Conference Schedule

Day1: August 16, 2018

Keynote Forum

Biography

Rongjun Chen obtained his MSc Degree in Materials Science from Tsinghua University (P R China) in 2003; pursued PhD Degree at Cambridge University (UK) during the period 2003-2007, with focus on polymer drug delivery. He carried out his Postdoctoral Research at Cambridge University first on lyophilisation of pharmaceuticals and then on manufacture of clinical-grade lentiviral vectors for gene therapy during the period October 2006 to September 2009. In May 2013, he moved to Imperial College London as a Lecturer and is currently a Senior Lecturer since 2016. From October 2009 to April 2013, he started his independent academic career by taking a tenure-track faculty position as the Group Leader and BHRC Senior Translational Research Fellow at the University of Leeds. His research interests focuses on biomaterials, nanomedicine, drug delivery and cell therapy.

 

 


Abstract

It remains a major challenge to effectively deliver therapeutic agents, in particular macromolecules, through negatively charged lipid membrane barriers. It is the most limiting step preventing successful implementation of macromolecule-based cell modification and intracellular therapies. This is due to endosomal entrapment of macromolecules and their degradation in lysosomes. Many researchers have used cationic delivery systems to address this challenge. However, the positive charge could cause some issues, such as unfavorable biodistribution, rapid renal clearance and high non-specific cytotoxicity. ThiS presentation presents an alternative delivery strategy based on an anionic drug delivery platform. It covers our recent efforts on design and synthesis of novel anionic, viral-peptide-mimicking, pH-responsive, metabolite-derived polymers, and evaluation of their use in intracellular drug delivery in vitro and in vivo. Strict control over the size, structure, hydrophobicity-hydrophilicity balance and sequence of the polymers can effectively manipulate interactions with lipid membrane, cell and tissue models. It has been demonstrated that the biomimetic polymers can successfully traverse the extracellular matrix in three-dimensional multicellular spheroids, and also enable efficient loading of a wide range of macromolecules into the cell interior. This can represent a versatile delivery platform, suitable for targeted therapeutic delivery and cell therapy for treatment of various diseases including but not limited to cancer.

 

 

 

Biography

Andrew David Miller is well known as a leading Chemist Expert in the understanding and exploitation of molecular mechanisms in biology. The overall goal of his academic research has been and continues to be the design and creation of advanced therapeutics and diagnostics that address unmet medical need in the treatment of chronic diseases (such as cancer, diabetes, pain and some infectious diseases). From 1990-2010, he was a Member of academic staff in the Chemistry Department of Imperial College London (UK) where he founded the Imperial College Genetic Therapies Centre (GTC) in 1998, and became Full Professor of Organic Chemistry and Chemical Biology in 2002. Since 2010, he has been affiliated with King’s College London (UK) and more recently with the Veterinary Research Institute (VRI) in Brno, Czech Republic, where he is the Director of OPVVV Project FIT and its Key Foreign Scientist (KFS). He Co-Founded KP Therapeutics Ltd in 2016 with a pipeline of Precision Therapeutic Approaches (PTAs) in discovery & development for the diagnosis and treatment of chronic diseases. He has currently published nearly 250 papers, book chapters and alike, including at least 26 patents and patent applications. He is also Principal Writer of the first textbook of chemical biology “Essentials of Chemical Biology”, John Wiley & Sons.

 

 


Abstract

Precision Medicine is considered by many to be a necessary future for the treatment for all diseases. Fundamentally, this can be divided into two subsections, namely personalized medicine and precision therapeutics. With personalized medicine, the aim is to understand the genetic, immunological and/or metabolic individuality of patients in order to match individual patients with the most appropriate active pharmaceutical ingredients (APIs) for treatment of their particular disease(s). With precision therapeutics, the aim is t take control of the delivery of APIs to disease target tissue, by means of nanomedicine, and/or make use of select APIs that have extreme target specificity. The focus of this lecture is in precision therapeutics, as demonstrated by four worked examples of precision therapeutic approaches (PTAs) that are currently being taken forward in my laboratories and the laboratories of key collaborators for the treatment of chronic diseases. The chronic diseases of interest are chronic pain, epilepsy, cancer, non-alcoholic fatty liver disease (NAFLD) /diabetes type II, and infectious diseases such as influenza, Zika virus and HIV. By way of example, the right-hand side panel outlines a PTA for the treatment of cancer. In effect, a combination of bio-imaging and the application of image-guided targeting enable anti-cancer drug delivery nanoparticles to accumulate in a tumour lesion of choice and no obvious place elsewhere in the body. Accumulated nanoparticles may then release these anti-cancer drugs for local activity against tumour tissue saving other body tissues from unwanted exposure to these otherwise cytotoxic drugs. Implementation of such a PTA in the clinic could radically improve patient chemotherapy outcomes whilst reducing both required drug doses and side effects to an unprecedented degree. Such potential step changes in disease treatment explain why precision therapeutics should be an indispensable part of future medicine.

 

 

 

 

Biography

Kent Peterson is a graduate with an honors from Boston University’s Graduate School of Management, and a Member of American Mensa Society. He has lead Fluid Imaging Technologies since joining as the founder of the firm 12 years ago. The Company has sold over 600 FlowCams in over 52 countries. Ship-based FlowCam sys­tems have also been at work sampling from every ocean in the world. He has served on a number of boards and is active in community affairs. He has also been named Mainebiz Business Leader of the Year. His achievements include: Fluid Imaging Technologies’ Awards and recognitions include, the Governor’s Award for Business Excel­lence, the SBA New England Exporter of the Year Award, and the Portland Regional Chamber’s Robert R Masterton Award.

 

 


Abstract

Flow imaging microscopy has proven to be an important tool for the analysis of subvisible particulates in parenteral drugs. Now, due to the combined resolving power of blue LED light and patented oil immersion technology, flow imaging microscopes can image and analyze particles as small as 300 nm. The ability to detect transparent particles and differentiate them based on morphology yields significantly more detailed and accurate information than can be acquired using common laser diffraction and light obscuration techniques. Along with sophisticated statistical pattern recognition algorithms, these systems can be used to distinguish between different particulate types such as silicon oil, protein aggregates, and air bubbles. This presentation will present the techniques used to accomplish this.

 

 

 

Biography

Robert k Prud’homme is a professor in the Department of Chemical and Biological Engineering at Princeton University. He is the founding director of the Program in Engineering Biology. His research program focusses on polymer self-assembly applied to drug delivery. The development of Flash Nano Precipitation (FNP) in his laboratory enabled the encapsulation of poorly soluble drug compounds and oligonucleotides for therapy directed towards cancer, TB, and injections. FNP is a scalable and continuous process that is enables integrated processing and spray drying for low cost oral and aerosol formulations. Under sponsorship by the Bill and Melinda Gates Foundation, the process is being adopted to formulate new compounds coming from TBA, MMV, and DNDi.

 

 


Abstract

The intravenous delivery of composite gel microparticles (cGMPs) offers a platform for localized treatment of lung cancer. We describe a method for fabrication of cGMPs with average diameters of 35 to 100 μm using shear emulsification and microfluidic droplet generation. We characterized the particles and describe the performance of these particles in vivoBiodistribution of the cGMPs was selective to the lung after intravenous injection and particle clearance from the lung occurred in 7 weeks. One-week biodistribution studies demonstrated that larger, uniform particles produced by microfluidics provided optimal targeting to lung tissue. We demonstrated that highly loaded cGMPs containing a long wavelength fluorophore allow in vivo analysis of particle biodistribution without the need for ex-vivo organ analysis. The release of camptothecin conjugates from the nanopartricles, and thus, gel microparticles, is tuned from minutes to days by altering the polarity of the nanoparticle core.

 

 

Biography

Patrick J Sinko is a Pharmacist (BS, Rutgers 1982) and a Pharmaceutical Scientist (PhD, University of Michigan 1988). He joined Rutgers, The State University of New Jersey in 1991 and rose through the academic ranks where he is currently a Distinguished Professor (II) and the Parke-Davis Endowed Chair in Pharmaceutics and Drug Delivery in the Ernest Mario School of Pharmacy. He is the Principal Investigator of an active research laboratory that focuses on biopharmaceutics, pharmaceutical formulations and molecular-, nano- and micro-scale drug delivery with specific applications to the treatment or prevention of HIV/AIDS, breast, brain and lung cancerchemical terrorism countermeasures. He has received prestigious National Institutes of Health FIRST and MERIT awards and his lab has been continuously funded by the NIH for over 25 years.

 

 


Abstract

Ductal carcinoma in situ (DCIS) is a noninvasive breast cancer (BC) with possible microinvasions into the breast stroma. DCIS accounts for more than 16% of new BC diagnoses in women. DCIS progresses to Invasive Ductal Carcinoma (IDC) over time in 39-53% of patients, if left untreated. The vast majority of BC cases originate in the mammary duct. In this presentation, a nanoscale delivery system will be described that utilizes transpapillary delivery to achieve molecularly targeted, pathway-specific therapy in cancerous areas of the mammary duct. Our preliminary results with a nanosuspension of ciclopirox (CPX) in an orthotopic model of BC established the concept that sustained ductal exposure could completely suppress BC occurrence in vivo. For these studies polymeric NPs (nanoparticles) as well as lipid-polymer hybrid (LPH) NPs were the primary delivery vehicles. In order to achieve sustained precision treatment, HER2, transferrin receptor and/or EGFR were targeted using peptide ligands covalently bound to the surface of NPs. Ligand surface densities of 5% and 10% were evaluated and it was found that surface functionalized NPs enhanced binding and uptake into target cells. Cytoxicity was significantly increased with EGFR or TfR targeted NPs as compared to CPX alone or non-functionalized CPX-loaded NPs. A synergistic effect was observed when CPX was administered with gedatolisib, a PI3K/Akt/mTOR inhibitor resulting in a dose reduction index of ~6. In addition, the treatments were effective not only in BC cells but also cancer stem-like cells. Our efforts in addition to describing these studies and results, the engineering of the NPs to enhance ductal retention and specificity will also be described.

 

 

Biography

Simon C W Richardson is a Founder, Director and CSO at Intracellular Delivery Solutions Ltd, and Reader (Associate Professor) in Membrane Trafficking and Drug Delivery, at the University of Greenwich, UK. The driving theme behind his research is the intracellular delivery of antisense and RNAi to the cytosol. He is currently leading the Cell Biology Research Cluster within the Faculty of Engineering and Science, located at the Medway campus. His lab is currently working with several technologies based upon attenuated virulence factors that have very low in vitro toxicity profiles (and are minimally disruptive to the cell), and very high efficiency intracellular delivery profiles. We are also examining several methodologies to modulate protein stability and intracellular trafficking to aid the oral delivery of vaccines.

 

 


Abstract

Many protein toxins have evolved to access a variety of relatively inaccessible intracellular compartments in order to exert virulence. Counted among this number are proteins such as ricin toxin, shiga toxindiphtheria toxin and anthrax toxin. These proteins display diverse architecture ranging from AB5 to AB configurations and depending upon the specific B chain in question, entertain a number of strategies from direct membrane penetration to utilizing retrograde trafficking pathways to access a plethora of intracellular compartments including the cytosol. Typically the A chain will exhibit catalytic activity proportional to both cellular intoxication and virulence. However given the facile nature of protein recombination, attenuation is relatively simple. Here we describe the ability of attenuated anthrax toxin (ATx) to manipulate endocytic cargo sorting for the purposes of drug delivery, traversing intracellular compartmental boundaries for nucleic acid delivery. We report not only the efficiency with which siRNA and antisense effectors are delivered but also the mechanisms they utilize to traverse the barriers responsible for intracellular compartmentalization. Attenuated Atx:ASO complexes had transfection efficiency approximately equivalent to Nucleofection®. In HeLa cells, at 200 pmol ASO expression of the target gene was 5.4±2.0% relative to an untreated control after 24 h. Using 200 pmol ASOs, Nucleofection® reduced Synt5 expression to 8.1±2.1% after 24 h. PA:LFn-GAL4:ASO transfection of non- or terminally-differentiated THP-1 cells and Vero cells resulted in 35.2±19.1%, 36.4±1.8% and 22.9±6.9% (respectively) target gene expression after treatment with 200 pmol of ASO and demonstrated versatility. Nucleofection® with Stealth RNAi™ siRNA reduced HeLa Synt5 levels to 4.6±6.1% whereas treatment with the PA:LFn-PKR:siRNA resulted in 8.5±3.4% Synt5 expression after 24 h (HeLa cells). These data underscore the tractability of this approach to both antisense and siRNA delivery.

 

 

Biography

Vladimir P Torchilin, PhD, DSc is a University Distinguished Professor of Pharmaceutical Sciences and Director, Center for Pharmaceutical Biotechnology and Nanomedicine of Northeastern University, Boston, USA. His interests include drug delivery and targeting, nanomedicine, multifunctional and stimuli-sensitive pharmaceutical nanocarriersbiomedical polymers, experimental cancer therapy. He has published more than 400 original papers, more than 150 reviews and book chapters, has written and edited 12 books, and holds more than 40 patents. Google Scholar shows more than 52,000 citations of his papers with H-index of 102. He is Editor in Chief of Current Drug Discovery Technologies, Drug Delivery, and OpenNano; Co Editor of Current Pharmaceutical Biotechnology and on the Editorial Boards of many other journals. He received more than $30 M from the governmental and industrial sources in research funding. He has multiple honors and awards and in 2011, Times Higher Education ranked him number 2 among Top World Scientists in Pharmacology for the period of 2000-2010.

 

 


Abstract

Tumor therapy, especially in the case of multidrug resistant cancers, could be significantly enhanced by using siRNA down-regulating the production of proteins, which are involved in cancer cell resistance, such as Pgp or survivin. Even better response could be achieved is such siRNA could be delivered to tumors together with chemotherapeutic agent. This task is complicated by low stability of siRNA in biological surrounding. Thus, the delivery system should simultaneously protect siRNA from degradation. We have developed several types of lipid-core polymeric micelles based on PEG-phospholipid or PEI-phospholipid conjugates, which are biologically inert, demonstrate prolonged circulation in the blood and can firmly bind non-modified or reversibly-modified siRNA. Additionally, these Nano preparations can be loaded into their lipidic core with poorly water soluble chemotherapeutic agents, such as paclitaxel or camptothecin. In experiments with cancer cell monolayers, cancer cell 3D spheroids, and in animals with implanted tumors, it was shown that such co-loaded preparations can significantly down-regulate target proteins in cancer cells, enhance drug activity, and reverse multidrug resistance. This is illustrated by the efficient treatment of MDR (multi-drug resistance) cancer cells with combi-nations of siRNA-Pgp or siRNA-survivin stabilized in polymeric mixed mi-celles and doxorubicin, or tariquidar (Pgp inhibitor) and paclitaxel loaded into the same lipo-some or lipid-core polymeric micelle. In order to specifically unload such nanopreparations inside tumors, we made them sensitive to local tumor-specific stimuli, such as lowered pH, hypoxia, or overexpressed certain enzymes, such as matrix metalloproteases. Using pH-, redox-conditions, hypoxia-, or MMP2-sensitive bonds between different components of nanopreparations co-loaded with siRNA and drugs, we were able to make the systems specifically delivering biologically active agents in tumors, which resulted in significantly improved therapeutic response. We have also developed approaches to target individual intracellular organelles to initiate the apoptosis in resistant cancer cells.

 

 

Tracks

  • Nano Biomaterials | Nano Materials Synthesis and Characterisation | Nano Sensors | Advanced Nanomaterials | Nanotechnology and Biomedical Applications | Drug Delivery Research | Nanomedicine | Pharmaceutical Nanotechnology
Location: Dublin, Ireland

Biography

Kent Peterson is a graduate with an honor from Boston University’s Graduate School of Management, and a Member of American Mensa Society. He has lead Fluid Imaging Technologies since joining as the founder of the firm 12 years ago. The Company has sold over 600 FlowCams in over 52 countries. Ship-based FlowCam sys­tems have also been at work sampling from every ocean in the world. He has served on a number of boards and is active in community affairs. He has also been named Mainebiz Business Leader of the Year. His achievements include: Fluid Imaging Technologies’ Awards and recognitions include, the Governor’s Award for Business Excel­lence, the SBA New England Exporter of the Year Award, and the Portland Regional Chamber’s Robert R Masterton Award.

 

 


Abstract

Flow imaging microscopy has proven to be an important tool for the analysis of subvisible particulates in parenteral drugs. Now, due to the combined resolving power of blue LED light and patented oil immersion technology, flow imaging microscopes can image and analyze particles as small as 300 nm. The ability to detect transparent particles and differentiate them based on morphology yields significantly more detailed and accurate information than can be acquired using common laser diffraction and light obscuration techniques. Along with sophisticated statistical pattern recognition algorithms, these systems can be used to distinguish between different particulate types such as silicon oil, protein aggregates, and air bubbles. This presentation will present the techniques used to accomplish this.

 

 

Biography

Robert k Prud’homme is a professor in the Department of Chemical and Biological Engineering at Princeton University. He is the founding director of the Program in Engineering Biology. His research program focusses on polymer self-assembly applied to drug delivery. The development of Flash NanoPrecipitation (FNP) in his laboratory enabled the encapsulation of poorly soluble drug compounds and oligonucleotides for therapy directed towards cancer, TB, and injections. FNP is a scalable and continuous process that is enables integrated processing and spray drying for low cost oral and aerosol formulations. Under sponsorship by the Bill and Melinda Gates Foundation, the process is being adopted to formulate new compounds coming from TBA, MMV, and DNDi.

 

 

 


Abstract

The intravenous delivery of composite gel microparticles (cGMPs) offers a platform for localized treatment of lung cancer. We describe a method for fabrication of cGMPs with average diameters of 35 to 100 μm using shear emulsification and microfluidic droplet generation. We characterized the particles and describe the performance of these particles in vivoBiodistribution of the cGMPs was selective to the lung after intravenous injection and particle clearance from the lung occurred in 7 weeks. One-week biodistribution studies demonstrated that larger, uniform particles produced by microfluidics provided optimal targeting to lung tissue. We demonstrated that highly loaded cGMPs containing a long wavelength fluorophore allow in vivo analysis of particle biodistribution without the need for ex-vivo organ analysis. The release of camptothecin conjugates from the nanopartricles, and thus, gel microparticles, is tuned from minutes to days by altering the polarity of the nanoparticle core.

 

 

Biography

Patrick J Sinko is a Pharmacist (BS, Rutgers 1982) and a Pharmaceutical Scientist (PhD, University of Michigan 1988). He joined Rutgers, The State University of New Jersey in 1991 and rose through the academic ranks where he is currently a Distinguished Professor (II) and the Parke-Davis Endowed Chair in Pharmaceutics and Drug Delivery in the Ernest Mario School of Pharmacy. He is the Principal Investigator of an active research laboratory that focuses on biopharmaceutics, pharmaceutical formulations and molecular-, nano- and micro-scale drug delivery with specific applications to the treatment or prevention of HIV/AIDS, breast, brain and lung cancer, chemical terrorism countermeasures. He has received prestigious National Institutes of Health FIRST and MERIT awards and his lab has been continuously funded by the NIH for over 25 years.

 

 


Abstract

Ductal carcinoma in situ (DCIS) is a noninvasive breast cancer (BC) with possible microinvasions into the breast stroma. DCIS accounts for more than 16% of new BC diagnoses in women. DCIS progresses to Invasive Ductal Carcinoma (IDC) over time in 39-53% of patients, if left untreated. The vast majority of BC cases originate in the mammary duct. In this presentation, a nanoscale delivery system will be described that utilizes transpapillary delivery to achieve molecularly targeted, pathway-specific therapy in cancerous areas of the mammary duct. Our preliminary results with a nanosuspension of ciclopirox (CPX) in an orthotopic model of BC established the concept that sustained ductal exposure could completely suppress BC occurrence in vivo. For these studies polymeric NPs (nanoparticles) as well as lipid-polymer hybrid (LPH) NPs were the primary delivery vehicles. In order to achieve sustained precision treatment, HER2, transferrin receptor and/or EGFR were targeted using peptide ligands covalently bound to the surface of NPs. Ligand surface densities of 5% and 10% were evaluated and it was found that surface functionalized NPs enhanced binding and uptake into target cells. Cytoxicity was significantly increased with EGFR or TfR targeted NPs as compared to CPX alone or non functionalized CPX-loaded NPs. A synergistic effect was observed when CPX was administered with gedatolisib, a PI3K/Akt/mTOR inhibitor resulting in a dose reduction index of ~6. In addition, the treatments were effective not only in BC cells but also cancer stem-like cells. Our efforts in addition to describing these studies and results, the engineering of the NPs to enhance ductal retention and specificity will also be described.

 

 

Biography

Simon C W Richardson is a Founder, Director and CSO at Intracellular Delivery Solutions Ltd, and Reader (Associate Professor) in Membrane Trafficking and Drug Delivery, at the University of Greenwich, UK. The driving theme behind his research is the intracellular delivery of antisense and RNAi to the cytosol. He is currently leading the Cell Biology Research Cluster within the Faculty of Engineering and Science, located at the Medway campus. His lab is currently working with several technologies based upon attenuated virulence factors that have very low in vitro toxicity profiles (and are minimally disruptive to the cell), and very high efficiency intracellular delivery profiles. We are also examining several methodologies to modulate protein stability and intracellular trafficking to aid the oral delivery of vaccines.

 

 


Abstract

Many protein toxins have evolved to access a variety of relatively inaccessible intracellular compartments in order to exert virulence. Counted among this number are proteins such as ricin toxinshiga toxin, diphtheria toxin and anthrax toxin. These proteins display diverse architecture ranging from AB5 to AB configurations and depending upon the specific B chain in question, entertain a number of strategies from direct membrane penetration to utilizing retrograde trafficking pathways to access a plethora of intracellular compartments including the cytosol. Typically the A chain will exhibit catalytic activity proportional to both cellular intoxication and virulence. However given the facile nature of protein recombination, attenuation is relatively simple. Here we describe the ability of attenuated anthrax toxin (ATx) to manipulate endocytic cargo sorting for the purposes of drug delivery, traversing intracellular compartmental boundaries for nucleic acid delivery. We report not only the efficiency with which siRNA and antisense effectors are delivered but also the mechanisms they utilize to traverse the barriers responsible for intracellular compartmentalization. Attenuated Atx:ASO complexes had transfection efficiency approximately equivalent to Nucleofection®. In HeLa cells, at 200 pmol ASO expression of the target gene was 5.4±2.0% relative to an untreated control after 24 h. Using 200 pmol ASOs, Nucleofection® reduced Synt5 expression to 8.1±2.1% after 24 h. PA:LFn-GAL4:ASO transfection of non- or terminally-differentiated THP-1 cells and Vero cells resulted in 35.2±19.1%, 36.4±1.8% and 22.9±6.9% (respectively) target gene expression after treatment with 200 pmol of ASO and demonstrated versatility. Nucleofection® with Stealth RNAi™ siRNA reduced HeLa Synt5 levels to 4.6±6.1% whereas treatment with the PA:LFn-PKR:siRNA resulted in 8.5±3.4% Synt5 expression after 24 h (HeLa cells). These data underscore the tractability of this approach to both antisense and siRNA delivery.

 

 

Biography

Vladimir P Torchilin, PhD, DSc is a University Distinguished Professor of Pharmaceutical Sciences and Director, Center for Pharmaceutical Biotechnology and Nanomedicine of Northeastern University, Boston, USA. His interests include drug delivery and targeting, nanomedicine, multifunctional and stimuli-sensitive pharmaceutical nanocarriersbiomedical polymers, experimental cancer therapy. He has published more than 400 original papers, more than 150 reviews and book chapters, has written and edited 12 books, and holds more than 40 patents. Google Scholar shows more than 52,000 citations of his papers with H-index of 102. He is Editor in Chief of Current Drug Discovery Technologies, Drug Delivery, and OpenNano; Co Editor of Current Pharmaceutical Biotechnology and on the Editorial Boards of many other journals. He received more than $30 M from the governmental and industrial sources in research funding. He has multiple honors and awards and in 2011, Times Higher Education ranked him number 2 among Top World Scientists in Pharmacology for the period of 2000-2010.

 

 


Abstract

Tumor therapy, especially in the case of multidrug resistant cancers, could be significantly enhanced by using siRNA down-regulating the production of proteins, which are involved in cancer cell resistance, such as Pgp or survivin. Even better response could be achieved is such siRNA could be delivered to tumors together with chemotherapeutic agent. This task is complicated by low stability of siRNA in biological surrounding. Thus, the delivery system should simultaneously protect siRNA from degradation. We have developed several types of lipid-core polymeric micelles based on PEG-phospholipid or PEI-phospholipid conjugates, which are biologically inert, demonstrate prolonged circulation in the blood and can firmly bind non-modified or reversibly-modified siRNA. Additionally, these nanopreparations can be loaded into their lipidic core with poorly water soluble chemotherapeutic agents, such as paclitaxel or camptothecin. In experiments with cancer cell monolayers, cancer cell 3D spheroids, and in animals with implanted tumors, it was shown that such co-loaded preparations can significantly down-regulate target proteins in cancer cells, enhance drug activity, and reverse multidrug resistance. This is illustrated by the efficient treatment of MDR (multi-drug resistance) cancer cells with combi-nations of siRNA-Pgp or siRNA-survivin stabilized in polymeric mixed mi-celles and doxorubicin, or tariquidar (Pgp inhibitor) and paclitaxel loaded into the same lipo-some or lipid-core polymeric micelle. In order to specifically unload such nanopreparations inside tumors, we made them sensitive to local tumor-specific stimuli, such as lowered pH, hypoxia, or overexpressed certain enzymes, such as matrix metalloproteases. Using pH-, redox-conditions, hypoxia-, or MMP2-sensitive bonds between different components of nanopreparations co-loaded with siRNA and drugs, we were able to make the systems specifically delivering biologically active agents in tumors, which resulted in significantly improved therapeutic response. We have also developed approaches to target individual intracellular organelles to initiate the apoptosis in resistant cancer cells.

 

 

Biography

Sergey Suchkov graduated from Astrakhan State Medical University and was awarded with MD; in 1985 maintained his PhD at the I M Sechenov Moscow Medical Academy and in 2001, maintained his Doctor Degree at National Institute of Immunology , Russia. From 1987 through 1989 was a Senior Researcher at Koltzov Institute of Developmental Biology. From 1989 through 1995, he was the Head of the Lab of Clinical Immunology, Helmholtz Eye Re-search Institute in Moscow. From 1995 through 2004, a Chair of the Department for Clinical Immunology, Moscow Clinical Research Institute (MONIKI). He has been trained at: Na-tional Institute of Health; Wills Eye Hospital, Pennsylvania, USA; University of Florida in Gainesville; University of California San Francisco; Johns Hopkins University, Baltimore, MD, USA respectively. 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, he is a Chair, Department 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; European Association for Predictive, Preventive and Personalized Medicine (EPMA), Belgium; American Association for Research in Vision and Ophthalmology (ARVO); ISER (International Society for Eye Research); Personalized Medicine Coalition (PMC), Washington, USA.

 

 


Abstract

Catalytic Abs (catAbs) are multivalent im-munoglobulins (Igs) with a capacity to hy-drolyze the antigenic (Ag) substrate. In this sense, proteolytic Abs (Ab-proteases) repre-sent Abs to provide proteolytic effects. Abs against myelin basic protein/ MBP with pro-teolytic activity exhibiting sequence-specific cleavage of MBP are of great value to moni-tor demyelination whilst in MS. The activity of Ab-proteases was first registered at the subclinical stages 1-2 years prior to the clin-ical illness. And the activity of the Ab-proteases revealed significant correlation with scales of demyelination and the disabil-ity of the patients as well. So, the activity of Ab-proteases and its dynamics tested would confirm a high subclinical and predictive (translational) value of the tools as applicable for personalized monitoring protocols. Of tremendous value are Ab-proteases directly affecting remodeling of tissues with multilevel architectonics (for instance, mye-lin). By changing sequence specificity one may reach reduction of a density of the neg-ative proteolytic effects within the myelin sheath and thus minimizing scales of demye-lination. Ab-proteases can be programmed and re-programmed to suit the needs of the body metabolism or could be designed for the development of new catalysts with no natural counterparts. Further studies are needed to secure artificial or edited Ab-proteases as translational tools of the newest generation to diagnose, to monitor, to con-trol and to treat and rehabilitate MS patients at clinical stages and to prevent the disorder at subclinical stages in persons-at-risks to secure the efficacy of regenerative manipula-tions.

 

 

Day2: August 17, 2018

Keynote Forum

Biography

Sergey Suchkov was born in the City of Astrakhan, Russia. In 1980, graduated from Astrakhan State Medical University and was awarded with MD. In 1985, maintained his PhD at the I.M. Sechenov Moscow Medical Academy and Inst of Med Enzymology. In 2001, and then his Doctor Degree at the Nat Inst of Immunology in Russia. From 1989 through 1995, was being a Head of the Lab of Clin Immunology, Helmholtz Eye Research Inst in Moscow. From 1995 through 2004 - a Chair of the Dept for Clin Immunology, Moscow Clin Research Institute (MONIKI). In 1993-1996, 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.At present, Dr Sergey Suchkov, MD, PhD

 

 


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 PM concept, it is necessary to create a fundamentally new strategy based upon the subclinical recognition of bio predictors 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 health care resources. A lack of medical guidelines has been identified by the majority of 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

Pauline Y Lau has been working in medical industry for over 35 years with experiences in both pharmaceutical and in-vitro medical device (IVD). Her accomplishment is well recognized by the international societies in the last 20 years. Suntec Medical Inc., USA (pharmaceutical company) under her leadership has successfully registered pharmaceutical products globally. Currently, the company is developing a portfolio of cancer therapies including immuno-oncology and targeted therapies utilizing its proprietary MNC technology platform. She is global Expert and accomplished Researcher in Precision Medicine. She was an Executive Director in Integrated Medicine at Roche Pharmaceuticals.

 

 


Abstract

Statement of the Problem: A common issue for anti-cancer drugs are the strong toxicity and low efficacy of most therapies. This is especially an issue to many immunotherapies such as IL12. This is caused by not enough drugs enter tumor tissues to have satisfactory efficacy, and left majority of the drugs enter normal tissues to cause high toxicities to the normal tissues. Target drug delivery technologies such as liposomes are effective in improving the therapeutic index (efficacy/toxicity) of small molecular chemotherapies. But, no effective technology has been demonstrated to effectively enhancing the therapeutic index of protein drugs including monoclonal antibody targeted therapies.

Breakthrough in Cancer Treatment: A new nanotechnology, MNC technology, has been dramatically improved the therapeutic index of all classes of anti-cancer drugs. Successful applications are demonstrated by Trastuzumab (monoclonal antibody), Interferon alpha (immunotherapy) and Sunitinib (chemotherapy).

Principle of Technology: MNC technology is a micelle nanocomplex that can reversably bind to drugs, target deliver majority of the drug to the tumor site plus effectively slow release of the drug from circulation to tumor to enhance the full usage of the drug and reduce toxicity.

 

 

Tracks

  • Nano Applications | Nano Science and Technology | Medical Applications in Nanotechnology | Biomarkers and Personalized Medicine | Cancer and Nanotechnology | Nanomaterials for drug delivery | Novel Drug Delivery Systems
Location: Dublin, Ireland

Biography

Muthu Madaswamy Sona earned his Bachelor’s Degree in Pharmacy in 2002; Master’s Degree in Pharmaceutical Technology from India in 2004 and PhD Degree in Pharmaceutics from IIT, Varanasi, India in 2009. He did his Postdoctoral trainings in the Department of Chemical Engineering at National University of Singapore as a Recipient of Boyscast Fellowship and CREST Award from India. He is also an Awardee of DST Young Scientist in 2012. His research interest is to develop advanced nanomedicine as novel platform for diagnosis and therapy. He has authored over 64 peer-reviewed publications with a cumulative impact factor of >240, citation of 2100 and h-index of 24.

 


Abstract

Nanotheranostics have shown the development of advanced platforms that can diagnose cancer at early stages, initiate first-line therapy, monitor it, and if needed, rapidly start subsequent treatments. In nanotheranostics, therapeutic and diagnostic agents are loaded with nanomedicine in a single theranostic platform, which can be further developed as clinical formulations for targeting different types of cancer. This speech is concerned about theranostic micelles developed using TPGS (tocopheryl polyethylene glycol succinate), docetaxel and gold nanoclusters for the early diagnosis and therapy of cancer with advanced features. Micelles are amphiphilic spherical nanostructures consisting of hydrophilic shell and hydrophobic core. Micelles have advantages such as thermodynamic stability, kinetic stability, higher payload and smaller dimension (less than 50 nm). In our group, various research studies were done on targeted micelles for cancer diagnosis and therapy. In future, nanotheranostics will be able to provide personalized treatment which can make cancer even curable or at least treatable at the earliest stage.

 

Biography

Lorenz De Neve started his carrier as a Researcher with his master thesis on the sorption behavior of cationic surfactants. During this period he obtained experience concerning the preparation and characterization of liposomal dispersions, including viscometry using rotational viscometers, submicron particle sizing by dynamic light scattering and adsorption analysis by both QCM-D and by the traditional depletion technique. Currently he is conducting research on pharmaceutical nanosuspension formulations. More specifically the purpose of his research is to enlarge the fundamental knowledge of the link between the formulation parameters and the macroscopic properties of nanosuspensions and to understand the interactions between the different formulation parameters.

 


Abstract

Poly(ethylene oxide)-poly(propylene oxide)-poly(ethylene oxide) block copolymer surfactants (poloxamers or pluronics) are used as stabilizer in various nanosuspensions, e.g. of rilpivirine, danazol, diclofenac, asulacrine and itraconazole. In order to have a stabilizing effect on hydrophobic particles, these PEO-PPO-PEO surfactants should adsorb to the particle surface. In this research, the adsorption behavior of pluronics with two different ethylene oxide contents (50% and 80%) and three different molecular weights of the propylene oxide part (i.e. 950, 1750 and 3250 g/mol) was studied at 20°C and 37°C onto gold sensors coated with 1-undecanethiol using a quartz crystal microbalance with dissipation (QCM-D). Pluronic solutions with 5 different concentrations were used, ranging from 0.02 mg/ml to 50 mg/ml. Our results indicate a significant (linear) effect of the pluronic concentration on the average adsorption during the adsorption steps. No clear effect could, however, be detected after rinsing of the sensors with ultrapure water. The molecular weight of the PPO part seemed to have a proportional effect on the adsorbed amounts after rinsing, but no clear effect during the adsorption steps. The ethylene oxide content seemed to have an effect during both the adsorption and rinsing steps. Also, our results indicated no significant difference in the average adsorbed amount during both the adsorption and rinsing steps at 20°C and 37°C. The obtained results were useful to gain more insight in the stability differences between nanosuspensions with different pluronic concentrations (and molecular structure).