hosted by · 2016-01-05 · *please mount your poster no later than 8:30 on october 1, 2015....

33
October 1-3 , 2015 Northeastern University, Boston, Massachusetts, USA HOSTED BY:

Upload: others

Post on 23-Jul-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: HOSTED BY · 2016-01-05 · *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00

October 1-3 , 2015 Northeastern University, Boston, Massachusetts, USA

HOSTED BY:

Page 2: HOSTED BY · 2016-01-05 · *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00

Symposium Chair Vladimir Torchilin, Ph.D., D.Sc. University Distinguished Professor, Director of Center for Pharmaceutical Biotechnology and Nanomedicine, Department of Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA

Thursday, October 1, 9:00 – 9:15

International Advisory Board: J. Irache (Spain); P. Couvreur (France); T. Webster (USA); C-M. Lehr (Germany); D. Poncelet (France); E. Fattal (France); M. Alonso (Spain); H. Smyth (USA); P. Mainsent (France); G. Colombo (Italy); Y. Capan (Turkey); J. Kreuter (Germany); P. Caliceti (Italy); J-C. Leroux (Switzerland); A. Goepferich (Germany); K. Park (USA); M. Amiji (USA); C. Alvarez Lorenzo (Spain); A. Bernkop-Schnurch (Austria); J.P. Benoit (France); A. Almeida (Portugal)

Local Organizing Committee: V. Torchilin, T. Webster, M. Amiji, T. Levchenko, M. Sheynina The Best Poster Committee: S. Salmaso, T. Minko, T. Konri

CONFERENCE SUPPORTERS

Our sincere thanks to the sponsors of this year’s conference:

Page 3: HOSTED BY · 2016-01-05 · *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00

IMS2015 Boston Northeastern University, Boston MA October 1-3, 2015

1 All talks, poster sessions, breakfasts and coffee breaks will take place in the Curry Student Center Ballroom on the 2nd (Building #50 on map) unless otherwise noted. Lunch will be served in the McLeod Suites on the 3rd floor of the building.

3 | P a g e

AGENDA1

Thursday, October 1, 2015 7:30 - 9:00 Registration/Continental Breakfast *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00 on October 1 and from 13.00 to 14.30 on October 2 to interact with poster viewers, judges, answer questions, and/or provide any clarifications regarding the work presented. The 6 best posters will be selected for 10 minute presentations (up to 5 slides) on October 3, 2015. Announcement about selected presentations will be made before Session 5 on October 2. One first place, two second and 3 third will be announced before the final general discussion and certificate will be sent to awardees by mail.

9:00 – 9:15 Introductory Remarks J. Reynolds, Professor and Interim Dean, Bouvé College of Health Sciences, Northeastern University, Boston, MA V. Torchilin, University Distinguished Professor, Director, Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA

9:15 – 10:00 Plenary Talk 1 F. Szoka, “On the road to clinical use of microencapsulated systems for drug and gene delivery: problems and hurdles”

10:00 – 11:00 Session 1 “Lipid-based systems for encapsulation” Chair – G. Borchard 10:00-10:30 – T. Minko, “Lipid-based microencapsulation for drug and gene delivery in lung diseases.” 10:30-11:00 – K. Wasan, “Lipid-based encapsulation for infectious diseases.”

11:00 – 11:30 Coffee Break

11:30 – 12:00 Session 1 “Lipid-based systems for encapsulation” continued Chair – G. Borchard H. Harashima, “Multifunctional lipid-based systems for drug and gene delivery.”

12:00 – 13:00 Session 2 “Microencapsulation for imaging” Chair – L. Zhu 12:00-12:30 – A. Bogdanov, “Encapsulated agents for Magnetic Resonance Imaging.” 12:30-13:00 – C. Allen, “The impact of heterogeneity in tumor microenvironment on treatment

efficacy.”

13:00 – 15:00 Lunch and Poster Viewing

15:00 – 15:30 Session 2 “Microencapsulation for imaging” continued Chair – L. Zhu 15:00-15:30 – V. Zharov, “Photothermal and photoacoustic visualization of drug carriers and nanoparticles in the blood.”

15:30 – 16:00 Session 3 “Microencapsulation for consumer products”

Page 4: HOSTED BY · 2016-01-05 · *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00

IMS2015 Boston Northeastern University, Boston MA October 1-3, 2015

1 All talks, poster sessions, breakfasts and coffee breaks will take place in the Curry Student Center Ballroom on the 2nd (Building #50 on map) unless otherwise noted. Lunch will be served in the McLeod Suites on the 3rd floor of the building.

4 | P a g e

Chair – S. Salmaso 15:30-16:00 – M. Rosenberg, “Microencapsulation for food products.”

16:00 – 17:00 Poster Viewing

19:00 Speakers’ Dinner – 406 Stuart Street, Boston, MA (by invitation only)

Friday, October 2, 2015 9:00 – 10:00 Plenary Talk 2

P. Hammond. “Materials for layer-by-layer microencapsulation.”

10:00 – 11:00 Session 4 “Translational Microencapsulation” Chair – M. Alonso 10:00-10:30 – R. Gray, “Microfluidic methods for precise, high throughput Microencapsulation.”

10:30-11:00 – T. Konry, “Lab on a chip microencapsulation technique for cellular spheroid generation and dynamic drug evaluation analysis.”

11:00 – 11:30 Coffee Break

11:30 – 12:00

Session 4 “Translational Microencapsulation” continued Chair – M. Alonso 11:30-12:00 – A. Wang, “Clinical application of encapsulated platinum preparations.”

12:00 – 14:30 Lunch and Poster Viewing

14:30 – 16:30 Session 5 “New Concepts in Microencapsulation” Chair – M. Rosenberg 14:30-15:00 – M. Alonso, “Multilayer polymer nanocapsules as drug delivery carriers.” 15:00-15:30 – L. Zhu, “Stimuli-sensitive microencapsulation.”

15:30-16:00 – Z. Medarova, “Encapsulation in design of miRNA targeted drugs.”

16:00-16:30 – C. Palivan, “Protein-polymer nanoreactors as artificial organelles.”

19:00 Conference Dinner - Odyssey Boston, 60 Rowes Wharf, Boston, MA 02110 (Bus transportation provided from campus at Ruggles circle at 17:30. Ship boarding begins at 18.00)

Saturday, October 3, 2015 9:00 – 10:30 Session 6 “New materials for microencapsulation”

Chair – V. Torchilin 9:00-9:30 – T. Webster, “New materials for microencapsulation.”

9:30-10:00 – S. Salmaso. “Multimodal microencapsulated system for logical delivery of anticancer drugs.”

10:00-10:30 – G. Borchard. “Layer-by-layer enzyme microencapsulation.”

10:00 - 10:30 Coffee Break

Page 5: HOSTED BY · 2016-01-05 · *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00

IMS2015 Boston Northeastern University, Boston MA October 1-3, 2015

1 All talks, poster sessions, breakfasts and coffee breaks will take place in the Curry Student Center Ballroom on the 2nd (Building #50 on map) unless otherwise noted. Lunch will be served in the McLeod Suites on the 3rd floor of the building.

5 | P a g e

PLENARY SPEAKERS

Francis Szoka, Ph.D. Professor, Department of Bioengineering and Therapeutic Sciences, UCSF School of Pharmacy, San Francisco CA, USA Thursday, October 1, 9:15 – 10:00

Paula T. Hammond, Ph.D. David H. Koch Professor of Engineering, Head of the Department of Chemical Engineering, MIT, Boston, MA, USA Friday, October 2, 9:15 – 10:00

11:00 – 12:30 Session 7 “Best poster presentations” Chair – S. Salmaso Short presentation of selected posters

12:30 – 13:30 Final General Discussion

13:30 Symposium Adjourn

Page 6: HOSTED BY · 2016-01-05 · *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00

IMS2015 Boston Northeastern University, Boston MA October 1-3, 2015

6 | P a g e

INVITED SPEAKERS

Tamara Minko, Ph.D. Distinguished Professor and Chair, Department of Pharmaceutics, Rutgers, The State University of New Jersey, NJ, USA Thursday, October 1, 10:00- 10:30

Kishor Wasan, Ph.D. Professor and Dean, College of Pharmacy and Nutrition, University of Saskatchewan, Canada Thursday, October 1, 10:30 – 11:00

Hideyoshi Harashima, Ph.D. Professor and Chair, Laboratory of Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Japan Thursday, October 1, 11:30 – 12:00

Alexei Bogdanov, Ph.D. Professor, University of Massachusetts Medical School, Amherst, MA, USA Thursday, October 1, 12:00 – 12:30

Page 7: HOSTED BY · 2016-01-05 · *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00

IMS2015 Boston Northeastern University, Boston MA October 1-3, 2015

7 | P a g e

Christine Allen, Ph.D. Professor, Associate Dean, Graduate Education, Leslie Dan Faculty of Pharmacy, University of Toronto, Canada Thursday, October 1, 12:30 – 13:00

Vladimir Zharov, Ph.D., D.Sc. Professor, Department of Otolaryngology; Director, Arkansas Nanomedicine Center, University of Arkansas for Medical Sciences, AR, USA Thursday, October 1, 15:00 – 15:30

Moshe Rosenberg, Ph.D., D. Sc. Professor and Specialist, Department of Food Science and Technology, University of California, Davis, CA, USA Thursday, October 1, 15:30 – 16:00

Richard Gray, MA. Director of Blacktrace Holdings, Regional Director of the Group’s US subsidiaries, Boston, MA, USA Friday, October 2, 10:00 – 10:30

Page 8: HOSTED BY · 2016-01-05 · *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00

IMS2015 Boston Northeastern University, Boston MA October 1-3, 2015

8 | P a g e

Tania Konry, Ph.D. Assistant Professor, Department of Pharmaceutical Sciences Bouvé Collge of Health Sciences, Northeastern University, Boston, MA, USA Friday, October 2, 10:30 – 11:00

Andrew Wang, M.D. Associate Professor and Director of Clinical and Translational Research, Department of Radiation Oncology, University of North Carolina, Chapel Hill, USA Friday, October 2, 11:30 – 12:00

Maria Jose Alonso, Ph.D. Professor, Biopharmaceutics and Pharmaceutical Technology, University of Santiago de Compostela, Santiago de Compostela, Spain Friday, October 2, 14:30 – 15:00

Lin Zhu, Ph.D. Assistant Professor, Irma Lerma Rangel College of Pharmacy, Texas A&M Health Science Center, Kingsville, TX, USA Friday, October 2, 15:00 – 15:30

Page 9: HOSTED BY · 2016-01-05 · *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00

IMS2015 Boston Northeastern University, Boston MA October 1-3, 2015

9 | P a g e

Zdravka Medarova, Ph.D. Assistant Professor, Radiology, Harvard Medical School; Massachusetts General Hospital, Boston, MA, USA Friday, October 2, 15:30 – 16:00

Cornelia Palivan, Ph.D. Professor, Department of Chemistry, University of Basel, Switzerland Friday, October 2, 16:00 – 16:30

Thomas Webster, Ph.D. Professor and Department Chair, Chemical Engineering, Northeastern University, Boston, MA, USA Saturday, October 3, 9:00 – 9:30

Stefano Salmaso, Ph.D. Associate Professor, Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Italy Saturday, October 3, 9:30 – 10:00

Page 10: HOSTED BY · 2016-01-05 · *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00

IMS2015 Boston Northeastern University, Boston MA October 1-3, 2015

10 | P a g e

Gerrit Borchard, Ph.D. Professor, University of Geneva, Geneva-Lausanne School of Pharmacy, Switzerland Saturday, October 3, 10:00 – 10:30

POSTER PRESENTERS

Poster # Presenter’s name

Organization, country Abstract Title

1 Simone Aleandri University of Zurich, Switzerland

Light-responsive lyotropic liquid crystal mesophases: triggered release of guest molecules

2 Barbara Blanco Fernandez

Michigan State University, USA

Glucose cryoprotectant hinders nanoparticle responsiveness to glutathione

3 Navroz Boghani Mondelez International, USA Microencapsulation for sustained release in chewing gums

4 Luis Braz University of Algarve, Portugal

Toxicological evaluation of locust bean gum/chitosan nanoparticles for an application in respiratory delivery

5 Ana Cadete Pires University of Angers, France and University of Santiago de Compostela, Spain

The potential of hydrophobically modified hyaluronic acid nanocapsules for targeted cancer therapy

6 Andrea Cerciello University of Salerno, Italy Prilling as a versatile technique for the development of controlled delivery systems containing anti-inflammatory drugs

7 Gaia Colombo University of Ferrara, Italy Thalidomide complexation by β-cyclodextrins in new nasal powder formulations for hereditary hemorrhagic telangiectasia

8 Ludmylla Cunha University of Algarve, Portugal

Fucoidan microparticles as inhalable antibiotic carriers

9 Felicetta De Cicco University of Salerno, Italy Combination of co-axial prilling and supercritical drying for the formulation of doxycycline particles for wound healing.

10 Connor Dennis Orbis Biosciences, Inc, USA Taste-masked Powders for Pediatric-centric Dispersed Dosage Formats

11 Pranali Deshpande

Northeastern University, USA

Dual-functional liposomes for effective cancer cell targeting and Improved Intracellular delivery

Page 11: HOSTED BY · 2016-01-05 · *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00

IMS2015 Boston Northeastern University, Boston MA October 1-3, 2015

11 | P a g e

12 Yuancai Dong Institute of Chemical & Engineering Science, Singapore

Hot-melt Extrusion Microencapsulation of Polyphenols for Taste-masking

13 Anna Maria Fadda University of Cagliari, Italy Delivery of liposomes across the skin using needle-free liquid jet injectors

14 Ada Ferri Politecnico di Torino, Italy Encapsulation of active principles in PCL for knitted fabrics functionalization

15 Ana Grenha University of Algarve, Portugal

Unveiling a role for polysaccharides on macrophage targeting in tuberculosis therapy

16 Juan Irache University of Navarra, Italy Polymeric nanoparticles for the oral delivery of camptothecin

17 Aditi Jhaveri Northeastern University, USA Resveratrol-loaded liposomes: A promising nanomedicine for malignant gliomas

18 Beatriz Miranda Harvard University, USA; University of São Paulo, Brazil,

Mucus-penetrating, pH sensitive polymersomes

19 Alexander Moncion

University of Michigan, USA Perfluorocarbon emulsions as ultrasound-mediated delivery vehicles in fibrin scaffolds.

20 Armin Mooranian Curtin University, Australia Novel multicompartmental microcapsules for the oraltargeted delivery of a potential antidiabetic drug, probucol, using bile acid-based formulation: anti-hyperglycaemic and anti-inflammatory effects in an animal model of Type 2 diabetes mellitus

21 Bhushan Pattni Northeastern University, USA

Targeted delivery of PEG-PE-based micelles encapsulating a novel pro-apoptotic drug, NCL-240, in 3D cancer cell spheroids

22 Can Sarisozen Northeastern University, USA

Stimuli-sensitive siRNA delivery by hypoxia-targeted nanoformulation

23 Marco Soto Pontifical Catholic University of Chile

Amphipathic tri-block co-polymers interaction with DPPC large unilamellar vesicles

24 Krzysztof Szczepanowicz

Jerzy Haber Institute of Catalysis and Surface Chemistry PAS, Poland

Polyelectrolyte nanocapsules as a promising drug delivery system

ATTENDEES

James Aborn Hideyoshi Harashima

E Ink Corporation, USA Hokkaido University, Japan

Simone Aleandri Juan M. Irache

University of Zurich, Switzerland University of Navarra, Spain

Christine Allen Tania Konry

University of Toronto, Canada Northeastern University, USA

Maria Jose Alonso Tatyana Levchenko

University of Santiago de Compostela, Spain Northeastern University, USA

Erhan Altinoglu Francesco Lai

Page 12: HOSTED BY · 2016-01-05 · *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00

IMS2015 Boston Northeastern University, Boston MA October 1-3, 2015

12 | P a g e

Novartis, USA University of Cagliari, Italy

Mansoor Amiji Lynne McCullough

Northeastern University, USA E-Ink Corporation, USA

Brian Ayling Zdravka Medarova

Microfluidics Corporation, USA Massachusetts General Hospital, USA

Barbara Blanco Fernandez Tamara Minko

Michigan State University, USA Rutgers State University, USA

Alexei Bogdanov Beatriz Miranda

University of Massachusetts Amherst, USA Harvard University, USA

Navroz Boghani Alexander Moncion Baez

Mondelez International, USA University of Michigan, USA

Gerrit Borchard Armin Mooranian

Geneva-Lausanne School of Pharmacy, Switzerland Curtin University, Australia

Luis Braz Christine Newbold

University of Algarve, Portugal Microfluidics Corporation, USA

Di Bush James Oxley

Avanti Polar Lipids, Inc., USA Southwest Research Institute, USA

Ana Cadete Pires Cornelia Palivan

University of Angers, France University of Basel, Switzerland

Andrea Cercielli Moshe Rosenberg

University of Salerno, Italy University of California, Davis, USA

Gaia Colombo Stefano Salmaso

University of Ferrara, Italy University of Padova, Italy

Ludmylla Cunha Chiara Sinico

University of Algarve, Portugal University of Cagliari, Italy

Felicetta De Cicco Megan Sonnenburg

University of Salerno, Italy E Ink Corporation, USA

Pasquale Del Gaudio Marco Soto-Arriaza

University of Salerno, Italy Pontificia Universidad Católica de Chile, Chile

Connor Dennis Yang Su

Orbis Biosciences, Inc., USA Microfluidics Coporation, USA

Wenyu Dong Krzysztof Szczepanowicz

Teva Pharmachemie, Netherlands Jerzy Haber Institue of Catalysis and Surface Chem, Poland

Yuancai Dong Francis Szoka

Institute of Chemical & Engineering Science, Singapore University of California, San Francisco, USA

Mario Fabiilli Ronald Versic

University of Michigan, USA Ronald T. Dodge Co., USA

Anna Maria Fadda Andrew Wang

University of Cagliari, Italy University of North Carolina Chapel Hill, USA

Ada Ferri Kishor Wasan

Politecnico di Torino, Italy University of Saskatchewan, Canada

Garret Figuly Thomas Webster

DuPont, USA Northeastern University, USA

Richard Gray Cuie Yan

Dolomite Microfluidics, United Kingdom PepsiCo, USA

Page 13: HOSTED BY · 2016-01-05 · *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00

IMS2015 Boston Northeastern University, Boston MA October 1-3, 2015

13 | P a g e

Ana Grenha Yuxian "Ashley" Zhang

University of Algarve, Portugal Microfluidics Corporation, USA

Paula Hammond Vladimir Zharov

Massachusetts Insitute of Technology, USA University of Arkansas for Medical Sciences, USA

Hiroki Haniuda Lin Zhu

Kyowa Hakko Kirin Co., Ltd, Japan Texas A & M Health Science Center, USA

POSTER ABSTRACTS (1) Light-responsive lyotropic liquid crystal mesophases: triggered

release of guest molecules

S. Aleandria, C. Spezialeb, R. Mezzengab and E. M. Landaua

a Department of Chemistry, University of Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland. b Department of Health Science & Technology, ETH Zurich, Schmelzbergstrasse 9, LFO, E23 8092 Zürich, Switzerland

Nanocarriers offer many advantages in drug delivery compared to conventional formulation methods.1, 2 With appropriate molecular design it is possible to affect triggered drug release in order to achieve better temporal and spatial control of the delivery of therapeutic agents, thereby obtaining a higher local concentration.3 In the field of controlled release, it is possible to obtain drug release in response to various internal and external stimuli, such as pH,4 temperature,3 ultrasound,5 and light.6 An ideal drug delivery system should be biodegradable, biocompatible, incorporate the active agent without loss or alteration of its activity, and provide an efficient and controlled delivery mechanism to the specific location in vivo.7 Lipidic mesophases (LMPs) constitute alternative delivery systems that can be triggered by external stimuli.7 One of the most commonly used lipids for the formation of LMPs is monoolein (MO), which exhibits 1-, 2- and 3-dimensional self-assembled mesophases in the presence of water, corresponding to the hexagonal (H), lamellar (L), and lipidic cubic phases (LCPs) We present novel light-responsive functional lyotropic liquid crystal system using host-guest lipidic mesophases (LMPs). 6 These consist of the neutral host lipids monoolein (MO) and oleic acid (OA), and a small amount of the designed light responsive guest lipids 1 and 2. The effectiveness of the investigated biomaterials in release and retention of embedded dye molecules in single step, as well as sequential light-triggering is demonstrated, thereby achieving exquisite temporal, spatial and dosage control of the release. This opens up the possibility of using such designed lipidic mesophase materials as effective matrices in therapy, when a continuous release of active drugs might be toxic. Significantly, because of the ability of LMPs to incorporate molecules of any polarity or charge, the strategy presented here is general, i.e. molecule independent, and shows great promise for various biomedical applications.

References:

Page 14: HOSTED BY · 2016-01-05 · *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00

IMS2015 Boston Northeastern University, Boston MA October 1-3, 2015

14 | P a g e

1. Lapteva, M. et al. Microstructured bicontinuous phase formulations: their characterization and application in dermal and transdermal drug delivery. Expert Opin Drug Del, 10: 1043-1059, 2013. 2. Yang, J. et al. Stimuli-responsive drug delivery systems. Adv Drug Deliv Rev, 64 : 965-966, 2012.

Page 15: HOSTED BY · 2016-01-05 · *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00

15 | P a g e

3. Fong, W.-K. et al. Stimuli responsive liquid crystals provide 'on-demand' drug delivery in vitro and in vivo. J. Controll Release 135: 218-226, 2009. 4. Negrini, R. et al. pH-Responsive Lyotropic Liquid Crystals for Controlled Drug Delivery. Langmuir 27: 5296-5303, 2011. 5. Cravotto, G. et al. Molecular self-assembly and patterning induced by sound waves. The case of gelation. Chem Soc Rev 2009, 38 (9): 2684-2697, 2009. 6. Aleandri, S, et al. Design of light-triggered lyotropic liquid crystal mesophases and their application as molecular switches in “on demand” release. Langmuir 31 (25): 6981–6987, 2015. 7. Brambilla, D. et al. Breakthrough discoveries in drug delivery technologies: the next 30 years. J. Controll Release, 190: 9-14. 2014.

(2) Glucose cryoprotectant hinders nanoparticle responsiveness to glutathione

Bárbara Blanco-Fernández a, Manuela Curcio a,b, Angel Concheiro a, Francesco Puoci b, Carmen Alvarez-Lorenzo a

a Dept. Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Santiago de Compostela, 15782-Santiago de Compostela (Spain). b University of Calabria, Department of Pharmaceutical Sciences, Edificio Polifunzionale,

Arcavacata di Rende (CS), 87036 Italy.(e-mail: [email protected])

Introduction. Cryoprotectans are frequently required to guarantee the stability of nanoparticles (NPs) systems and ensure their reconstitution; glucose (5-10% wt.) being one of the most widely used. However, certain sugars used as cryoprotectants have been recently shown to negatively impact the cellular uptake of NPs [1]. Moreover, under diabetic conditions glucose can react in vivo with glutathione (GSH)/glutathione disulfide (GSSG) leading to Amadori products, which in turn lowers GSH levels [2]. One can hypothesize that, if locally administered (i.e., intratumorally), glucose-protected NPs may temporally lead to glucose levels that may inactivate the GSH stimulus. Aim. To elucidate the effect of glucose as cryoprotectant on GSH-responsiveness of polysaccharide NPs. Experimental. NPs were prepared via complex coacervation of a chitosan thiomer and chondroitin sulfate or dermatan sulfate, and then, cross-linked with N,N’-bis(acryloyl)cystamine. The obtained NPs were freeze-dried using glucose as cryoprotectant (0.5 and 5.0 % wt) and, subsequently, loaded with methotrexate (MTX). Drug release experiments were conducted in reducing media mimicking the extra- and intracellular spaces. Cellular uptake and antitumoral efficiency was verified against two different cancer cell lines (HeLa and CHO-K1) [3]. Results. Freeze-drying with 0.5% glucose led to NPs that exhibited GSH-dependent release of MTX. Oppositely, those freeze-dried with 5.0% glucose did not show GSH-responsiveness. Moreover, confocal microscopy studies revealed that cellular uptake of NPs freeze-dried with 5.0% glucose occurred to a less extent. Conclusions. The concentration of glucose or any other reducing sugar as cryoprotectant should be considered during the development of GSH-sensitive systems. References: 1. K.S. Tang et al. The effect of cryoprotection on the use of PLGA encapsulated iron oxide nanoparticles for magnetic cell labeling. Nanotechnology, 24:125101, 2013. 2. I. Jeric and S. Horvat. Screening for glucose-triggered modifications of glutathione. J. Pept. Sci. 15:540–547, 2009. 3. M. Curcio et al. Glucose cryoprotectant affects glutathione-responsive antitumor drug release from polysaccharide nanoparticles. Eur. J. Pharm. Biopharm. 2015; doi: 10.1016/j.ejpb.2015.04.010.

Page 16: HOSTED BY · 2016-01-05 · *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00

16 | P a g e

(3) Microencapsulation for sustained release in chewing gums

Navroz Boghani Ph.D.

Research Principal, Mondelēz International

Although encapsulations are used in a wide variety of industries, identifying encapsulation systems that are resistant to mastication, compatible with gum base, provide controlled release of ingredients during chewing, and utilize food approved or potentially approvable ingredients is challenging. The pharmaceutical industry utilizes many encapsulations for drugs – however these focus on providing stability during shelf life and on controlled release during digestion in gastrointestinal track. Further, the encapsulation materials are generally not food approved and would break down quickly during chewing. Also there are several encapsulation technologies utilized in food and agricultural industries for shelf life stability and controlled release. Although these encapsulations work satisfactorily for those applications, they do not withstand the chewing process in gums and lose their efficacy due to break down during chewing. A patent protected, high strength long lasting encapsulation, has been developed using food approved ingredients. The technology uses a unique formula with a special grade Polyvinylacetate polymer and process technology. Analytical results from in-vitro chew-out release, from chewing gum containing the encapsulated sweeteners, will be presented. The analytical release correlates well with sensory testing.

(4) Toxicological evaluation of locust bean gum/chitosan nanoparticles for an application in respiratory delivery

L Braz1,2; S Rodrigues1, AM Rosa da Costa2, A Grenha1

1CBMR – Centre for Biomedical Research, Drug Delivery Laboratory; 2Center for Research in Chemistry of Algarve; University of Algarve, Campus de Gambelas, Faro, Portugal

The use of carriers to mediate the delivery of biomolecules through the most varied routes has been proposed consistently, in line with the need to provide in vivo protection and, when applicable, to improve mucosal contact. Polymeric nanoparticles have been proposed as vehicles, with natural polymers assuming a prominent role as matrix materials, owing to their propensity for biocompatibility and biodegradability, along with their structural flexibility.1 A previous work demonstrated the ability of sulfated locust bean gum/chitosan (SLBG/CS) nanoparticles to associate different protein-based molecules.2 In the present work, a toxicological evaluation of these carriers regarding a pulmonary application is provided. A home-made sulfate derivative of LBG was complexed with CS (SLBG/CS = 2/1), forming nanoparticles by polyelectrolyte complexation. After centrifugation (16000 g, 30 min, 15 ºC), nanoparticle sediments were resuspended in 200 μL of ultrapure water. Morphology (transmission electron microscopy), size and zeta potential (dynamic light scattering) were characterized. Nanoparticle biocompatibility was evaluated in two respiratory cell lines (A549 and Calu-3), by the MTT assay (metabolic activity), LDH release (cell membrane integrity) and quantification of released IL-6 and IL-8 (inflammatory response). Cells were exposed to different concentrations of nanoparticles (0.1, 0.5, 1 mg/mL), for 3h and 24h. Nanoparticles with spherical shape, evidencing a size around 180 nm and zeta potential of +14 mV were obtained. The MTT assay revealed absence of toxicity in the range of tested concentration, with cell viabilities above 90% in all cases. LDH release upon 24h exposure to nanoparticles was ~25% higher than that of the control. Finally, a slight inflammatory response was found to occur, with release of IL-8 and IL-6 around 145-150% comparing with the 100% considered for the negative control. In this assay, LPS was used as positive control, inducing 220-350% release. More tests are therefore needed to conclude on the biocompatibility of the formulation. Funding from Portuguese Foundation for Science and Technology (PTDC/SAU-FCF/100291/2008, PTDC/DTP-FTO/0094/2012, UID/BIM/04773/2013) is acknowledged. S Rodrigues acknowledges PhD scholarship (SFRH/BD/52426/2013). References: 1. Anwunobi and Emeje. Recent applications of natural polymers in nanodrug delivery. J Nanomed Nanotechnol, S4:2-7, 2011. 2. Braz et al. Locust Bean Gum-based nanoparticles as antigens carriers. Biomed Biopharm Res, 2(10): 281, 2013.

Page 17: HOSTED BY · 2016-01-05 · *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00

17 | P a g e

(5) The potential of hydrophobically modified hyaluronic acid nanocapsules for targeted cancer therapy

Ana Cadete1, 5, Ana Olivera1, Angela Molina2, Lídia Gonçalves3, Magnus Besev4, Pradeep Dhal4, Gema Moreno3, Guillaume Bastiat5, JP Benoit5, Dolores Torres1, MJ Alonso1

1Center for Research in Molecular Medicine and Chronic Diseases (CIMUS) and Dept. Pharmacy, University of Santiago de Compostela, Spain; 2 Dept. Biochemistry,, University Autonoma of Madrid, Spain; 3 Faculty of Pharmacy, University of Lisbon,

Portugal; 4 Genzyme-Sanofi, Boston, USA; 5 INSERM-U1066, University of Angers, France

Background: Hyaluronic acid (HA) is attracting increasing attention for the design of targeted drug delivery systems. [1] Tailoring HA with a lipophilic chain is a good strategy to modify its original hydrophilic character. [2][3] Here, we set out to synthesize a hydrophobically modified HA derivate as a component to prepare HA nanocapsules (HA-NCs) using a spontaneous emulsification technique. These NCs were designed to efficiently encapsulate docetaxel (DCX) and for the attachment of a protein to the polymeric shell aimed for intracellular delivery.

Materials and methods: DCX loaded HA-NCs were prepared using a recently developed self-emulsification method in which the drug was solubilized in the oil core. Protein-associated HA-NCs were prepared by the same method by incubating blank NCs with the protein at different concentrations. NCs were characterized by size, polydispersion index (PDI) and zeta potential. [4] Cellular uptake and toxicity was done in A549 lung tumor cells and the effective delivery of the protein to the intracellular compartment was evaluated by confocal microscopy.

Results: After the optimization of the self-emulsification technique, DCX loaded HA-NCs showed a particle size around 150 nm, negative zeta potential and a high DCX entrapment (around 90%). The use of an oil/water extraction appears as a new strategy for IVR assays. The results shown that HA-NCs slowed the release of DCX comparing to the free drug, where we observed a burst release of 40% at time zero and a sustained profile of 70% after 24h. In-vitro cytotoxicity assays showed that HA-NCs were able to maintain DCX toxicity in A549 cells. Confocal microscopy images showed the ability of HA-NCs to carry the protein to the intracellular compartment. Conclusions and future perspectives: Up to now we have developed a versatile system for anticancer drug delivery: an oil core for hydrophobic anticancer agents and a polymeric surface for protein association. The ability of DCX loaded HA-NCs to effectively reduce the tumor is being evaluated in a xenograft subcutaneous model in mice. The delivery of proteins for specific intracellular targeting is an ambitious approach where polymeric NCs appear as promising carriers for accurate delivery.

References: 1. S. Mizrahy et al.” J. Control. Release, 156: 231–8, 2011; [2] H. Lee et al. Macromol. Biosci., 9: 336–42, 2009; [3] X. Dong et al. J. Nanomater., 2010:1–9, 2010.; [4] G. Bastiat, J. Control. Release, 170: 334–42, 2013.

(6) Prilling as a versatile technique for the development of controlled delivery systems containing anti-inflammatory drugs.

Andrea Cerciello, Felicetta De Cicco, Maria Chiara Pellegrino, Rita P. Aquino, Pasquale Del Gaudio, Paola Russo Department of Pharmacy, University of Salerno, Fisciano (SA), Italy

Purpose: The current study aimed to investigate the ability of the prilling technique to produce controlled release dosage forms containing poorly water soluble anti-inflammatory drugs with different biopharmaceuticals and pharmacodynamics

Page 18: HOSTED BY · 2016-01-05 · *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00

18 | P a g e

properties as well as similar short half-life (about 2 h), belonging to the non-steroidal (ketoprofen, K) or steroidal (prednisolone, P) class. Methods: Prilling/ionotropic gelation was selected as micro-encapsulation technique to produce drug loaded hydrophilic microparticles (gel-beads) in very mild and reproducible conditions. The influence of process parameters such as composition and viscosity of the aqueous feed solutions (sodium alginate 2.0-2.75% w/v in different ratio with drugs 1:10-1:4), aqueous bulks for ionotropic gelation and cross-linking time (2-10 min) on the obtained particles was evaluated. Beads morphology, size and solid state characteristics were analysed (SEM microscopy, FTIR, DSC). In vitro release study was assessed in conditions simulating the gastrointestinal environment (USP XXXVI). Results: Particles morphology as well as release kinetics of alginate formulations were strongly related to the amount of alginate and K or P loaded into the feed solutions. For both active pharmaceutical ingredients, better results were obtained increasing alginate and drug content in order to achieve a compact polymeric matrix able to better retain the drug. In particular, ketoprofen, with a lower water solubility (51 mg/L) requested a lower amount of alginate for an adequate control of drug release, compared to P (235 mg/L). The best formulations, loaded with K or P, released around 20% of drugs in simulated gastric fluid (SGF, pH=1); complete drugs release is achieved after pH change in simulated intestinal fluid (SIF, pH=6.8) in about 3.5 h. This behavior may be explained by a combination of alginate pH-dependent solubility, cross-linking properties of Zn2+ and polymeric matrix density; beads did not swell or erode in SGF and still keep intact matrix, whereas in SIF (at pH 6.8) they started to swell and further erode due to the ion-exchange. Conclusions: Conventional therapies of chronic inflammatory diseases (such as arthritis, arthrosis etc.) consists of multiple daily administrations of different anti-inflammatory drugs leading to a variable drug blood level and ineffective therapy. This study suggested that prilling is an appropriate technological approach to manufacture drug delivery systems able to control both non-steroidal and steroidal drug release. The formulations can be proposed as dosage forms for the treatment of chronic inflammatory diseases, able to achieve delayed and controlled therapeutic drug levels, reducing the number of daily dose and, consequently, the well-known side effects associated with NSAID and SAID long term use.

(7) Thalidomide Complexation by -cyclodextrins in New Nasal Powder Formulations for Hereditary Hemorrhagic Telangiectasia

A. Rossi1, V. Chiapponi1, P. Colombo1, M.T. Stigliani2, P. Russo2, B. Marchesano3, F. Bortolotti3, G. Colombo3, 1Department of Pharmacy, University of Parma, Parma 43124, Italy

2Department of Pharmacy, University of Salerno, Fisciano (SA) 84080, Italy 3Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara 44121, Italy

AIM This work aimed to design and manufacture thalidomide nasal powders using β-cyclodextrin, hydroxypropyl-β-cyclodextrin or sulphobutylether-β-cyclodextrin as carriers. Thalidomide nasal powder could enable topical treatment of nose bleeding in hereditary hemorrhagic telangiectasia as maintenance therapy complementing systemic administration per os. Powders were manufactured and characterized in vitro both for technological and biopharmaceutical features. EXPERIMENTAL METHODS Powders were obtained by kneading thalidomide (THAL) and cyclodextrins (CDs) in a mortar with an ethanol solution of lecithin. After drying, product size was calibrated through a 0.4 mm mesh sieve. THAL transport across and accumulation in freshly excised rabbit nasal mucosa was studied in vitro using Franz type cells in comparison with an aqueous drug saturated solution. After 4 h, THAL was quantified in all compartments, including the mucosa (upon homogenization and extraction). Powder muco-adhesion was also measured in vitro. Insufflation from different nasal devices and deposition into a human nasal model cast were studied. RESULTS AND DISCUSSION All powders showed good flow properties, with differences depending on the CD used (HP-β-CD > β-CD > SBE-β-CD). In vitro THAL transport across excised nasal mucosa in 4 hours was negligible from all powders. About 2-3% of the thalidomide initially loaded in the donor was found within the mucosal tissue, without significant differences among the powders. Thalidomide complexation by CDs improved muco-adhesion compared to the drug raw material as powder. The average THAL amount retained on the mucosa surface after 3 min washing with 45 ml of simulated nasal fluid was 8.1 ± 4.2, 12.5 ±

Page 19: HOSTED BY · 2016-01-05 · *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00

19 | P a g e

5.7 and 54.8 ± 25.7% of the total amount recovered for the powders with SBE-β-CD, HP-β-CD and β-CD, respectively. The faster the powder solubility in water, the greater the muco-adhesion.

Powders with β-CD and HP-β-CD were quantitatively emitted with MIAT device (mean emitted amounts of 19.0 0.7 mg and

21.0 0.7 mg for β-CD and HP-β-CD, respectively). The emitted amount was lower for the SBE-β-CD powder, likely due to its lower flow properties. A similar behavior was observed with the Teijin multidose device. However, powder deposition patterns in the human cast differed. With Teijin device, the powders deposited more in the vestibular, pre-turbinate and turbinate regions, than in the rhino-pharynx. In contrast, most of powder deposition upon delivery from MIAT device occurred beyond the nasal cavity, i.e., in the rhino-pharynx (from where the product could be swallowed more rapidly). This is due to the different insufflation air flow of the two devices.

(8) Fucoidan microparticles as inhalable antibiotic carriers

LC Cunha, AD Alves, A Grenha

CBMR – Centre for Biomedical Research, Drug Delivery Laboratory, University of Algarve, Campus Gambelas, 8005-139 Faro, Portugal

The lung has been explored as an effective route for the delivery of drugs in the ambit of respiratory diseases, allowing direct

targeting of the infected organ and possibly reducing systemic drug toxicity, which is a special advantage in diseases involving

long-term treatments, such as tuberculosis (TB) [1]. In order to overcome some of the limitations of lung delivery (airway

structure, local degradation) drug microencapsulation appears as a potential approach [2]. This work aimed at using fucoidan

(FUC) to produce microparticles (MP) that efficiently associate isoniazid (INH) and rifabutin (RFB) in combination, for an

application in pulmonary tuberculosis therapy. FUC MP were successfully produced by spray drying (Buchi mini-spray dryer,

B-290) an aqueous solution containing 2% (w/v) FUC with or without INH (10%, w/w) and RFB (5%, w/w). Unloaded and drug-

loaded microparticles were characterized regarding morphology (scanning electron microscopy, SEM), Feret diameter

(optical microscopy) and tap densities. The cytotoxicity of the formulations was determined by the MTT assay in A549 cells,

models of the alveolar epithelium, upon 24h exposure. Macrophage-differentiated THP-1 cells (using phorbol myristate

acetate) were exposed to fluorescently labelled FUC MP and their phagocytosis determined by flow cytometry. The number

of cells associated with fluorescence was considered the definition for uptake. FUC MP revealed a spherical shape with

irregular surfaces. The Ferret’s diameter of FUC MP was 1.5 - 1.6 μm, while tap density of unloaded FUC-MP was 0.80 ± 0.30

g/cm3, which decreased considerably to 0.35 ± 0.02 g/cm3 after drug association. The determination of drug encapsulation

efficiency is in course. A549 cells registered viability above 70% after 24h exposure to MP. Preliminary assays revealed that

around 20% of macrophages phagocytosed FUC MP. FUC MP thus showed theoretically adequate properties for pulmonary

delivery in the ambit of tuberculosis therapy. Funding from Portuguese Foundation for Science and Technology (PTDC/DTP-

FTO/0094/2012 and UID/BIM/04773/2013) is acknowledged. LC Cunha acknowledges PhD grant supported by Coordination

for the Improvement of Higher Education Personnel through the Brazilian Government scholarship programme Science

without Borders.

References: 1. R. Pandey et al. Antitubercular inhaled therapy: opportunities, progress and challenges. Indian Journal of Experimental Biology, 44: 357-66, 2005. 2. António J. Almeida and Ana Grenha. (2014). Technosphere®: An inhalation system for pulmonary delivery of biopharmaceutical. In Mucosal Delivery of Biopharmaceuticals (pp 483-98). DOI 10.1007/978-1-4614-9524-6_22.

(9) Combination of co-axial prilling and supercritical drying for the formulation of doxycycline particles for wound healing.

Felicetta De Cicco, Andrea Cerciello, Paola Russo, Rita P. Aquino, Pasquale Del Gaudio

Page 20: HOSTED BY · 2016-01-05 · *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00

20 | P a g e

Department of Pharmacy, University of Salerno, Fisciano (SA), I – 84084, Italy

Introduction: Natural polymers such as alginate with high mannuronic content and amidated pectin are known as biopolymers able to promote wound healing1; doxycycline is an inexpensive tetracycline with broad spectrum of action that also inhibits

MMPs and TNF-α converting enzyme (TACE) activity promoting wound healing, too2. The aim of this work was to develop a controlled-release doxycycline topic formulation in form of sterile dressing loaded with microparticles able to protect from infection, absorbing exudate and promoting healing. Combination of innovative microencapsulation techniques as prilling in co-axial configuration and supercritical antisolvent extraction (SAE) has been used to produce core/shell aerogel particles.

Materials and Methods: Amidated pectin (3.5% w/w solution) with different amounts of doxycycline suspended and sodium alginate aqueous solutions (1.5% and 1.75% w/w) were prepared. Microcapsules were produced using prilling apparatus in optimized operative conditions, equipped with coaxial nozzle, consisting of two concentric nozzles, one for the alginate annular solution (600μm) and the other for the doxycycline/pectin core (400μm). 0.5 M CaCl2 ethanol solution was used as gelling bath, obtaining alcohol-gel particles suitable for SAE, conducted at T 38°C, p 150 bar, flow 0,6 kg/h. Scanning electron microscopy (SEM), fluorescence microscopy (FM) and calorimetric analysis (DSC) were used to characterize core/shell particles. Drug release profiles were determined by Franz type diffusion cells.

Results: Prilling in co-axial configuration allowed the production of core/shell spherical microparticles with narrow size distribution. SAE allowed rapid and complete solvent elimination from hydrated particles, producing homogeneous dried aerogel retaining size and nanostructured texture of the gel. Good encapsulation efficiency (e.e. between 62.25% and 87.55%) was obtained mainly due to doxycycline low solubility both in the gelling solution and supercritical CO2. DSC analyses showed the interaction between polymers and drug, also confirmed by fluorescence microscopy analyses. In vitro drug release profiles showed an intense burst effect in the first 3 hours and complete drug release within 24h or 40 h. Furthermore, only after 48 h doxycycline begins its degradation in vitro. Conclusions: This study highlighted the advantages of a tandem technique based on prilling and SAE in the production of doxycicline controlled release microparticles. The intense burst effect followed by a sustained release of the drug till 48 h may allow an early control of bacteria infection and help the long-term treatment of infected wounds. Microparticles may be applied on wounds preventing the spreading of the infection and promoting healing.

References: 1. Thomas, A. et al. Alginates from wound dressings activate human macrophages to secrete tumour necrosis factor-alfa. Biomaterials, 21 (17): 1797-1802, 2000. 2. Anumolu, S. S. et al. Doxycycline loaded poly (ethylene glycol) hydrogels for healing vesicant-induced ocular wounds. Biomaterials, 31 (5): 964-974, 2010.

(10) Taste-masked Powders for Pediatric-centric Dispersed Dosage Formats

N. Dormer1, D. Shi1, C. Dennis1, C. Berkland1,2

1Orbis Biosciences, Inc., Lenexa, KS, 66214, USA; 2Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, 66045, USA; [email protected]

Purpose Format-flexible oral drug powders that are palatable and can be accurately-dosed provide an effective alternative to standard tablets and capsules. Here, we describe a scalable microencapsulation technology that produces powders that can be palatable, precisely dosed and incorporated into a range of dosage formats. The microsphere powder technology was used to precisely mimic a common over–the-counter expectorant, Guaifenesin.

Methods

Page 21: HOSTED BY · 2016-01-05 · *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00

21 | P a g e

Uniform microcapsules of guaifenesin were prepared in a single step using Orbis’ Precision Particle Fabrication technology. Briefly, the API was co-melted with a mix of waxes, hydrophilic excipients, and binders to create an amorphous drug-matrix dispersion. The resulting solution was sprayed through a co-axial nozzle which was acoustically excited using an ultrasonic transducer controlled by a frequency generator to produce uniform microspheres droplets. The microspheres were collected in a stainless steel collection vessel and concentrated for further use. Microspheres were placed on accelerated stability at 40 °C/75% RH, and dissolution tested at 0, 7, 14, and 21 days. In vitro release from the micropsheres was tested using United States Pharmacopeia (USP) Type II apparatus at 37 °C, and compared to the commercially-available , Maximum Strength Mucinex®, 1200 mg extended-release bi-layer tablets.

Results Transmitted light microscopy of the guaifenesin microspheres revealed smooth, free-flowing, spherical particles with a narrow size distribution. Compared to the respective commerical tablet formulation, the microencapsulated powder dose exhibited identical release kinetics with the same 1200 mg dose, with a 97.2 similarity factor. Differential scanning calorimetry also suggested that the guaifenesin was amorphously dispersed within the wax-excipient matrix.

Conclusion Precision Particle Fabrication was successfully used to create uniform microspheres that, compared to a commercial tablet formulation, exhibited identical release kinetics. It stands to reason that the guaifenesin powder formulation could easily be scaled back to mimic a smaller does (such as a 600 mg Mucinex® tablet). In addition, the particle fabrication technology could be used for other active pharmaceutical ingredients, such as ibuprofen, erythromycin, levetiracetam, prednisone, ritonavir. These formulations would be highly relevant in pediatric, geriatric, and persons with dysphagia, where large or foul-tasting tablets and capsules are not easily palatable. We are currently developing a pilot-scale system capable of producing up to 40 kg clinical batches of these novel microencapsulated formulations.

(11) Dual-Functional Liposomes for Effective Cancer Cell Targeting and Improved Intracellular Delivery

Pranali P. Deshpande a, Swati Biswas Ph.D.b and Vladimir P. Torchilin Ph.D., D.Sc.a*

a Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115.

b Department of Pharmacy, Birla Institute of Technology and Sciences-PiIani, Hyderabad Campus, Jawahar Nagar, Hyderabad, Andhra Pradesh 500078, India.

Purpose: Formulation of doxorubicin-loaded liposomes modified with Octa-arginine (R8) and Transferrin (Tf) with an aim to achieve better cancer cell targeting and enhanced intracellular drug delivery in cancer cells.

Methods: Initially, commercially available Doxil® was modified using an arginine-rich cell penetrating peptide R8 with a goal to improve the intracellular delivery of doxorubicin. Liposome uptake and internalization were studied using flow cytometry and confocal microscopy, respectively. In vitro cytotoxicity and in vivo tumor regression studies were performed in breast and lung tumor models to compare and establish the usefulness of R8-Doxil® over Doxil®. To overcome the issue of nonspecificity of R8 towards cancer cells, we chose a second ligand Tf as a targeting molecule and designed dual-functional liposomes (DualL) using both R8 and Tf. Tf receptors (TfRs) are over-expressed on tumor cells and have a high turnover rate that is proportional to the proliferation potential of tumors. TfR over-expression along with their ability to internalize makes them an attractive target for chemotherapy and specific cancer-cell targeting. We determined the usefulness of the DualL over single-ligand modified liposomes, by studying their uptake, contribution of both ligands in internalization and their therapeutic potential in vitro, in TfR over-expressing ovarian cancer cells.

Results: Flow cytometry and confocal imaging studies showed that R8-Doxil® exhibited higher association with lung and breast cancer cells at two tested time-points, and also showed improved nuclear delivery of therapeutic potential for R8-Doxil®doxorubicin. In vitro cytotoxicity and in vivo tumor regression studies showed a higher therapeutic potential for R8-Doxil®. In an ovarian cancer model, DualL demonstrated a 2-fold increase in uptake over R8-liposomes and mechanistic analysis proved the contribution of both ligands. This effect is also translated into improved toxicity in vitro.

Page 22: HOSTED BY · 2016-01-05 · *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00

22 | P a g e

Conclusion: Modification of Doxil® with R8, improves intracellular delivery of doxorubicin enhancing its delivery to the nucleus which is its site of action. Further, formulating DualL helps to circumvent the problem of non-specificity of R8 towards cancer cells and imparts a cancer-cell targeting effect. All studies helped us establish that the DualL possess several properties like passive targeting, improved intracellular delivery and cancer cell-surface targeting, all in the same system.

(12) Hot-melt Extrusion Microencapsulation of Polyphenols for Taste-masking

Yuancai Dong, Chia Miang Khor, Wai Kiong Ng, Parijat Kanaujia

Institute of Chemical and Engineering Sciences, 1 Pesek Road, Jurong Island, Singapore 627833 Email: [email protected]

Background: With the greater health awareness, plant-derived polyphenols (e.g. quercetin) with anti-inflammatory and anti-oxidant properties have gained increasing attention in functional food development. However, polyphenols are bitter in taste and direct incorporation of such ingredients into staple diets has been the challenge for the food industry to gain the consumer’s acceptance. Microencapsulation, by forming a physical barrier between the bitter bioactives and the taste buds, is an ideal strategy for masking the unpleasant taste of the functional food ingredients, such as minerals, vitamins, antioxidants, etc [1-2]. The purpose of this work is to develop taste-masked quercetin particles by use of hot-melt extrusion microencapsulation technique.

Experimental: Quercetin and encapsulant (carnauba wax, shellac and zein) were mixed at the weight ratio of 60/40, 70/30 and 80/20, manually fed and extruded using Prism Eurolab 16 Twin Screw Extruder (Thermo Scientific, Germany) at appropriate temperature and screw speed. The extrudates were collected as strands, cooled at room temperature and ball-milled (Retsch Mill 300, Germany) for 2 min at30 Hz. In vitro taste-masking efficiency was evaluated by investigating dissolution in pH 6.8 medium (saliva) within 2 min

Results and conclusions: Quercetin was successfully encapsulated through hot-melt extrusion with three GRAS encapsulants at weight ratio of 60/40 and 70/30 except 80/20. The milled sample had the size of 200-500 µm. 14% of the pure quercetin was dissolved in pH 6.8 medium within 2 min, which was reduced to only 2% for the encapsulated formulation suggesting an excellent taste-masking efficiency. In pH 1.0 medium (gastric), the dissolution of quercetin from the encapsulated formulation was comparable to the pure quercetin. Our work demonstrates that hot-melt extrusion microencapsulation is a promising technique for masking the bad taste of the functional food ingredients.

References: 1. Nazzaro et al., Microencapsulation in food science and biotechnology. Curr Opin in Biotech 23:182–186, 2012 2. Champagne et al., Microencapsulation for the improved delivery of bioactive compounds into foods. Curr Opin Biotech 18:184–190, 2007

(13) Delivery of liposomes across the skin using needle-free liquid jet injectors

Anna Maria Faddaa, Michele Schlicha, Francesco Laia, Sergio Murgiab, Donatella Valentia, Chiara Sinicoa

aDept. Scienze della Vita e dell'Ambiente, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy bDept. Scienze Chimiche e Geologiche, University of Cagliari, S.S. 554 Bivio Sestu, 09042 Monserrato (CA), Italy

Introduction Liquid jet injectors are devices that use a high-speed stream of fluid to deliver molecules across the skin into the intradermal, subcutaneous or intramuscular region without the use of a needle. These devices, which have been studied for more than 50

Page 23: HOSTED BY · 2016-01-05 · *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00

23 | P a g e

years, are clinically used for mass immunization and delivery of macromolecules such as insulin and growth hormones as well as small molecules [1]. Non-steroidal anti-inflammatory drugs, including diclofenac, are usually applied on the skin surface to reach deep tissue below the skin for the relief of pain symptoms in muscular strains, sprains and contusions. Local application could be beneficial also in the treatment of rheumatic and osteoarthritic diseases but the difficulty of overcoming the barrier of the skin and subsequent cutaneous clearance make the drug pharmacokinetics complex [2]. In this study, we investigated for the first time the ability of liquid jet injectors to deliver intact lipid vesicles. The key innovation of this work is the combination of two technologies such as liposomes and jet injectors. Indeed, in this study, different phospholipid vesicle formulations, such as conventional liposomes and transfersomes, were developed and tested for subcutaneous administrations of diclofenac sodium by means of a liquid jet injector. Methods Data concerning size, size distribution and surface charge of vesicles before and after the injections were measured by dynamic light scattering experiments. Bilayer integrity and thickness was checked by means of X-ray scattering techniques. The effect of vesicle fast jet injection through the skin on drug release kinetics was checked by in vitro experiments. Results Diclofenac sodium loaded phospholipid vesicles, were prepared and fully characterized. The lipid vesicles were delivered through a new born pig skin specimen using a jet injector and the collected samples were analyzed to assess both vesicle structural integrity and drug retention after the injection. Both liposomes and transfersomes showed the same features before and after injection, thus suggesting the integrity of vesicles after skin crossing as a high-speed liquid jet. Overall results obtained in this study may broaden the range of application of liquid jet injectors to lipid vesicle based formulations thus combining beneficial performance of painless devices with those of liposomal drug delivery systems.

References: 1. S. Mitragotri, Current status and future prospects of needle-free liquid jet injectors. Nat. Rev. Drug Discov. 5: 543-548, 2006. 2. C.F. Goh, M.E. Lane, Formulation of diclofenac for dermal delivery. Int. J. Pharm. 473: 607-616, 201.4

(14) Encapsulation of active principles in PCL for knitted fabric functionalization

Ada Ferri, Roberta Peila, Naveeta Kumari, Manuela Mihailiasa, Antonello Barresi All Department of Applied Science and Technology, Politecnico di Torino, Italy

Micro- and nanocapsules containing active principles are widely used for dermal and transdermal applications in cosmetics and pharmaceutical preparations. Recently, the dispersion of microcapsules on fabrics have paved the way to new types of products, named cosmeto-textiles [1]. Such products, combining ease of use of a garment and controlled-release from microcapsules, are ideal candidates for complementary therapy of diseases like psoriasis, which require long-term treatment and dedication to the therapy. Nanoencapsulation of caffeine, menthol and melatonin in PCL is discussed in this work, where two different systems for solvent displacement (the confined-impinging jet mixer and the multi inlet vortex mixer) have been extensively investigated. For each mixer, several process parameters, such as fluid dynamics, type of solvent and polymer-to-drug ratio, have been considered to find the optimal configuration for micro- or nanocapsule formation. In Figure 1, an example of nanoparticles prepared in the same conditions (polymer type and concentration, solvent, mixing conditions) loading different substances in the confined impinging jet mixer is shown. The initial mass ratio (MR) of loading substance and polymer may not be the only or main factor determining final size, as it can be noted that nanocapsule formed by using the triglyceride oil are significantly larger than particles prepared dissolving a solid component.

Figure 1 Size of nanoparticles prepared loading PCL

Mw= 14000, 6 mg/mL in acetone, with different

substances, dissolved in acetone with the polymer :,

Figure 2 In-vitro release from a knitted cotton

fabric with embedded melatonin-PCL

nanoparticles (6 mg/ml melatonin- 25 mg/ml

0 20 40 60 80 100 120 140 160 180 200

0,0000

0,0001

0,0002

0,0003

Higuchi model

Cum

ula

tive

rele

ase

of

mel

atonin

(g)

time (minutes)

0.1 1 10

1000

vj, m s

-1

dp,

nm

100

Page 24: HOSTED BY · 2016-01-05 · *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00

24 | P a g e

Complex phenomena can take place during the solvent displacement process, with the formation of a new liquid phase by oiling out: menthol in water/acetone is an example of a substance for which this phenomenon has been observed directly by the authors. Finally, in-vitro release test from fabrics with embedded nanocapsules has been carried out in a Franz cell equipment confirming that fabrics can act as comfortable substrates for controlled release of active principles (Figure 2). References:

1. S.Y. Cheng, C.W.M. Yuen, C.W. Kan, K.K.L. Cheuk, J.C.O. Tang, Systematic Characterization of Cosmetic Textiles, Textile Research Journal Vol 80 (6): 524–536

(15) Unveiling a role for polysaccharides on macrophage targeting in tuberculosis therapy

Ana D. Alves1, Susana Rodrigues1, Ana M. Rosa da Costa2 and Ana Grenha1

1CBMR – Centre for Biomedical Research, Drug Delivery Laboratory; 2Center for Research in Chemistry of Algarve; University of Algarve, Campus de Gambelas, Faro, Portugal

Tuberculosis remains a leading cause of death and therapeutic failure is attributed to non-compliance with prolonged treatments.1 New therapeutic strategies are demanded and the direct targeting of Mycobacterium hosts (alveolar macrophages) is an interesting approach, concentrating drugs at the main infection site.2 Many polysaccharides present in their structure mannose (Man) or galactose (Gal), structural units potentially recognized by macrophage receptors, promoting phagocytosis.2 In this context, we proposed to design aerodynamically suitable and biocompatible polysaccharide-based microparticles (MPs), while verifying their ability to directly target and activate alveolar macrophages. Three different polysaccharides were tested for the production of MPs: k-carrageenan (CRG) composed of Gal, locust bean gum (LBG) composed of Man/Gal (4/1) and partially hydrolyzed guar gum (PHGG) also composed of Man/Gal (2/1). A solution of each polymer was spray dried (Büchi B-290 mini spray dryer) at a concentration of 2% (m/v), alone or with the antitubercular drugs isoniazid and rifabutin in combination. MPs based on each of the three polysaccharides were successfully obtained, and exhibited adequate Feret diameter (1.1 – 1.96 µm), aerodynamic diameter (1.3 - 2.65 µm) and real density (1.07 – 1.39 g/cm3) for the objective of reaching the alveolar zone upon inhalation.3 The drug loading capacity was determined by UV-Vis spectrophotometry or high-performance liquid chromatography. The polysaccharides used as matrix materials enabled the association of the two antibiotics with efficiencies between 57 and 100%. Fluorescently-labelled MPs of each formulation were prepared to determine their capacity to undergo macrophage capture. This ability was tested in macrophage-like THP-1 cells, which were differentiated using phorbol 12-myristate 13-acetate. Fluorescently-labelled MPs were aerosolized with Dry powder Insufflator™ onto a monolayer of macrophages. After 2h exposition, the occurrence of phagocytosis was analyzed by flow cytometry (results compared with cells not exposed). When the higher dose of MPs was tested (200 µg/cm2), all polysaccharides registered macrophage uptake above 99%. However, at a lower dose (50 µg/cm2) macrophage uptake preference was LBG > PHGG > CRG, suggesting a preference of phagocytosis of Man units in detriment of Gal. The ability of MPs to activate macrophages is currently being determined. Considering the high affinity of macrophages for the proposed MPs, these carriers are considered adequate as the basis of an inhalable therapy for pulmonary tuberculosis. This work was supported by National Portuguese funding through FCT - Fundação para a Ciência e a Tecnologia, projects PTDC/DTP-FTO/0094/2012 and UID/BIM/04773/2013. The PhD scholarship to Susana Rodrigues (SFRH/BD/52426/2013) is also acknowledged.

References: 1. Pham et al. Pulmonary drug delivery systems for the treatment of tuberculosis. Drug Deliv Syst, 23:474–480, 2015. 2. Patel et al. Particle engineering to enhance or lessen particle uptake by alveolar macrophages and to influence the therapeutic outcome. Eur J Pharm Biopharm, 89:163–174, 2015. 3. Grenha et al. Microencapsulated chitosan nanoparticles for lung protein delivery. Eur J Pharm Sci, 25:427–437, 2005.

Page 25: HOSTED BY · 2016-01-05 · *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00

25 | P a g e

(16) Polymeric nanoparticles for the oral delivery of camptothecin

J.M. Irache1, J. Huarte1, S. Espuelas1, Y. Lai2, N. Martín-Arbella1, R. Penalva1, B. He2

1Dep. of Pharmacy and Pharmaceutical Technology, University of Navarra, Pamplona, Spain. 2National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China.

Camptothecin (CPT) is a potent anticancer compound that blocks Topoisomerase 1, an enzyme required for DNA transcription and replication. However, the oral delivery of this drug is faced to a number of challenges. Firstly, CPT shows a very low aqueous solubility. Secondly, the chemical structure of CPT includes an unstable lactone ring that is highly susceptible to spontaneous and reversible hydrolysis, yielding an inactive and more toxic carboxylate form [1]. Finally, CPT is also a P-glycoprotein substrate [1]. The aim of this work was to evaluate the capability of polymeric nanoparticles to promote the oral absorption of CPT. For that, a PK study in Wistar rats was carried out. In this study, animals were divided in the following groups: (a) CPT suspension iv administered, (b) CPT suspension orally, and (c) CPT-loaded nanoparticles. In all cases, the CPT dose was 1 mg/kg. The amount of CPT was determined in an HPLC system with fluorescence detector. The pharmacokinetic analysis was performed with the WinNonlin 5.2 software (Pharsight Corporation, USA). The oral administration of CPT after its encapsulation into the nanoparticles offered prolonged and sustained plasma levels of the anticancer drug for at least 48 h. As a consequence, these nanoparticles displayed a higher AUC than the traditional drug suspension, improving 7-times the oral relative bioavailability of CPT. Interestingly, the toxicity assays did not reveal any important secondary effect associated to the delivery of CPT encapsulated into nanoparticles. In addition, when CPT was orally administered in the form of nanoparticles, its half-life in plasma and volume of distribution increased and the drug clearance decreased as compared with the suspension. These findings would be explained by the capability of these nanoparticles to cross the mucus layer and reach the surface of the enterocyte, in which they would release their cargo, increasing the residence time of nanoparticles within the gut and disturbing the intestinal P-glycoprotein. In any case, this approach would be in line with an improved efficacy of this anticancer drug. In fact, it has been described that a prolonged and sustained Topo 1 inhibition (including with daily low-dose dosing regimens) can mediate a hypoxia-inducible factor 1 alpha inhibition mechanism, augmenting the efficacy of the drug [2]. In summary, the study revealed that these nanoparticles would be capable to improve the absorbed fraction of the given dose of CPT offering prolonged and sustained regimens of the drug for at least 48 h. Acknowledgements: This work was partially supported by grant from the UE FP7 (Heptag Exchange project, IRSES, Marie Curie Action # 295218). References: [1] Venditto VJ, et al., Cancer therapies utilizing the camptothecins: A review of the in vivo literature. Mol Pharmaceutics, 7:307–349, 2010. [2] Onnis B, et al., Development of HIF-1 inhibitors for cancer therapy. J Cell Mol Med, 13:2780–2786, 2009.

(17) Resveratrol-loaded liposomes: A promising nanomedicine for malignant gliomas

Aditi Jhaveri1 and Vladimir Torchilin1 Ph.D., D.Sc. 1 Center for Pharmaceutical Biotechnology and Nanomedicine (CPBN), Department of Pharmaceutical Sciences, Northeastern

University, Boston, MA 02115

Glioblastomas (GBMs) are highly aggressive brain tumors with a poor prognosis. They harbor a small population of slow-dividing cells known as tumor-initiating cells (TICs) that resist therapy, are highly tumorigenic and are responsible for the formation, maintenance, invasiveness and recurrence of GBMs. Conventional chemotherapeutics act on the bulk of the tumor cells, sparing the TICs, and also exhibit severe toxicities to normal tissues, thus greatly hampering the quality of life of patients. Lasting tumor regression thus necessitates the use of drugs that are well tolerated and can affect both the bulk tumor cells and TICs.

Page 26: HOSTED BY · 2016-01-05 · *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00

26 | P a g e

In this project, we investigated resveratrol (RES), a natural polyphenol that has pleiotropic health benefits, and the chemopreventive effects of which have been demonstrated in all the major stages of cancer including initiation, promotion and progression (1). However, using RES as a free drug has several limitations including a poor water solubility, chemical instability, low bioavailability and poor pharmacokinetics (2). To counter these, we developed and characterized a pegylated liposomal formulation of RES (RES-L). We also developed and characterized in vitro TIC models from two glioblastoma cell lines (LN-18 and U-87MG) using the neurosphere (NS) assay. Cytotoxicity studies with free RES and RES-L demonstrated significant dose and time-dependent cytotoxic effects of RES on both the cell lines. Our preliminary results also show that RES was able to inhibit the anchorage-independent growth of GBM NS in a dose-dependent manner. We examined the general mechanisms of action of RES on GBM and found that at low concentrations (20-75 µM) it caused a significant arrest of cells in the S phase of the cell cycle and blocked the S to G2/M progression of cells. At higher concentrations, (>75µM), RES resulted in a significant increase in the reactive oxygen species (ROS) in GBM cells, suggesting that it probably acts as a pro-oxidant at higher doses, which maybe one of the mechanisms by which it induces cell death. From our initial studies, liposomal RES seems like a promising agent for further development against gliomas.

References: 1. Jang, M et al. Cancer chemopreventive activity of resveratrol, a natural product derived from grapes. Science, 275 (5297) 218-20, 1997 2. Neves, A.R. et al. Resveratrol in medicinal chemistry: A critical review of its pharmacokinetics, drug-delivery and membrane interactions Curr. Med. Chem. 19, 1663-1681, 2012

(18) Mucus-penetrating, pH sensitive polymersomes

Beatriz N. M. Miranda1,2, Li-Heng Cai2, Laura R. Arriaga2, Emanuel Carrilho1, and David A. Weitz2

1Instituto de Quimica de Sao Carlos, Universidade de Sao Paulo, Sao Carlos, Brazil.

2 John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, United States

Mucus protects the human body by trapping foreign particulates and also poses a barrier for drug delivery by slowing down the mobility of drug carriers. To design mucus penetrating carriers, solid particles are typically coated with inert polymers such as polyethylene glycol or Pluronic polymers to prevent mucoadhesion. However, the solid structure of the particles limits their loading and the process to coat them requires a complex synthesis. Here we develop a simple method to fabricate nanocarriers with an exceptional combination of properties; these include a good mucus-penetration capability and a large loading capacity. Unlike conventional coating methods, we use a diblock copolymer, consisting of both hydrophobic and hydrophilic blocks, which self-assembles into polymersomes under hydration. Because of the inert attribute of the hydrophilic block, these polymersomes are mucus-penetrating by nature. Moreover, their hollow structure provides the polymersomes with a large loading capacity. Importantly, by introducing small quantities of pH sensitive polymers into the polymersome membrane, we demonstrate that these polymersomes can release contents upon application of external stimuli such as pH. Together with its simple fabrication, this mucus-penetrating, pH-responsive polymersome provides a new platform for mucosal drug delivery. References: 1. Min Liu, Jian Zhang, Wei Shan & Yuan Huang. Developments of mucus penetrating nanoparticles. Asian J. Pharm. Sci. 10, 275–282 (2015). 2. Lai, S. K., Wang, Y. Y. & Hanes, J. Mucus-penetrating nanoparticles for drug and gene delivery to mucosal tissues. Adv. Drug Deliv. Rev. 61, 158–171 (2009). 3. Discher, D. E. & Ahmed, F. Polymersomes. Annu. Rev. Biomed. Eng. 8, 323–41 (2006).

Page 27: HOSTED BY · 2016-01-05 · *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00

27 | P a g e

(19) Perfluorocarbon emulsions as ultrasound-mediated delivery vehicles in fibrin scaffolds

Alexander Moncion, Keith J. Arlotta, Oliver D. Kripfgans, Renny T. Franceschi, Andrew J. Putnam, Mario L. Fabiilli

University of Michigan, Ann Arbor, MI USA Introduction: Fibrin scaffolds are protein-based hydrogels that are often used in regenerative medicine as a reservoir for macromolecules and substrate for cellular invasion. Molecular diffusion from a fibrin hydrogel is typically a passive process. Spatio-temporally controlled release can be achieved by doping fibrin scaffolds with sonosensitive perfluorocarbon (PFC) vesicles containing a desired payload. Release from the resulting acoustically responsive scaffold (ARS) is then triggered by focused ultrasound (US) applied non-invasively, on-demand, and with sub-millimeter precision – leading to the release of the encapsulated payload. This work focuses on the US-modulated payload release from ARSs with varying PFC emulsion formulation.

Materials and Methods: Sonosensitive perfluoropentane (PFP, C5F12), perfluorohexane (PFP, C6F14), and a 1:1 PFP:PFH ad-mixture double emulsions (W1/PFC/W2, mean diameter: 4.95 ± 0.59 μm, 3.86 ± 0.06 μm, 2.71 ± 0.10 μm, respectively) were prepared as described previously by our group with AlexaFluor 680 dextran (AF680, 10 kDa) contained in the W1 phase. ARSs (0.5 mL volume) were cast in 24 well plates by doping 5 mg/mL fibrin gels with 1% (v/v) emulsion, adding 0.5 mL of overlying DMEM, and then incubating at 37°C. A positive control was prepared by incorporating AF680 directly into the hydrogel, i.e. without emulsion. A subset of the ARSs were exposed to US (2.5 MHz, Pr = 8 MPa, 13 cycles, 10 Hz pulse repetition frequency) for ~2 min one day after polymerization. Samples of the overlying media were taken throughout the experiment and analyzed with a plate reader.

Results and Discussion: In all cases, ARSs exposed to US had a higher percentage of AF680 released at 144 h versus the corresponding –US condition: 49% vs. 24% for PFP ARSs, 20% vs. 5% for PFP:PFH admixture, and 12% vs. 3% for PFH. However, AF680 release from ARS was in all cases less than AF680 release from the positive control (88%). A B C

Figure: (A,B,C) In vitro release profile of AF680 from PFP, 1:1 PFP:PFH, PFH ARSs (+/- US) and a positive control (n = 5). US was applied 24 h after polymerization. *p < 0.05 vs. –US condition. Conclusions: US can increase the release of AF680 from the ARSs, with a burst release of 21%, 9%, and 5% occurring after exposure for PFP, PFP:PFH admixtures, and PFH ARSs. ARSs containing higher boiling point PFCs yielded less AF680 release over time. On-going experiments aim to optimize release from ARSs and translate to in vivo models.

0%

20%

40%

60%

80%

100%

0 20 40 60 80 100 120 140 160

Ma

ss F

ra

cti

on

Rele

ase

d

Time (h)

PFP Release Profile

Positive Control +US -US

* * * * * *

0%

20%

40%

60%

80%

100%

0 20 40 60 80 100 120 140 160

Ma

ss F

ra

cti

on

Rele

ase

d

Time (h)

PFH Release Profile

Positive Control +US -US

* * * * * *

0%

20%

40%

60%

80%

100%

0 20 40 60 80 100 120 140 160

Ma

ss F

ra

cti

on

Rele

ase

d

Time (h)

PFP/PFH Release Profile

Positive Control +US -US

* * * * * *

Page 28: HOSTED BY · 2016-01-05 · *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00

28 | P a g e

(20) Novel multicompartmental microcapsules for the oral targeted delivery of a potential antidiabetic drug, probucol, using

bile acid-based formulation: anti-hyperglycaemic and anti-inflammatory effects in an animal model of Type 2 diabetes

mellitus

Armin Mooranian1, Rebecca Negrulj1, Ryusuke Takechi2, Hani Al-Salami1

1Biotechnology and Drug Development Research Laboratory, School of Pharmacy, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia 2School of Public Health, Curtin Health Innovation Research Institute,

Curtin University, Perth, Western Australia, Australia

Introduction: This study aimed to develop and characterise PB-microcapsules, incorporated with the bile acid ursodeoxycholic acid (UDCA), in vitro, and examine its potential anti-hyperglycaemic and anti-inflammatory effects in vivo.

Method: Two types of microcapsules were prepared: one without UDCA (PB-SA, control) and one with UDCA (PB-UDCA-SA, test). Microcapsules were characterised in vitro and in vivo. In vitro: morphology, surface composition and topography, physico-chemical stability, excipient’s thermal and chemical compatibility, rheology, mechanical strength, and PB release profile at various conditions micking the upper and lower gastrointestinal tract (GIT). In vivo: 50 mice (5-weeks old) were randomly allocated into 5 equal groups and given: low fat diet (LFD), high fat diet (HFD), metformin (M), PB-SA and PB-UDCA-SA daily for 1 month, and their blood glucose (BG), glycated haemoglobin (HbA1c), visceral fat deposition (abdominal lipid accumulation) and inflammatory serum biomarkers (TNF-α, IFN-γ, IL-1β, IL-6, IL-10, IL-12 and IL-17) were analysed (n=3). PB concentrations in serum, ileal mucosa and faeces were also analysed.

Results and Discussion: PB-UDCA-SA microcapsules showed good morphological and release properties, compared with PB-SA, in vitro. Oral administration of PB-UDCA-SA reduced BG and HbA1c in HFD mice (p<0.01) to levels similar to those treated with M and also significantly reduced visceral fat depositions to an extent greater than PB-SA and M. Compared with HFD, PB-UDCA-SA reduced serum levels of TNF-α (p<0.5) and IL-10 (p<0.01), while PB-SA reduced serum levels of TNF-α (p<0.01) and IFN-γ (p<0.05) and exerted a hypoglycaemic effect similar to that of PB-UDCA-SA. In addition, PB concentrations from PB-UDCA-SA microcapsules were lower in serum and ileal tissues (p<0.01) and higher in faeces (p<0.01) compared with PB-SA.

Conclusion: The incorporation of UDCA into PB-microcapsules improved its in vitro delivery characteristics and both microcapsules showed hypoglycaemic and anti-inflammatory effects suggesting potential applications in the treatment of diabetes.

(21) Targeted delivery of PEG-PE-based micelles encapsulating a novel pro-apoptotic drug, NCL-240, in 3D cancer cell spheroids

B. Pattni1*, S. Nagelli1, B. Aryasomayajula1, P. Deshpande1, A. Degterev2, V. Torchilin1

1Center for Translational Cancer Nanomedicine, Northeastern University, Boston, MA 2Department of Biochemistry, Tufts University, Boston, MA

*email: [email protected]

Abstract: A targeted micellar delivery system for the novel pro-apoptotic anticancer drug candidate, NCL-240, was prepared and evaluated using a cancer cell spheroid model. NCL-240 was successfully loaded in transferrin (Tf)-modified PEG2000-PE micelles. NCI-ADR cells (ovarian cancer) were used to form spheroids by the non-adhesive liquid overlay technique. The spheroids partly mimicked the in vivo tumor, indicated by the presence of live cells on the periphery and a necrotic core as demonstrated with H&E staining. Cytotoxicity studies demonstrated that the targeted delivery markedly improved NCL-240 drug efficacy. Confocal microscopy showed that the Tf-conjugated (targeted) micelles had increased spheroid penetration into the spheroids as compared to the untargeted micelles. It is speculated that the nanosized targeted micelles induced a

Page 29: HOSTED BY · 2016-01-05 · *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00

29 | P a g e

‘sink effect´ after penetrating the tumors because of endocytosis action by the transferrin receptors on the surface of the cells, thus inducing more micelles to enter the spheroids. This effect could not be seen with untargeted micelles which were rather associated more with the cells around the periphery of the spheroids. Acknowledgement: This work was supported NCI grant 5U54CA151881 to V. Torchilin

(22)Stimuli-sensitive siRNA delivery by hypoxia-targeted nanoformulation

Sarisozen C1*, Perche F1, Biswas S1, Wang T1, Zhu L1, Torchilin VP1,2

1Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA, USA 2Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia

*[email protected]

ABSTRACT: The altered vasculature and the resultant abnormal blood flow rate in the tumor mass lead to the presence of regions with low oxygen tension and acute hypoxia. In this study, we synthesized a novel hypoxia-sensitive conjugate where the polycationic polymer polyethyleneimine (PEI), which is conjugated to the fusogenic lipid DOPE, was linked to a PEG2000 chain via a hypoxia sensitive azobenzene group. Resulting conjugate (referred as PAPD) is able to efficiently complex siRNA in aqueous environment due to PEI and form nano-sized ‘core-shell’ micelle-like structures. Once in hypoxic environment, the hypoxia-sensitive azobenzene group led to the detachment of the PEG chain allowing for an increased internalization of the remaining DOPE-PEI/siRNA complex. PAPD and its hypoxia-insensitive counterpart PPD, fully complexed siRNA starting from N/P ratios of 10. The siRNA in the complexes was found to be stable for 5 days without visible degradation. The conjugates protected siRNA from degradation by RNase. The PEG chain detachment in hypoxic conditions resulted in a slight particle size decrease (from 222±96nm to 167±61nm) and considerable positive charge increase (from 0.1±6.5mV to 13.2±3.7mV) of the complexes. The hypoxia-sensitive PAPD complexes were able to significantly down-regulate the expression of GFP in various GFP over-expressing cancer cell lines under hypoxia conditions (up to 55%) whereas non-silencing effect was observed under normoxia conditions or with the hypoxia-insensitive PPD complexes. The PAPD complexes significantly downregulated the therapeutic proteins selectively in hypoxic conditions compared to normoxic conditions and PPD complexes. Survivin, a caspase activation inhibitor negatively regulating apoptosis, was downregulated 85.1±5.3% by PAPD in multidrug resistant SKOV-3TR cells under hypoxic conditions. P-gp, an efflux protein responsible of decreased intracellular drug concentrations was also significantly downregulated 35±1.1% in the same cell line. The FAM-labeled-siRNA and rhodamine-labeled-PAPD penetrated into deeper layers of the 3D cancer cell spheroids under hypoxic conditions compared to PPD complexes. The in vivo tumor accumulation of the developed systems resulted in 2-fold increase only after treatment with fluorescently labeled PAPD, while the PPD was not detectable in the tumors. In vivo GFP downregulation was also investigated in A2780/GFP tumor model. After the intravenous injection of the hypoxia-sensitive formulation, GFP downregulation of about 25% was detected by ex vivo imaging and ca. 30% GFP downregulation was confirmed by flow cytometry. Overall, the developed hypoxia-sensitive nanopreparations have been proven to be effective for both in vitro and in vivo stimuli-sensitive siRNA delivery. ACKNOWLEDGEMENTS: This work was supported by the NIH grant (U54CA151881) to Vladimir P. Torchilin.

(23) Amphipathic tri-block co-polymers interaction with DPPC large unilamellar vesicles

Soto-Arriaza, M.A. 1,2*, Palominos M.A.1

1: Departamento de Química-Física, Facultad de Química, Pontificia Universidad Católica de Chile, Av. Vicuña Mackenna 4860, Casilla 306, Correo 22, C.P. 7820436, Santiago, Chile.

2: Centro de Investigación en Nanotecnología y Materiales Avanzados CIEN-UC, Pontificia Universidad Católica de Chile, Av. Vicuña Mackenna 4860, C.P. 7820436, Santiago, Chile.* e-mail: [email protected]

ABSTRACT: We report the effect of amphipathic tri-block copolymers arranged as PCL-PEO-PCL on the dynamic and structural

Page 30: HOSTED BY · 2016-01-05 · *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00

30 | P a g e

properties o f a dipalmitoylphosphatidylcholine in large unilamellar vesicles. Copolymers interaction was observed at concentration below and above its cmc. Turbidity at 50 or 150 mM NaCl was higher than the one at 1 mM. Flocculation was not observed even after 48 h of samples incubation at room temperature. On the other hand, steady-state fluorescence measurements demonstrated a typical behavior; at low temperatures, the presence of the copolymers produce a decrease but in the liquid crystalline phase produced an increase in the GP value of Laurdan probe. This behavior can be explain due to that PEO2000 and PEO4600 both reduce the quantity or mobility of water molecules in the interface of the lipid bilayer in liquid crystalline and gel phase, respectively. In order to understand how copolymer interacts with lipid bilayer, we determined the time-resolved fluorescence anisotropy parameters of DPH. The most important result is related to the increase of the order parameter (S) at 25°C (gel phase) and decrease at 55°C (liquid crystalline phase). On the other hand, rotational correlation time (Φ) increase with temperature and decrease in presence of PEO2000 below and above cmc unlike to PEO4600 that Φ value increase at high polymer concentration (above cmc). On the other hand, the presence of PEO4600 at concentration above cmc increases the rate of water outflow compared with PEO2000 which had no effect on the water outflow. On the other hand, at low concentration, permeability of calcein decrease but at high concentration, above cmc, calcein permeability increase reaching the initial values in absence of polymers. According to the preliminary results we can indicate that the presence of copolymers in DPPC LUV shown an important effect upon the physicochemical properties of the bilayer. In gel phase, both copolymers increase the order of acyl chain in the inner part of the bilayer and increase the water mobility in the interface of the bilayer, but in liquid crystalline phase both copolymers decrease the order of acyl chain in the inner part of the bilayer but reduce water mobility in the interface of the bilayer. This behavior could be explained through a preferably interaction between copolymers with the interface of lipid bilayer in liquid crystalline phase.

ACKNOWLEGMENT(S): We thank Dr. A. Leiva and Esteban Bossel (Polymers Laboratory) for the kind gift of PEO2000 and PEO4500. This work was supported by Fondo Nacional de Desarrollo Científico y Tecnológico (FONDECYT), Chile, Grant N° 1141012. Also acknowledged support of Dirección de Investigación de Posgrado, Facultad de Química, Pontificia Universidad Católica de Chile.

(24) Polyelectrolyte nanocapsules as a promising drug delivery system

K. Szczepanowicz, K. Podgórna, T. Kruk, M. Piotrowski, M. Łapczyńska, S. Lukasiewicz and P. Warszyński Jerzy Haber Institute of Catalysis and Surface Chemistry PAS, Niezapominajek st 8, 30-239 Krakow, Poland

Traditional pharmaceuticals rarely demonstrate specific affinity towards the site of their action and as a rule, they distribute throughout the body upon administration. To reach the action site, a pharmaceutical agent has to overcome the inactivating action of the aggressive biological medium and cross a variety of biological barriers, which frequently results in at least partial drug inactivation/degradation and unfavorable pharmacokinetics and biodistribution. In addition, many pharmaceutical agents could provoke multiple undesirable side effects in normal organs, tissues and cells. The solution of these complicating problems are targeted drug delivery systems. The use of polymeric nanocapsules in targeted drug delivery has huge expectations because nanoscale materials interact effectively with biological systems and may overcome many intractable health challenges. Development of a new nanocarriers requires better understanding of interactions between nanocaspsules and immune system which allows optimization of nanosystems properties for effective drug delivery. Therefore, in the present studies we focused on the investigation of the interactions between polyelectrolyte nanaocapsules and mouse murine macrophages cell line (RAW 264.7) and human leukemic monocyte cell line (THP-1). Nanocapsules based on a liquid core encapsulation with polyelectrolyte multilayer shells were prepared by the technique of sequential adsorption of polyelectrolytes (LbL) using AOT (docusate sodium salt) as the emulsifier and biocompatible polyelectrolytes: polyanion PGA (Poly-L-glutamic acid sodium salt) and polycation PLL (poly L-lysine). The average size of obtained capsules was 80nm. Pegylated external layers were prepared using PGA-g-PEG (PGA grafted by PEG poly(ethylene glycol)). Influence of physicochemical properties of the nanocapsules (charge, size, surface modification) on viability, phagocytosis potential, endocytosis was studied. Internalization of tested nanocapsules was determined by flow cytometry and confocal microscopy. Moreover, we assessed whether addition of the PEG chains, downregulates particle uptake by phagocytic cells. Presented results confirm that obtained PEG-grafted NPs are promising candidates for drug delivery.

Page 31: HOSTED BY · 2016-01-05 · *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00

31 | P a g e

ACKNOWLEGMENT: This study was partially supported by the NCN project 2011/03/D/ST5/05635

DIRECTIONS AND CAMPUS MAP

Curry Student Center:

All talks, poster sessions, breakfasts and coffee breaks will take place in the Curry Student Center Ballroom (2nd floor, building

#50 on map) unless otherwise noted. Lunch will be served in the McLeod Suites (room 318-320-322) on the 3rd floor.

Transport to Northeastern:

Public Transit: Northeastern is well served by public transit: Green Line – Northeastern stop, Orange Line - Ruggles.

Parking: Parking is available in the Renaissance (Building #62 on map) and Gainsborough (Building #45 on map) Garages.

Parking rates are based on an hourly fee structure. Weekend rates are available.

Odyssey Cruises, Rowes Wharf – Conference Banquet on Friday:

Bus transportation:

A bus shuttle will be offered for conference attendees in front of Ruggles station (#61 on map) to Rowes Wharf at 17:30

returning to the campus, or the conference hotels Inn at Longwood and Colonnade per request, after the cruise ends

(expected return time to campus between 22:30 – 23:00).

Public transportation:

Take the Orange Line train from Ruggles station to Oak Grove; connect to the Blue Line station Wonderland and exit at

Aquarium station; walk for 4 minutes to Rowes Wharf.

Parking at Rowes Wharf Parking:

Discounted parking is available at Rowes Wharf Garage located directly under the Boston Harbor Hotel at 30 Rowes Wharf on

Atlantic Avenue. Validation for discounted parking can be obtained at the Odyssey ticket booth. Weeknight rate after 17:00:

$12.

Cruise Attire:

Business casual and dress attire (for women). Casual jeans, t-shirt, athletic shoes or flip flops are discouraged.

Page 32: HOSTED BY · 2016-01-05 · *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00

32 | P a g e

Page 33: HOSTED BY · 2016-01-05 · *Please mount your poster no later than 8:30 on October 1, 2015. Presenters or co-presenters are expected to stand near their poster from 16.00 to 17.00

33 | P a g e

20th International Symposium on

MICROENCAPSULATION

Northeastern University Boston MA USA September 17-20, 2015