Conference Schedule

Day1: August 29, 2019

Keynote Forum

Biography

Hervé Hillaireau is Associate Professor of Pharmaceutical Technology at the School of Pharmacy of Université Paris-Sud since 2009. His research at Institut Galien Paris-Sud relates to the design and the toxicological evaluation of biodegradable nanoparticles as drug nanocarriers, with a focus on: (i) nanocarrier design for nucleoside/nucleotide analogues and nucleic acids delivery in anticancer and antiviral therapies; (ii) nanocarrier surface functionalization for molecular targeting; (iii) toxicological evaluation of biodegradable nanoparticles. He has been supervising or co-supervising 10 PhD theses. He is the author of 4 book chapters and 38 peer-reviewed international research articles, and has recently co-edited a special issue of Advanced Drug Delivery Reviews on Aptamers in therapeutics and drug delivery


Abstract

Downregulation of genes involved in cancer progression using therapeutic nucleic acids remains a promising alternative to conventional chemotherapy. This approach however necessitates an appropriate delivery system allowing not only intracellular delivery but also the targeting of cells involved in cancer progression. To this end, CD44 has been characterized a major biomarker, being overexpressed by many tumors including lung, breast, colon, pancreatic, head, and neck cancers.

Our group has developed over the past years hyaluronic acid (HA)-decorated nanocarriers able to deliver small interfering RNA (siRNA) and DNA plasmids [1] to CD44-expressing lung cancer cells. In particular, HA-decorated lipoplexes exhibited a lamellar structure able to deliver nucleic acids in vitro to the CD44-expressing A549 lung cancer cells through CD44-mediated endocytosis, and inhibited the expression of the luciferase (luc) reporter gene [2]. In vivo, these lipoplexes significantly inhibited luc after intravenous administration to mice bearing an A459-based lung cancer [3].

In order to increase the specificity of this delivery, we investigated the use of aptamers to functionalize the surface of nanocarriers. Aptamers are nucleic acid-based ligands that can bind their targets with a high affinity and specificity, with virtually no toxicity and immunogenicity. A 2ʹ-F-pyrimidine RNA aptamer was successfully selected against the CD44 protein by the SELEX method and named Apt1 [4]. Apt1 was conjugated to the surface of liposomes by a thiol-maleimide coupling. Such liposomes exhibited a strong affinity to various CD44-expressing cells, even higher than Apt1 alone [5]. SiRNA-loaded, Apt1-decorated aptamers successfully inhibited luc in the CD44-expressing breast cancer MDA-MB-231 cells in vitro. After intravenous administration to mice bearing an orthotopic model of MDA-MB-231-based breast cancer, Apt1-targeted liposomes successfully inhibited luc in the tumor cells, in a specific and targeted manner [6]. These results show the potential of aptamers to selectively target oncogenes to CD44-expressing cancer cells in vivo

Tracks

  • Advanced Nanomaterials | Nanotech for Energy and Environment | Graphene and its Applications Nanotechnology in Water Treatment | Nano Biomaterials | Nano Materials Synthesis and Characterisation Pharmaceutical Nanotechnology
Location: Johnson

Mohtada Sadrzadeh

University of Alberta, Canada

Chair

Biography

Prof. Per Hansson has his expertise in colloidal systems of relevance for developing drug delivery systems. Most of his studies have dealt with polyelectrolytes and their interaction with colloids of opposite charge, in particularly phase transitions and phase separation in polyelectrolyte hydrogels in relation to the loading and release of amphiphilic drugs, proteins and peptides. He is currently investigating the self-assembling properties of amphiphilic drugs, and the fate of biomacromolecular drug formulations after injection into the subcutaneous tissue. This involves the development of in vitro methods to study the release from formulations and transport through the extracellular matrix. Hansson is leading the Pharmaceutical Physical Chemistry group at the Department of Pharmacy, Uppsala University, and the Parenteral Drug Delivery Platform of the Swedish Drug Delivery Forum


Abstract

Statement of the Problem: Microgels have the capacity to store large amounts of amphiphilic, peptide and protein drugs. This makes them interesting as delivery systems for drugs needing protection against degradation, and as components of slow-release and depot formulations for parenteral administration. Despite the fact that microgels are used clinically, e.g., to deliver doxorubicin in the treatment of liver cancer [1], little attention has been given to how the drug self-assembly inside microgels affects the release properties. The purpose of this study is to show that that the release of amphiphilic drugs is strongly influenced by self-assembly [2] and its effect on the swelling of the microgel network. Methodology & Theory: Loading and release of cationic amphiphilic drugs are investigate by means of micropipette-assisted microscopy of single microgels [2, 3] (Fig. 1) and μDiss apparatus studies. We have developed and evaluated two new models combining transport dynamics and thermodynamics of drug self-assembly, one for non- swelling responsive microgels based on the Nernst-Planck equation [2], and one for responsive networks handling coexisting phases with different degrees of network swelling. Findings: For two polyelectrolyte microgel systems, DC bead® used clinically, and polyacrylate microspheres (diameter: ~100 μm), the drug self-assembly has a profound effect on the loading and release kinetics as well as on the distribution of the drug inside the beads and the distribution between different beads in a suspension. The roles played by salt in the release medium and the responsiveness of the polymer network are highlighted. The micropipette technique is found to be a powerful in vitro method.  Conclusions & Significance: The results highlight the importance of the electric coupling of diffusive fluxes, the amphiphilic self-assembly and the responsiveness of the microgel. The results are important for the development of microgel drug delivery systems with improved release kinetics

Biography

Christoph Alexiou, in the year 2000 received his degree as an ENT-Physician and 2002 he changed to the ENT-Department in Erlangen, Germany, where he performed his postdoctoral lecture qualification (Habilitation). He is working there as an assistant medical director in the clinic and leads the Section for Experimental Oncology and Nanomedicine (SEON). Since 2009 he owns the Else Kröner-Fresenius-Foundation-Professorship for Nanomedicine at the Universityhospital Erlangen. He receives grants from the European Union, German Research Community (DFG), Ministry of Education and Science (BMBF) and Bavarian State Ministry of the Enviroment and Consumer Protection and is a member of the Executive Board of the European Technology Platform for Nanomedicine (ETPN). His research is addressing the emerging fields of  Diagnosis, Treatment and Regenerative Medicine using magnetic nanoparticles and the translation from basic research into clinical trials and published >150 papers in peer reviewed journals. He received for his research several national and international renowned awards


Abstract

Nanoparticles offer promising new possibilities for a multiplicity of medical applications including therapy and diagnosis of various diseases. Especially iron oxide nanoparticles provide a broad application spectrum as contrast agents, magnetic transporters, or heat carriers in hyperthermia treatment. While particles used for imaging should circulate for extended periods of time in the vascular system, nanoparticles designed for use as drug carriers should be efficiently taken up by the target cells. Importantly, nanoparticles for medical (and several other) applications must be biocompatible, meaning that after contact with cells no adverse effects are elicited. To translate basic findings into clinical trials several requirements such as detailed synthesis and characterization of the nanoparticles, nanotoxicological testings, ex vivo models to simulate in vivo conditions for appropriate adjustment of the necessary parameters and pre-clinical animal studies have to be addressed. These results are of pivotal importance to start with respective GMP production and approval, which is essential for translating these products into clinical trials (scheme). SEON (Section of Experimental Oncology and Nanomedicine) addresses these issues with a special focus on drug delivery in oncology (1,2,3,4,5,6) and their promising potential applications in cardiovascular (7,8), regenerative medicine (9), imaging (1,10) and infections (11). The aim is the translation of the preclinical results into clinical trials and the respective steps necessary to gain this ambitious object

Biography

Cesar Torres is a third-year PhD student at the University of Maryland, in College Park, MD, US.  He has a dual bachelor’s degree in Chemical Engineering and Chemistry, and a minor in Material Science & Engineering from the University of Idaho, in Moscow, ID, USA. As an undergraduate student, he participated in a summer research internship program at the Whitesides Group in Harvard University, working on nano-fabrication and soft-lithography.  As a PhD student, he is focused on the application of nanotechnology to controlled release systems in contact lenses.


Abstract

Statement of the problem: Ophthalmic diseases are most commonly treated with eye drops due to the low costs and self-administration. However, it has been reported that eye drops could deliver only about 5% of functional ingredients contained in a burst dosage since they suffer from tear drainage in addition to corneal and sclera barriers.  To address the limitations of eye drops, researchers have explored the use of therapeutic contact lenses. Nevertheless, a major limitation of contact lenses in drug delivery is that most of the drug absorbed is released within the first few hours, fact that limits the use for extended release.  The purpose of this study is to demonstrate the application of vitamin E diffusion barriers with ionic surfactants into contact lenses, in order to achieve controlled release of non-steroidal-anti-inflammatory drugs (NSAIDs).  Methodology: Silicone-hydrogel commercial contact lenses were soaked in ethanol solutions containing vitamin E and an ionic surfactant. Secondly, a NSAID was added to each lens by soaking the lenses in drug solutions for determined periods of time. The in vitro release experiments were carried by immersing the drug-containing contact lenses in phosphate buffer saline at physiological pH.  Results: Contact lenses containing vitamin E extend drug delivery up to a factor of 50, depending on the amount of vitamin E loaded and the physichochemical properties of the drugs.  Vitamin E in contact lenses acts as a diffusion barrier that retards the diffusion of the drugs to the release medium. Contact lenses containing both vitamin E and a long-chain cationic surfactant further extend drug delivery due to electrostatic interactions between the anionic NSAIDs and the surfactant.  Conclusion: Contact lenses containing vitamin E and cationic surfactants can provide extended release of NSAIDs, and could be potential biomaterials to be used for the treatment of ophthalmic diseases. However, ocular irritation and toxicity studies would be needed to evaluate the safety of the approach. 

Biography

Peptide-based vaccines: Dr. Hussein focus on the development of peptide-based vaccines against infectious diseases and cancer. This includes (1) synthesis of adjuvanting moieties for stimulation of immune system; (2) applying different methods for conjugation of peptides and lipids; (3) determination of the size, shape and charge of the self-assembled vaccine particles; (4) investigate the biological efficiency of vaccines in both in vitro and in vivo. Gene delivery: Currently, Dr. Hussein is working on the development of targeted nanoparticle delivery system to deliver the siRNA to cytoplasm. This delivery system includes peptide-based, micelles and/or liposome formulations


Abstract

Conjugation of multiple peptides by their N-termini is a promising technique to produce branched multiantigenic vaccines. We established a double conjugation strategy that combines a mercapto-acryloyl Michael addition and a copper-catalysed alkyne-azide 1,3-dipolar cycloaddition (CuAAC) reaction to synthesise self-adjuvanting branched multiantigenic vaccine candidates. These vaccine candidates aim to treat cervical cancer and include two HPV-16 derived epitopes and a novel self-adjuvanting moiety. This is the first report of mercapto-acryloyl conjugation applied to the hetero conjugation of two unprotected peptides by their N-termini followed by a CuAAC reaction to conjugate a novel synthetic lipoalkyne self-adjuvanting moiety. In vivo experiments showed that the most promising vaccine candidate completely eradicated tumours in 46% of the mice (6 out of 13 mice).

Biography

Glòria Garcia Ortega is a PhD student at the Universitat Autònoma de Barcelona whose work deals with lipo-based delivery systems for anticancer applications. The group where she stays focuses their research on the challenging area of bioinorganic chemistry (metallothioneins, synthesis of Cu and Pt chemotherapeutic complexes and metallosomes for biomedical applications). Her final Bachelor’s degree project lied on the synthesis, characterization and application of metal and metal-oxides nanoparticles. After her Master’s degree in Industrial Chemistry and Introduction to Chemical Research, she did an industrial research internship in order to develop nanocomposites for superconducting materials


Abstract

Metallosurfactants (MTS) are tensioactive metal complexes that gather properties of both constituents: the surfactant and the metal ion.1,2These molecules exhibit self-assembly properties and, when mixed with phospholipids, are able to form stable vesicles, known as metallosomes, which size ranges from nanoparticles to microns. They have been proposed as reliable and efficient tools for drug delivery systems in cancer treatment.3,4Although platinum drugs are widely used in the treatment of several types of solid tumors, the inclusion of Pt chemotherapeutic agents in such vesicles is reported to enhance their biocompatibility and allows to reduce immunogenicity and non-specific toxicity, diminishing side-effects in front of the free drug.5,6

An unexplored Pt(II)-MTS family of linear anionic alkyl sulfonated ligands have been synthesised and fully characterised by several techniques (1H-NMR, 13C-NMR, 195Pt-NMR, ESI-MS, ICP-OES and ATR-FTIR). This particular MTS family bears the polar head group opposite to the coordinated Pt(II) centre, hence giving rise to a non-standard MTS structure.7The aggregation properties of both, the amphiphilic ligands and the corresponding Pt(II)-MTS have been studied by DLS and UV-vis, as well as by optical microscopy and cryo-TEM. Tensioactive ligands have not shown a suitable hydrophilic/hydrophobic moieties ratio to form aggregates by themselves. However, the coordination of some of these ligands to Pt(II) moiety allows their assembly in the micron-range (Figure 1). Mixed vesicles of Pt(II)-MTS and phospholipids at different concentrations have been obtained in aqueous media to study the Pt internalisation in the phospholipid bilayer and the metallosomes size.8Finally, preliminary cytotoxic studies have shown the potentiality of these compounds to be used as promising anticancer lipodrug candidates.

Biography

Hervé Hillaireau is Associate Professor of Pharmaceutical Technology at the School of Pharmacy of Université Paris-Sud since 2009. His research at Institut Galien Paris-Sud relates to the design and the toxicological evaluation of biodegradable nanoparticles as drug nanocarriers, with a focus on: (i) nanocarrier design for nucleoside/nucleotide analogues and nucleic acids delivery in anticancer and antiviral therapies; (ii) nanocarrier surface functionalization for molecular targeting; (iii) toxicological evaluation of biodegradable nanoparticles. He has been supervising or co-supervising 10 PhD theses. He is the author of 4 book chapters and 38 peer-reviewed international research articles, and has recently co-edited a special issue of Advanced Drug Delivery Reviews on Aptamers in therapeutics and drug delivery


Abstract

Nanomedicine hav brought tremendous hope of targeted and personalized medicine. In particular, they have improved the bioavailability of poorly soluble drugs, reduced side effects of several drugs, and enabled drug targeting to diseased tissues, especially in the field of cancer treatment. However, a widespread use of nanomedicine still faces several important impediments, especially in the case of hydrophilic drugs for several reasons. (i) Despite variable drug encapsulation efficiencies into nanocarriers, the drug loading achieved is generally low, which limits the drug dose and/or requires the administration of high amounts of excipients (eg. lipids or polymers forming the nanocarrier). (ii) The drug release is often poorly controlled and prone to the ‘burst release’ effect, and the stability of nanocarriers in physiological media may be unsatisfying, therefore limiting the targeting potential of nanomedicine. (iii) Each drug often requires a specific nanocarrier system, which is often challenging to scale-up, which dramatically limits the applicability into the industry and the clinics [1].

To address these limitations, we have designed a drug delivery platform tailored to hydrophilic drugs, especially nucleotides/nucleotide analogues. This platform relies on the preparation of chitosan-iron coordination complexes, which form nanogels in presence of hydrophilic drugs. Such nanogels act as nanocarriers of therapeutic nucleotides such as adenosine triphosphate (ATP), as well as nucleotide analogues such as azidothymidine triphosphate (AzT-TP).

Our group has first demonstrated the ability of triphosphate group-containing drugs to induce ionotropic gelation of chitosan. This was demonstrated using the nucleotide ATP and the nucleotide analogue AzT-TP. The resulting nanogels can be seen as nucleotide/nucleotide analogue nanocarriers, showing both efficient encapsulation and drug loading, up to 44% w/w [2]. To further investigate the potential of chitosan-metal complexes to improve the nanogel stability in physiological media, coordination complexes of chitosan and Fe(III) have been synthesized and used to form nanogels in presence of ATP, which clearly demonstrated a high resistance to physiological ionic strengths, increasing with the Fe(III) content. These results highlight the potential of metal association to chitosan to form nanogels with tunable stability and drug release profile [3].

Day2: August 30, 2019

Keynote Forum

Biography

Thierry Delair is full professor at Claude Bernard University, Lyon 1, since November 2008. Before, he spent 20 years in different R&D departments in the bioMérieux group where he developed polymers for in vitro medical diagnostics and nanodelivery systems for sub-unit or DNA vaccines. He supervised 26 PhD students, numerous trainees and postdoctoral students. He published 155 articles in international peered-reviewed journals (h-index =34 ,number of citations 3855), filed 22 patents, participated in 8 book chapters, and gave 62 oral conferences (12 as invited speaker). His results allowed the creation of three companies: Ademtech (Pessac, France), producing magnetic particles, Anabior (Saint Ismier, 38), pharmaceutical society specialized in the production of particles-based adjuvants for vaccines and CYTOSIAL Biomedic (Lyon) a cosmetics company. His current research activities are focused on the elaboration and the characterization of nano-structured physical hydrogels based on polysaccharides for tissue engineering applications, drug or vaccine delivery


Abstract

Drug delivery systems (DDS) prove essential for a better efficacy and safety of the chemotherapies and that fight diseases and improve the quality of life of patients.

The efficiency of DDS lies in their capabilities at delivering the right amount of an Active Pharmaceutical Ingredient (API), at the right location in the body, in the safest manner and more comfortable way for the patients. Moreover, DDS should be efficiently manufactured via robust, green and low cost processes involving the use of raw materials complying with a sustainable development. Hence, new generations of DDS will need a global and multidisciplinary approach, addressing the above-mentioned challenges.

Here, we report on our strategy of DDS elaboration, using polyelectrolyte complexes (PECs) of biosourced polysaccharides leading to tailored and multifunctional nanodelivery systems.

PECs result from the self-assembly of oppositely charged polymers in water.  During the charge neutralization step, counter-ions and water molecules are released, inducing a gain in entropy of the system. Therefore, PECS formation is spontaneous, energy efficient as it takes place in water under mild experimental conditions, without needing high shear rates nor organic solvents. In that sense, polyelectrolyte complexation is a safe and green process. When this process is used with biosourced polysaccharides, generally regarded as safe, we have at hand a formulation process that meets many of the above requirements.

We will establish that by a precise control of the physico-chemistry properties of chitosan, and also of the parameters of the assembly (for example by switching the assembly from a kinetically controlled to a more thermodynamically controlled process) we produced carriers featuring a fair colloidal stability in physiological media, a capacity at encapsulating molecular drugs and at interacting with macromolecules like proteins, either for targeting purposes or for inducing appropriate immune responses for vaccine delivery purposes.

Biography

Sergey Suchkov was born in the City of Astrakhan, Russia, in a family of dynasty medical doctors. In 1980, graduated from Astrakhan State Medical University and was awarded with MD. In 1985, Suchkov maintained his PhD as a PhD student of the I.M. Sechenov Moscow Medical Academy and Institute of Medical Enzymology. In 2001, Suchkov maintained his Doctor Degree at the National Institute of Immunology, Russia.

From 1989 through 1995, Dr Suchkov was being a Head of the Lab of Clinical Immunology, Helmholtz Eye Research Institute in Moscow. From 1995 through 2004 - a Chair of the Dept for Clinical Immunology, Moscow Clinical Research Institute (MONIKI). In 1993-1996, Dr Suchkov was a Secretary-in-Chief of the Editorial Board, Biomedical Science, an international journal published jointly by the USSR Academy of Sciences and the Royal Society of Chemistry, UK.

 


Abstract

A new systems approach to diseased states and wellness result in a new branch in the healthcare services, namely, personalized and precision medicine (PPM). To achieve the implementation of PPM concept, it is necessary to create a fundamentally new strategy based upon the subclinical recognition of biomarkers of hidden abnormalities long before the disease clinically manifests itself.

Each decision-maker values the impact of their decision to use PPM on their own budget and well-being, which may not necessarily be optimal for society as a whole. It would be extremely useful to integrate data harvesting from different databanks for applications such as prediction and personalization of further treatment to thus provide more tailored measures for the patients resulting in improved patient outcomes, reduced adverse events, and more cost effective use of the latest health care resources including diagnostic (companion ones), preventive and therapeutic (targeted molecular and cellular) etc. A lack of medical guidelines has been identified by responders as the predominant barrier for adoption, indicating a need for the development of best practices and guidelines to support the implementation of PPM! Implementation of PPM requires a lot before the current model “physician-patient” could be gradually displaced by a new model “medical advisor-healthy person-at-risk”. This is the reason for developing global scientific, clinical, social, and educational projects in the area of PPM to elicit the content of the new branch

Biography

Dr Mohammad Najlah has an extensive experience in Pharmaceutical and Allied Health Higher Education as a teacher, researcher and manager. He is currently Deputy Head of School of Allied Health at Anglia Ruskin University. Mohammad has an excellent research profile focusing on the development of novel nanomaterials, polymeric prodrugs/conjugates and polymeric nano-particulates for drug delivery. In addition to overcoming mutli-drug resistance by nanoformulations, Mohammad is mainly interested in developing formulations for disulfiram and its metabolites for cancer therapy


Abstract

Disulfiram (DSF), an antialcoholism drug, has shown a selective cytotoxicity towards a wide range of cancer cells.  The anti-cancer activity of DSF is copper (Cu+2) dependent as the reaction between both yields cancer-targeting reactive oxygen species (ROS) and diethyldithiocarbamate (DDC) complex with Copper (DDC-Cu) owning a strong NFkB-inhibiting activity leading to cancer cell death. However, the clinical use of DSF as an anti-cancer drug is limited by its biological instability in the blood (t1/2 < 4 min). The aim of this study is to design, formulate and characterize PEGylated nanoparticles of DSF to improve stability and provide a proposed long circulation to target cancer cells. Several formulations of PEGylated PLGA/PCL nanoparticles (NPs) loading DSF have been manufactured by direct nanoprecipitation using two water-miscible organic solvents, acetone/methanol (3:1). The resulting NPs were fully characterized; size analysis, zeta potential measurements were performed using Laser Doppler Velocimetry. Drug loading efficiency and stability studies in horse serum were performed and DSF was detected using a validated HPLC method. Freez-dried NPs were also tested using DSC, TGA, FT-IR and SEM. Cytotoxicity in vitro on triple negative breast cancer cells MDA-MB 231 and MDA-MB 231pac10 (resistant to paclitaxel 10 nM) was evaluated by MTT assay. All NPs were within a size range of 175-225 nm with narrow PDIs (less than 0.3) and negative zeta potential rang (-25 to -45 mV); loading DS had no impact on these properties. Encapsulation efficiency (EE) was around 55% for all NPs but the none PEGylated PLGA/PCL showing the highest EE% (76%). The NPs provided high stability for DSF in horse serum; (t1/2 > 90 min for all NPs) and the PEGylated NPs demonstrated sustainable release for 96 h. The MTT cytotoxicity assay demonstrated high cytotoxicity of the DSF-NPs on both cell lines. PLGA/PCL PEGylated nanoparticles have a great potential for breast cancer treatment.

Tracks

  • Nanomedicine | Cancer and Nanotechnology | Novel Drug Delivery Systems | Personalised Nanomedicine | Nanomaterials for Drug Delivery | Drug Delivery Research | Drug Delivery and Device Development
Location: Johnson

Mohammad Najlah

Anglia Ruskin University, UK

Chair

Biography

Sergey Suchkov graduated from Astrakhan State Medical University and awarded with MD, then in 1985 maintained his PhD at the I.M. Sechenov Moscow Medical Academy and in 2001, maintained his Doctorship Degree at the Nat Inst of Immunology, Russia. From 1987 through 1989, he was a senior Researcher, Koltzov Inst of Developmental Biology. From 1989 through 1995, he was a Head of the Lab of Clinical Immunology, Helmholtz Eye Research Institute in Moscow. From 1995 through 2004, a Chair of the Dept for Clinical Immunology, Moscow Clinical Research Institute (MONIKI. Dr Suchkov has been trained at: NIH; Wills Eye Hospital, PA, USA; Univ of Florida in Gainesville; UCSF, S-F, CA, USA; Johns Hopkins University, Baltimore, MD, USA. He was an Exe Secretary-in-Chief of the Editorial Board, Biomedical Science, an international journal published jointly by the USSR Academy of Sciences and the Royal Society of Chemistry, UK. At present, Dr Sergey Suchkov is a Chair, Dept for Personalized and Translational Medicine, I.M.Sechenov First Moscow State Medical University. He is a member of the: New York Academy of Sciences, USA; American Chemical Society (ACS), USA; American Heart Association (AHA), USA; EPMA (European Association for Predictive, Preventive and Personalized Medicine), Brussels, EU; ARVO (American Association for Research in Vision and Ophthalmology); ISER (International Society for Eye Research); PMC (Personalized Medicine Coalition), Washington, USA.


Abstract

Abs against myelin basic protein/MBP endowing with proteolytic activity (Ab-proteases with functionality) are of great value to monitor demyelination to illustrate the evolution of multiple sclerosis (MS). Anti-MBP autoAbs from MS patients and mice with EAE exhibited specific proteolytic cleavage of MBP which, in turn, markedly differed between: (i) MS patients and healthy controls; (ii) different clinical MS courses; (iii) EDSS scales of demyelination to correlate with the disability of MS patients to predict the transformation prior to changes of the clinical course.

Ab-mediated proteolysis of MBP was shown to be sequence-specific whilst demonstrating five sites of preferential proteolysis to be located within the immunodominant regions of MBP and to fall inside into 5 sequences fixed. Some of the latter (with the highest encephalitogenic properties) were proved to act as a specific inducer of EAE and to be attacked by the MBP-targeted Ab-proteases in MS patients with the most severe (progradient) clinical courses. The other ones whilst being less immunogenic happened to be EAE inducers very rare but were shown to be attacked by Ab-proteases in MS patients with moderate (remission-type) clinical courses.

The activity of Ab-proteases was first registered at the subclinical stages 1-2 years prior to the clinical illness. About 24% of the direct MS-related relatives were seropositive for low-active Ab-proteases from which 22% of the seropositive relatives established were being monitored for 2 years whilst demonstrating a stable growth of the Ab-associated proteolytic activity. Moreover, some of the low-active Ab-proteases in persons at MS-related risks (at subclinical stages of MS), and primary clinical and MRT manifestations observed were coincided with the activity to have its mid-level reached. Registration in the evolution of highly immunogenic Ab-proteases would illustrate either risks of transformation of subclinical stages into clinical ones, or risks of exacerbations to develop.

The activity of Ab-proteases in combination with the sequence-specificity would confirm a high subclinical and predictive (translational) value of the tools as applicable for personalized monitoring protocols. Ab-proteases can be programmed and re-programmed to suit the needs of the body metabolism or could be designed for the development of principally new catalysts with no natural counterparts. Further studies on targeted Ab-mediated proteolysis may provide a translational tool for predicting demyelination and thus the disability of the MS patients

Biography

Fiorenza Rancan has a degree in Pharmaceutical Chemistry and Technology at the University of Padua (Italy) and gained her PhD at the Humboldt University of Berlin (Germany). Since 2012 she is associated scientist and laboratory manager at the Clinical Research Center for Hair and Skin Science at the Charité Medical University of Berlin (Germany). Her expertise lays in drug delivery systems, dermal and transdermal drug delivery as well as ex vivo skin models and organ culture as alternatives to animal studies. Her main research fields are inflammatory skin diseases, transcutaneous vaccination, as well as skin and chronic wound infections. 


Abstract

Skin is a very sophisticated barrier and to deliver adequate amounts of drug to healthy as well as diseased tissue is still a challenge. Nanotechnology has the potential to improve the treatment of several skin diseases. To test these new formulations, it is necessary to have models reproducing not only the physical skin barrier but also the environmental and biological feature of skin diseases. Three-dimensional systems like reconstructed skin offer the possibility to reproduce partially the skin barrier and to test biological effects on both keratinocytes and fibroblasts. However, these models do not consider skin immune active cells like dendritic and mast cells. Skin appendages like the pilosebaceous unit are also missing. Animal studies offer the possibility to include all physical and biological parameters of skin. However, beside ethical reasons, they are expensive and results are not always reproducible in humans. Ex vivo skin has been used since years to test skin penetration of drugs. Thus, in order to test innovative nanoparticle-based drug delivery systems our group has developed ex vivo skin models reproducing skin inflammatory diseases and wound infections. These models were used to test drug delivery kinetics, toxicity, anti-inflammatory activity as well as antibacterial efficacy of different types of nanocarrier formulations. The results show that ex-vivo human skin models are realistic animal-free systems useful to investigate drug efficacy as well as the interactions between applied material and skin immune system.

Biography

NH Nazirah Kamarudin has her interest in empowering materials for various applications, especially using porous and nano materials for adsorption and catalysis. During her master study, she introduced acidic metals into zeolites and studied its intrinsic acidity for the catalytic isomerization, which brought her further into insight into modification of mesoporous silica nanoparticles (MSN) during phD, By using ibuprofen as the model drug, various modification was conducted to tailor the mesoporous silica structures to suit the demand in controlled drug release application. These include the study of physiochemical properties of the material as well as understanding the mechanisms of adsorption and release through the surface. The use of nanoscale technologies to design the drug carrier had been her passion since then. Currently, she works on the modification of MSN using carbon and polymeric materials for the anti-cancer drugs adsorption and release.


Abstract

The mesostructured silica nanoparticles (MSN) have been extensively studied for the purpose of drug delivery application due to their unique properties such as biocompatibility, high stability, porous structure to withhold the bulky drugs, high and tailorable internal and external surface area, as well as functionalizable surface. However, MSN is hydrophilic while most anti-cancer drugs are hydrophobic.  The purpose of this study was to modify the MSN with activated carbon (AC) which is expected to produce more efficient adsorption and release medium for the anticancer drug doxorubicin (DOX) as the drug model. MSN was modified with AC through co-condensation (CMSCN) and post-grafting (PMSCN) method. MSCN has been characterized using X-Ray Diffraction, Nitrogen gas physisorption, Fourier Transform Infrared, Transmission Electron Microscope, and Field Emission Scanning Electron Microscope-Energy Dispersive X-Ray, while adsorption and release of drugs was observed using UV Spectrophotometer.  The characterization demonstrated that the MSN produces relatively higher number of pores with larger diameters while addition of AC led to the decrease in the crystallization, surface area and diameter of the pores. The contact angle test shows that MSN modified with AC displayed higher hydrophobicity. PMSCN showed highest adsorption percentage with 95.92%, followed by MSN and CMSCN with 90.88% and 82%, respectively. To investigate the release ability of DOX from DOX-MSN and DOX-MSCN, the release activity was conducted at 37 ° C, and in acidic pH solution, since the cancer cells were more acidic than normal cells. At pH 3.0, DOX released by MSN, PMSCN and CMSCN were 75.9%, 86.8% and 80.2%, respectively. While at pH 5.5, DOX released by MSN, PMSCN and CMSCN were 48.8%, 65.5% and 56.3% respectively. At pH 7.4, 27.8%, 30.4 and 28.8% of DOX were released by MSN, PMSCN and CMSCN, respectively. This study recommend carbon-functionalized MSN to be further studied for drug delivery application.