-NIMs retained its cytotoxicity (and therefore its

8

therapeutic effect) after spray drying, a MTS assay was utilized to measure cell viability of lung

9

adenocarcinoma cells after D-NIMs exposure. We hypothesized that D-NIMs would quickly

10

release the doxorubicin and would have similar or increased cytotoxicity compared to free

11

doxorubicin in A549 cells. The left panel of Figure 2 compares A549 cells treated with D-

12

NIMs, as well as controls consisting of spray-dried lactose, SPIONs alone, and free doxorubicin

13

solution. A549 cells were exposed to low, medium, and high doses of free doxorubicin solution

14

or D-NIMs for a total of 48 hours (Fig. 2). It is important to note that the concentration of

15

doxorubicin was approximately doubled in the free doxorubicin solution control group

16

compared to the D-NIMs dry powder group at all three doses. The low concentration of free

17

doxorubicin solution (0.03µg) or D-NIMs (0.016µg) showed minimal cytotoxicity, and cells

18

were as viable as untreated A549 cells after 48 hours of exposure (Fig. 2A, MTS panel).

19

However, medium and high doses of doxorubicin solution and D-NIMs were toxic to 12 ACS Paragon Plus Environment

Page 13 of 31

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

1

adenocarcinoma cells compared to SPION and lactose controls. Importantly, at both medium

2

and high doses, no significant differences were seen between the free doxorubicin and D-NIMs

3

groups ((67±1.67)% and (58±0.51)% respectively), despite D-NIMs containing doxorubicin at

4

nearly half the concentration of the free doxorubicin group. (Fig 2C, MTS panel).

5 6

Figure 2. MTS viability and LDH cytotoxicity assay of A549 cells exposed to doxorubicin-

7

loaded NIMs. MTS (left panel) and LDH (right panel) assay measuring viability and

8

cytotoxicity, respectively, of A549 lung adenocarcinoma cells after 48 hours of exposure to A) 13 ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 14 of 31

1

low dose of D-NIMs compared to free doxorubicin (1µg NIMs containing 0.016µg doxorubicin

2

compared to 0.03µg free doxorubicin), B) medium dose of D-NIMs compared to free

3

doxorubicin (10µg NIMs containing 0.16µg doxorubicin compared to 0.3µg free doxorubicin),

4

and C) high dose of D-NIMs compared to free doxorubicin (100µg NIMs containing 1.6µg

5

doxorubicin compared to 3.0µg free doxorubicin). Free doxorubicin control (dark gray), Spray-

6

dried lactose control (light gray), SPIONs control (dashed gray), D-NIMs (black); for both MTS

7

and LDH panels. Lactose and SPIONs controls were used at the ratios from the spray dried feed

8

solution and were held constant relative to doxorubicin. A two-way ANOVA with Sidak’s

9

multiple comparison test was used to determine statistical significance. *p