52
III International Conference on & CLINICAL SCIENCES DRUG DISCOVERY November 9-11 2017 Sheraton Reston Hotel 11810 Sunrise Valley Dr, Reston VA 20191 USA UNITED Scientific Group CSDD-2017

CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

III International Conference on

&CLINICAL SCIENCES DRUG DISCOVERY

November 9-11

2017

Sheraton Reston Hotel11810 Sunrise Valley Dr, Reston

VA 20191USA

UNITED Scientific Group CSDD-2017

Page 2: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Notes

Page 3: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Index

Keynote Presentations .......07 - 11

.......12 - 44Featured Presentations

.......45 - 49Poster Presentations

.......50 - 51About Organizer

.......04Key Concepts

.......05Committee Members

III International Conference on

&CLINICAL SCIENCES DRUG DISCOVERY

November 9-11, 2017

Page 4: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

◆ Drug Discovery in Preclinical Research

◆ Drug Design: Novel Approaches

◆ Small Molecules for Cancer Immunotherapy

◆ Drug Development & Delivery System

◆ Computer Aided Drug Designing (CADD)

◆ Anti-Infectives

◆ Clinical Trials and Regulatory Affairs

◆ Cancer Targeted Drug Delivery

◆ Cardiovascular Drug Discovery & Therapy

◆ Chemistry & Medical Imaging

◆ CNS Drug Discovery & Therapy

◆ Diabetes and Obesity Drug Discovery & Therapy

◆ Inflammation and Immunology

◆ Innovative Drug Discovery and Nanotechnology

◆ Biomarkers

◆ Nutraceutical Drug Discovery & Therapy

◆ Pharmaceutical Biotechnology

◆ Protein and Peptide Sciences

◆ Regenerative Medicine

◆ Structural & System Biology

◆ Translational Medicine

◆ Bio-therapeutics

Key Concepts

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 4

Page 5: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Harold C. SmithUniversity of Rochester

USA

Brent E. KorbaGeorgetown University

USA

Nikolai ZhelevAbertay University

UK

Alan D. RogolUniversity of Virginia Medical

School, USA

Steven LaPlanteNMX Research and Solutions Inc.

Canada

Mahin KhatamiNIH (Retired)

USA

Committee Members

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 5

Page 6: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

1November 9

Thursday

Page 7: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

New paradigms in GPCR drug discovery

Kenneth A. Jacobson

Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, Bethesda, MD, USA

Abstract

G protein-coupled receptors (GPCRs) remain a major domain of pharmaceutical discovery. The identification of GPCR lead compounds and their optimization are now structure-based, thanks to advances in X-ray crystallography, molecular modeling, protein engineering and biophysical techniques. Roughly 200 high resolution GPCR structures have already been reported, in complex with agonists, antagonists and allosteric modulators. In silico screening provides useful hit molecules and modeling guides their optimization. New pharmacological approaches to tuning the pleotropic action of GPCRs include: allosteric modulators, biased ligands, GPCR heterodimer-targeted compounds, manipulation of polypharmacology, receptor antibodies and tailoring of drug molecules to fit GPCR pharmacogenomics. Measurements of kinetics and drug efficacy are factors influencing clinical success. With the exception of inhibitors of GPCR kinases, targeting of intracellular GPCR signaling or receptor cycling for therapeutic purposes remains a futuristic concept. New assay approaches are more efficient and multidimensional: cell-based, label-free, fluorescence-based assays, and biosensors. Tailoring GPCR drugs to a patient’s genetic background is now being considered. New imaging technology visualizes drug action in vivo. Thus, there is reason to be optimistic that new technology for GPCR ligand discovery will help reverse the current narrowing of the pharmaceutical pipeline.

Biography

Dr. Kenneth A. Jacobson, Ph.D. is Chief of the Laboratory of Bioorganic Chemistry and the Molecular Recognition Section at the National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health in Bethesda, Maryland, USA. Dr. Jacobson is a medicinal chemist with interests in the structure and pharmacology of G protein-coupled receptors, in particular receptors for adenosine and for purine and pyrimidine nucleotides. Dr. Jacobson has served as Chair of the Medicinal Chemistry Division of the American Chemical Society and was inducted into the ACS MEDI Division’s Hall of Fame in 2009.

Keynote Presentations

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 7

Page 8: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 8

Targeted imaging and therapeutic agents for cancer, autoimmune and inflammatory diseases

Philip S. Low

Purdue University, West Lafayette, IN, USA

Abstract

We have developed small molecule ligands for use in targeting attached drugs to pathologic cells, thereby avoiding collateral toxicity to healthy cells. In the case of cancer, we began by exploiting up-regulation of the folate receptor on cancers of the ovary, lung, kidney, endometrium and breast to target imaging and therapeutic agents to these cancers. Clinical trials of six folate-linked drugs demonstrate that the ligand-targeting strategy holds promise for increasing drug potency while reducing unwanted toxicity. Data on treatment of tumor-bearing mice, dogs, and humans will be presented.

We have also developed low molecular weight targeting ligands to deliver attached drugs selectively to cancers that over-express prostate specific membrane antigen (PSMA), CCK2 receptor (GI tract cancers), neurokinin 1 receptor (neuroendocrine cancers), carbonic anhydrase IX (kidney cancers and all hypoxic tumors), and several other tumor-specific receptors. Imaging and therapeutic studies suggest that these targeting ligands can not only improve the diagnosis of the respective cancers, but also enhance treatment of these malignant diseases. Recent pre-clinical and/or clinical data on these new targeting ligands confirm this anticipation.

Additional cancer-specific ligands that target malignancies of the bladder, pancreas, stomach, brain, liver, colon, skin and esophagus are also under development. Moreover, use of low molecular weight targeting ligands to “light up” cancer tissues with tumor-targeted fluorescent dyes during surgeries will be described, with videos of several of these surgeries being presented.

Finally, ligand-targeted imaging and therapeutic agents for a number of autoimmune, inflammatory, and infectious diseases (e.g. malaria, rheumatoid arthritis, multiple sclerosis, psoriasis, atherosclerosis, osteoarthritis, etc.) will also be described.

Biography

Dr. Philip S. Low is a Presidential Scholar in Drug Discovery and Corley Distinguished Professor of Chemistry. Dr. Low has spent >40 years exploring 1) targeted therapeutic and imaging agents for human diseases (including cancers, autoimmune, and infectious diseases) and 2) the structure, function, and pathologies of the erythrocyte membrane. He has published >400 scientific articles and has >60 US patents/patents pending. Eight drugs stemming from his research are undergoing human clinical trials and three companies (Endocyte Inc., OnTarget Laboratories Inc., and HuLow LLC) have been founded to commercialize these discoveries. Dr. Low received his B.S. in Chemistry from Brigham Young University and his Ph.D. in Biochemistry from UCSD.

Page 9: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 9

The comprehensive solution to characterize and treat every disease

Ram Samudrala

University at Buffalo, Buffalo, NY, USA

Abstract

Traditional drug discovery is focused on single targets (molecule/disease) and devising broadly applicable treatments.  Typically, an essential molecule responsible for pathogenesis is targeted so as to completely inhibit its function, and then determine side effects/toxicity of the inhibitor.  The exponentially decreasing efficiency of new drug discovery illustrates the limitations of the traditional paradigm.  Our solution is the Computational Analysis of Novel Drug Opportunities (CANDO) platform that screens and rank every existing human use drug or compound for every disease/indication, using structural bioinformatics tools developed as part of our basic research on multiscale modelling of protein and proteome structure, function, interaction, evolution, and design.  The shotgun screening is made intelligent and tailored to individual genotypic and phenotypic features, such as mutations in DNA, ethnicity, sex, age, weight, etc.  Given an individual’s genomic sequence, and other phenotypic features or laboratory results, the platform outputs a set of optimal drug regimens for any indication.  As new drugs are developed or new indications arise, the platform can immediately evaluate the efficacy of a new drug against all existing indications, or the efficacy of all existing drugs against a new indication, in any arbitrary context, which is a marked departure from the current single target focused approach. Information from the preliminary validations obtained is fed back into the platform to increase its accuracy. Similarly, other novel types of genotypic and phenotypic data can be fed back into platform to make it increasingly precise, with a tremendous incalculable impact on our health and quality of life.

Biography

Dr. Ram Samudrala is Professor and Chief, Division of Bioinformatics, University at Buffalo (2014-present) researching multiscale modelling of protein and proteome structure, function, interaction, design, and evolution.  His work has led to more than 130 publications in journals such as Science, Nature, PLoS Biology, PNAS, and JAMA.  Dr. Samudrala joined the faculty of the University of Washington (2001-2014) after completing his postdoctoral work with Michael Levitt (2013 Chemistry Nobel) at Stanford University.  His many honors include a Searle Scholarship (2002), a top young innovator (TR100) selection by MIT Technology Review (2003), NSF CAREER Award (2005), and the NIH Director’s Pioneer Award (2010).

Page 10: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 10

Chemistry and health: now is the time for a comprehensive approach

Stephen A. Matlin1*, Goverdhan Mehta2, Alain Krief3 and Henning Hopf4

1Institute for Global Health Innovation, Imperial College London, UK2School of Chemistry, University of Hyderabad, India3International Organization for Chemical Sciences in Development, Belgium4Institute of Organic Chemistry, Technische Universität Braunschweig, Germany

Abstract

Chemistry has made many outstanding contributions to health that have greatly enhanced both the quality and length of life of human beings during the last 1-2 centuries. Health benefits include effective and safe medicines and anesthetic agents, relief of pain and suffering, prosthetics to improve impaired functions, better nutrition, fertility regulation and a cleaner and safer environment. In chemistry terms, an extremely rich variety of examples can be drawn from diverse fields where chemistry plays a fundamental role – including, among others: medicinal and pharmaceutical chemistry and pharmacology; analytical and clinical chemistry; biological and nutritional chemistry; materials science; and physical chemistry.

There are major current and emerging global threats to health that require strong contributions from chemistry. However, while chemistry’s potential to go on making contributions is huge: its actual capacity is constrained by a number of current systemic factors and threats, so that its delivery is likely to be substantially less than optimal.

The most serious threats involve three systemic fragmentations: (1) in the discipline; (2) in the industrial sector; and (3) in the regulatory sector. The diversity and scale of problems and challenges is such that piecemeal fixes are insufficient – a comprehensive overall approach is required, that employs ‘systems thinking’ and engages widescale, systemic reform to achieve ambitious goals in ‘chemistry for better health’.

The analysis of systemic fragmentations and essential elements of the comprehensive approach we propose to overcome them will be presented in the talk.

Biography

Dr. Stephen Matlin is Secretary and Head of Strategic Development for the International Organization for Chemical Sciences in Development (IOCD) and Adjunct Professor in the Institute of Global Health Innovation, Imperial College London.

Page 11: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 11

Cancer a severe allergic response in tissue? Histamine role in immune dysfunction toward multistep tumorigenesis and angiogenesis, accidental discoveries

Mahin KhatamiInflammation and Cancer Research, Retired, NCI/NIH, Bethesda, MD, USA

AbstractOngoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is

a cause of carcinogenesis (‘foe’) have been very costly for the public in the last century. Failure rates of claimed ‘targeted’ therapies, ‘precision’ or ‘personalized’ medicine for solid tumors are 90 (+/-5%). In 1980’s we developed experimental models of acute and chronic inflammatory diseases in ocular tissues. Recent analyses of data suggest the first series of evidence for direct association between inflammation and developmental phases of immune dysfunction in conjunctival-associated lymphoid tissues (CALTs). At least three phases of inflammation-induced interactions and synergies between resident (host) and recruited immune cells were identified: (a), acute phase, strong clinical reactions, activation of mast cells (MCs) and B/ plasma cells, biosynthesis of IgE Abs, release of histamine and prostaglandin PGF1α; (b), intermediate phase, down-regulation phenomenon,, exhausted/degranulated MCs, heavy eosinophils (Eos) infiltrations into epithelium and mucus secreting goblet cells (GCs), and (c), chronic phase; induction of lymphoid hyperplasia, activated macrophages (MΦs), loss of integrity of lymphoid tissue capsular membrane, presence of histiocytes (activited dendritic cells), follicular and germinal center formation, increased ratios of local IgG1/IgG2, epithelial thickening and/or thinning.

Systematic studies to monitor early events and interactions between resident and recruited cells in site-specific tissues should provide key information for identification of the loss of immune surveillance that could be preventable, reversible or drugable. Promotion of Yin and Yang that we describe for effective immunity would help making informed decisions for effective preventive and therapeutic approaches to cancer and for universal safe vaccines development as well as accurate risk assessment formula toward improvement of public health.Selected References

Khatami M: In: Cancer research and therapy: scam of century–promote immunity [Yin-Yang]. ISBN-10:153043100X; ISBN-13: 978–1530431007; Amazon-Createspace. pp 1–166, 2016.https://www.createspace.com/6123573

Khatami M. “Yin and Yang” in inflammation: duality in innate immune cell function and tumorigenesis. Expert Opin Biol Ther. 2008;8:1461–1472.

Khatami M. Unresolved inflammation: loss of natural cancer killing ability of immunity as correct ‘target’ for therapy: seeing the ‘elephant’ in the light. Cell Biochem Biophys. 2012; 62:501–509.

Khatami M. Is cancer a severe delayed hypersensitivity reaction and histamine a blueprint? Clin Transl Med 2016 Dec; 5(1):35. doi: 10.1186/s40169-016-0108-3.

Khatami M. Safety concerns and hidden agenda behind HPV vaccines: another generation of drug-dependent society? Clinical and Translational Medicine.

BiographyDr. Mahin Khatami received her Ph.D. in Molecular Biology from Univ. PA (1980). She was a research

faculty at Univ. PA, involved in cell biology of diabetes complications and ocular inflammatory diseases. In 1998, at NCI/NIH extension of her discoveries and efforts to promote role of inflammation in cancer research met with serious opposition. Currently, topic of inflammation in cancer research and therapy is the focus of numerous funded projects. She authored over 100 articles, book chapters and proceedings. She has lectured internationally; was President/VP- GWIS; member of scientific and editorial organizations. In 2012, she edited 2 books on inflammatory diseases, aging, cancer and therapies.

Page 12: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 12

Technical Session – I

Evaluating CNS pharmacology to improve drug delivery to brain tumors

Sadhana JacksonNational Cancer Institute/National Institutes of Health, MD, USA

AbstractThe blood-brain barrier (BBB) severely limits the entry of systemically administered drugs including

chemotherapy. Few studies have centered on identifying agents which can transiently increase drug delivery across the BBB with close tracking via neuropharmacokinetics. In rodents, regadenoson (adenosine A2A receptor agonist) caused brief BBB disruption and increased drug concentrations in normal brain. Our studies evaluated regadenoson’s ability to increase intra-tumoral temozolomide concentrations in patients with glioblastoma.

Patients scheduled for a clinically indicated surgery for recurrent glioblastoma were eligible for this study. Microdialysis catheters (MDC) were placed intraoperatively in brain tissue. On post-operative days #1 and 2, patients received oral temozolomide (150mg/m2). On day #2, they received one intravenous dose of regadenoson (0.4mg). Blood and MDC samples were collected to determine temozolomide concentrations via LC-MS/MS.

Six patients were enrolled. Five patients had no complications from MDC placement or regadenoson; with successful collection of blood and dialysate samples. Mean plasma AUC was 16.4±1.4 hr*µg/ml for temozolomide alone and 16.6±2.87 hr*µg/ml with addition of regadenoson. Mean dialysate AUC was 2.9±1.2 hr*µg/ml with temozolomide alone and 3.0±1.7 hr*µg/ml with regadenoson. Mean brain:plasma AUC ratio was 18.0±7.8% and 19.1±10.7% for temozolomide alone and with regadenoson respectively. Peak concentration and Tmax in brain were not significantly different.

Although previously shown to be efficacious in rodents to increase BBB drug entry, our data suggest that regadenoson does not increase temozolomide concentrations in brain. Further studies exploring alternative regadenoson dosing and schedules are needed; as well as understanding differences of glioma vascular biology in an effort to enhance BBB drug delivery.

BiographyDr. Sadhana Jackson is an assistant clinical investigator within the Neuro-Oncology Branch.  Her research

centers on evaluating the blood-brain barrier of malignant gliomas, to enhance chemotherapy delivery.  She has experience with brain microdialysis, pharmacokinetic analysis, and studying CNS pharmacology of tumor models. Dr. Jackson received her medical doctorate from Eastern Virginia Medical School, completed her pediatric residency at Orlando Health and then pediatric hematology/oncology fellowship at St. Jude Children’s Research Hospital.  She concluded her training at Johns Hopkins in concurrent pediatric neuro-oncology and clinical pharmacology fellowships and has been at the NIH since 2015.

Featured Presentations

Page 13: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 13

Toward a functional cure for HIV/AIDS: regulatory interactions between RNA and the HIV restriction factor APOBEC3G

Harold C. Smith1*, Bogdan Polevoda1 and Ryan P. Bennett2

1Department of Biochemistry and Biophysics, University of Rochester, School of Medicine and Dentistry, NY, USA2OyaGen, Inc Rochester, NY, USA

Abstract APOBEC3G (A3G) is a member of the AID/APOBEC protein family of cytidine deaminases that bind to

nucleic acids. A3G mutates viral genomic DNA by deaminating dC to dU during reverse transcription, leading to accumulation of mutations that can inactivate the virus. RNA binding to A3G is essential for A3G assembly with virions and antiviral activity. However, binding to bulk cellular RNAs inhibits A3G binding to substrate DNA and antiviral mutagenic activity. Mass spectroscopy (MS) revealed that RNA and ssDNA bind to the same three A3G tryptic peptides (amino acids 181–194, 314–320 and 345–374). MS also revealed A3G peptides that uniquely bound to 7SL1 ncRNA and HIV genomic RNA involved in packaging A3G in virions. Y315A mutation alone significantly reduced A3G binding to both ssDNA and bulk RNA whereas Y181A and Y182A mutations only moderately affected A3G nucleic acid binding. Y315A mutant also showed markedly reduced ability to packaging into viral particles and lost most of its antiviral activity. In uninfected cells RNA binding reduction due to the Y315A mutation was evident by a shift of A3G from high molecular mass RNP complexes to low molecular mass complexes. We conclude that interactions of A3G with bulk nucleic acids strongly depends on Y315 and hypothesize that while the N-terminal domain binds RNAs such as 7SL1 and HIV genomic RNA, competitive binding of RNA and ssDNA to Y315 may be an important drug development target for activating the innate antiviral host defense potential of A3G both therapeutically and during viral reservoir reactivation.

BiographyDr. Harold C. Smith is a Ph.D. cell and molecular biologist and full professor at the University of Rochester.

His research laboratory has published 140 pier reviewed manuscripts on the biological control mechanisms affecting cancer, elevated cholesterol and heart disease and HIV/AIDS. He is a key opinion leader on APOBEC proteins. In 2003, Dr. Smith founded and is CEO for OyaGen, Inc. that conducts antiviral drug discovery and drug development.

Normalizing urine exosome biomarkers: a second look at hidden gems

Peter Yuen*, Erik Koritzinsky, Jonathan Street and Robert StarNational Institutes of Health, MD, USA

Abstract Non-invasive biomarkers for kidney disease are needed for accurate and timely diagnosis, including risk

assessment, early diagnosis, prognosis of disease progression, recovery from kidney disease and/or response to therapy. In recent years, the field has focused on the most urgent need, early diagnosis, and a few promising early diagnostic markers are being developed for clinical use. Kidney biopsies can fill in some of the gaps, but are rarely used beyond an initial diagnosis of kidney disease. Urine is a proximal fluid, enriched with biomarkers that derive from kidney cells, which has the potential to provide kidney- and GU tract-selective biomarker candidates.

While urine contains many kidney cell constituents, debris from damaged and dead cells can obscure information about the health status from kidney epithelial cells. In contrast, urine exosomes are small (~30-200 nm), intact vesicles, where intracellular contents are preserved, including mRNA, miRNA, and proteins, any of which can be promising biomarkers for kidney disease. Exosomes can come from any cell lining the nephron and can be readily separated from cellular debris.

Urine volume can vary by over an order of magnitude, which can obscure the cutoff values for urine biomarkers. Biomarker excretion rate, the gold standard for urine biomarkers, can be obtained with a 24 hour urine collection. While accurate, this method is impractical for most patients and represents a significant barrier to widespread clinical adoption. Instead, if a biomarker value is properly normalized with an appropriate denominator value, a

Page 14: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 14

single “spot” urine collection can be used.Similarly, urine exosome excretion rates can vary by over an order of magnitude, which does not correspond to

urine volume. Therefore, we examined normalization strategies for exosomal biomarkers in rats, and after kidney ischemia-reperfusion injury, we were able to identify a novel biomarker that was not statistically significant unless normalization was used.

BiographyDr. Peter S.T. Yuen obtained his Ph.D. from University of Minnesota and received further postdoctoral

training at Vanderbilt University and University of Texas, Southwestern Medical Center as a HHMI associate. He is currently a Staff Scientist in the Renal Diagnostics and Therapeutics Unit in the Kidney Diseases Branch of NIDDK, NIH. He has published over 60 publications, primarily on kidney disease, sepsis, and biomarkers.

Non-inhibitory pharmacological chaperones of glucocerebrosidase for the treatment of gaucher’s and parkinson’s disorders

Juan Jose Marugan1*, Samarjit Patnaik1, Elma Aflaki2, Wendy Westbroek2, Marc Ferrer1, Noel Southall1 and Ellen Sidransky2

1Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, MD, USA2Medical Genetics Branch, Human Genome Research Institute, National Institutes of Health, MD, USA

AbstractProtein folding is a thermodynamically-driven process by which newly synthesized polypeptides acquire

their minimum energy conformation. In many cases, the process is kinetically slow, requiring the binding of specific protein chaperones that catalyze the folding process by recognizing specific sequences and conformers. In the ER, properly folded proteins are incorporated into vesicles toward the Golgi apparatus, where they are post-transcriptionally modified, sorted and eventually distributed to specific membranes. Mutant proteins with diminished recognition by protein chaperones tend to accumulate in the ER and are eventually tagged for proteolysis, reducing the levels of protein at the intended site of action. Gaucher’s disease, the most common of the lipidosis disorders, is a recessive disorder caused by mutations in the lysosomal enzyme glucocerebrosidase (GCase). In addition, several clinical association studies have shown a clear link between GCase mutants and the development of Parkinson’s disease. Our group has identified and studied the biological effects of two series of non-inhibitory small molecule chaperones able to promote folding of GCase and increase its levels in the lysosome. Treatment with our molecules reduces the accumulation of glucosylceramide in Gaucher’s macrophages and iPSC-derived neurons. Our molecules are also able to reduce accumulated alpha synuclein in phenotypic dopaminergic Parkinson’s neurons carrying wild type and mutant GCase. These results open the door to development of a new class of molecules for the treatment of Gaucher’s and Parkinson’s disorders.

BiographyDr. Juan Marugan holds a group leader position at the National Center for Advancing Translation Sciences.

For more than 20 years he has been personally involved in numerous aspects of the translational process, with emphasis on medicinal chemistry and preclinical research including assay development, library screening and hit selection, structure activity relationship optimization studies, in vivo pharmacological, tolerability and efficacy evaluation, preclinical development and IND filing. He has extensive experience as team leader of programs in preclinical lead optimization, producing numerous series out licensed to pharmaceutical companies, advanced toward late development and entering in clinical trials.

Page 15: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 15

Antiviral nucleoside phosphonate prodrugs: potency amplifiers

Charles E. McKennaUniversity of Southern California, CA, USA

AbstractAcyclic nucleoside phosphonates (ANPs), in particular (S)-HPMPC (cidofovir) and (S)-HPMPA are highly

potent broad spectrum antiviral agents. However, poor ADME properties limit the utilization of these therapeutics in the clinic. Specifically, the nucleoside phosphonates are poorly absorbed, owing to the presence of a phosphonic acid group that ionizes at physiological pH and, when dosed intravenously, display dose-limiting nephrotoxicity due to their accumulation in the kidney.1 To address this issue, an amino acid phosphonate ester prodrug approach is being developed, which explores the use of benign single amino acids to mask the negative charges of the ANPs, and the incorporation of a long alkyl chain in the promoiety to increase prodrug lipophilicity to improve cell membrane permeability (e.g. by passive diffusion, membrane flippase activity or phospholipid uptake), which might translate into enhanced potency by increasing intracellular drug concentration following activation by endogenous enzymes. The corresponding prodrugs exhibit excellent antiviral activities against HCMV, HSV, VZV, cowpox and vaccinia viruses. USC-087 and USC-505, two orally available tyrosinamide prodrugs of HPMPA and HPMPC, showed protection of immunosuppressed Syrian hamsters against lethal intravenous challenge with human type 5 and 6 adenoviruses.2 (Supported in part by NIH-NIAID).

(1) I.S. Krylov, B.A. Kashemirov, J.M. Hilfinger, C.E. McKenna CE. “Evolution of an amino acid based prodrug approach: stay tuned.” Mol. Pharm. 2013 10:445-458.

(2) K. Toth, J.F. Spencer, B.A. Kashemirov, C.E. McKenna, M. Prichard, W.S.M. Wold. “USC-087 and USC-505 Protect Immunosuppressed Syrian Hamsters against Lethal Intravenous Challenge with Human Type 5 and 6 Adenoviruses.” International Society for Antiviral Research 2016.

BiographyDr. Charles McKenna is Professor of Chemistry, Pharmacology & Pharmaceutical Sciences at the University

of Southern California (USC), where he is also Dornsife College Vice Dean for Natural Sciences. He received his Ph.D. in Chemistry at UC San Diego and was an NIH postdoctoral Fellow at Harvard University. His research interests are primarily in medicinal and organophosphorus chemistry, focusing on bisphosphonate imaging probes for bone, nucleotide probes of DNA polymerases, and design of novel chemotherapeutic agents. Author of over 225 publications and patents and the recipient of numerous awards for his research and teaching, Dr. McKenna has founded several biotech companies.

Technical Session – II

New structure based drug discovery opportunities for GPCRs and transporters

Sid Topiol3D-2drug LLC, NJ, USA

AbstractStructure based drug design is now well established as a highly efficient approach in pharmaceutical research.

The approach uses atomic level detail structures of target proteins, most generally determined through X-ray crystallography. For many years this approach has not been possible for the most common classes of drug targets, particularly membrane bound proteins such as GPCRs and Transporters. Approximately 10 years ago, the first X-ray structures in these areas started to emerge and the generation of more structures has begun to accelerate in the recent past. These structures cover a rapidly increasing number of targets and categories including class A, B, C and F GPCRs, endogenous amine transporters, proteins in various states, novel sites for ligand modulation and other characterizations. This information has provided valuable information facilitating the design of new compounds with specific features such as intrinsic activity (from active to inactive), ligand bias, selectivity, allosterism and others. This

Page 16: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 16

growing body of information will be illustrated along with examples of successes and emerging new opportunities.

BiographyDr. Sid Topiol received his B.S. from CCNY and his Ph.D. from NYU. He trained as a theoretical quantum

chemist. He post-docked at Northwestern with Mark Ratner and Arthur Frost, and at Carnegie-Mellon with John Pople (Noble laureate.) He first joined The Mount Sinai School of Medicine in the Department of Pharmacology. He has spent over 20 years in Pharma, starting with Berlex followed by Sandoz/Novartis and then Lundbeck. He has worked on all aspects of Computer Aided Drug Design in interdisciplinary project environments. He now combines industry and academic activities. In 2010 he formed 3D-2Drug, a consulting company which provides the means for the most efficient discovery activities through advanced scientific approaches. In 2011 he began teaching as an Adjunct Lecturer at New Jersey Institute of Technology and, in 2014, joined the newly forming Center for Healthcare Innovation at the Stevens Institute of Technology. He serves on the Editorial Board of Drug Development Research.

Hydrogen/deuterium ratio is a key regulator of energy production and cell proliferation – submolecular dimensions of drug development

Gábor Somlyai1*, Miklós Molnár2, Ildikó Somlyai1, István Fórizs3, György Czuppon3 and László G. Boros4-7

1HYD LLC for Cancer Research and Drug Development, Budapest, Hungary2Institute of Pathophysiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary3Research Centre for Astronomy and Earth Sciences, Hungarian Academy of Sciences, Budapest, Hungary4Department of Pediatrics, Harbor-UCLA Medical Center, UCLA School of Medicine, Los Angeles, CA, USA5The Los Angeles Biomedical Research Institute (LABiOMED), Torrance, CA, USA6SIDMAP, LLC, Culver city, CA, USA7UCLA Clinical and Translational Science Institute, Torrance, CA, USA

AbstractThe concentration of deuterium (D) is about 150 ppm (over 16 mM/L) in surface waters and 12-14 mM/L

in living organisms, which is clearly a biologically relevant ion considering that circulating Ca2+ is only 2.24-2.74 mM/L. In order to reveal the possible role of naturally occurring D in living organisms, the replacement of normal tap water with deuterium depleted water (DDW) in a range from 25 ppm to 135 ppm was investigated in cell cultures, animal studies, as well as in prospective blinded and retrospective clinical trials. DDW inhibited cell growth of multiple cancer cell lines in in vitro culturing studies and readily induced tumor xenograft regression in mice. Double blind, controlled, human Phase II clinical trial with prostate cancer, in compliance with GCP principles exhibited a significant difference between the control (n=22) and DDW treated (n=22) patients with respect to end point parameters and confirmed significant antitumor efficacies.

Thirty volunteers with decreased glucose tolerance underwent 90 days long DDW treatment. Fasting glucose decreased significantly after 90 days DDW treatment (6.06±0.66 mmol/L at day0 and 5.74±0.94 mmol/L at day 90, p=0.029). Evaluation of individual cases showed that glucose uptake increased in 11 volunteers (6.9±2.4 mg/kg/min at day 0, 8.6±2.5 mg/kg/min at day 90, p=0.0014).

Our data demonstrate that the depletion of D increases submolecular proton transfers with improved protein machines for complete substrate oxidation with limited anabolism and cell proliferation. Orally administered deuterium depleted water opens submolecular dimensions for drug development that can replace many underperforming drug development efforts of the genomics era.

BiographyDr. Gábor Somlyai graduated as a biologist at the University of Szeged in 1982. In 1988 he defended his Ph.D.

thesis in molecular biology. In the same year he worked at Georg August University in Göttingen for 6 months, from the end of 1988 held a postdoctoral fellowship at the University of Missouri (Columbia, Missouri, USA. Since 1990 he started to investigate the possible role of naturally occurring deuterium in living organism at the Hungarian Institute of Oncology. Gábor Somlyai is the author of numerous scientific publications in Hungarian and English and recognized speaker of international conferences.

Page 17: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 17

Bisphosphonates:  bones and beyond

Graham Russell

The Mellanby Centre, Sheffield University and the Botnar Research Centre, Oxford University, UK

Abstract The biological effects of the bisphosphonates (BPs), originally called diphosphonates, were first reported in

1969. (See Russell RG. Bisphosphonates: The first 40 years. Bone. 2011;49:2-19). Their earliest use was for bone scintigraphy, and the treatment of Paget’s disease. This was later extended to the prevention of skeletal-related events in patients with myeloma or bone metastases, and to osteoporosis. BPs are still the leading drugs used for these disorders.

The pharmacological effects of BPs as inhibitors of bone resorption appear to depend upon two key properties; their affinity for bone mineral, and their inhibitory effects on osteoclasts. After being internalised by osteoclasts the nitrogen-containing BPs (including alendronate, risedronate, ibandronate, minodronate and zoledronate) inhibit farnesyl pyrophosphate synthase (FPPS), a key enzyme in the mevalonate pathway, which generates isoprenoid lipids utilized for the post-translational modification of small GTP-binding proteins that are essential for osteoclast function.

BPs have now become largely generic drugs, as key patents have expired, but are likely to remain major drugs for treating bone diseases. There are obvious opportunities for extending the use of BPs to other areas of medicine. Several recent studies suggest that BPs may be associated with other clinical benefits outside the field of bone diseases, eg on mortality, cardiovascular disease, and reduction of colon cancer. The non-skeletal effects of BPs include inhibition of several protozoan parasites, increasing longevity in animal progeria models, and enhancing human stem cell life span, DNA repair and tissue regeneration. BPs can also be used for selectively delivering other drugs to bone.

BiographyDr. Graham Russell graduated in Biochemistry from Cambridge University and spent his formative years

in Leeds, Davos, Bern, Oxford and Boston. He moved to Sheffield in 1976, and subsequently back to Oxford University in 2000. His early work with Herbert Fleisch in Switzerland led to the discovery of the biological effects of bisphosphonates, and to their eventual successful clinical use in the treatment of bone resorption disorders, including bone metastases and osteoporosis. Later his group elucidated how bisphosphonates act within cells, as inhibitors of the mevalonate pathway of cholesterol biosynthesis, thereby enabling multiple new potential medical uses for these compounds.

New drug candidates for liposomal delivery identified by computer modeling of liposomes’ remote loading and leakage

Ahuva Cern1, 2, David Marcus3, Alexander Tropsha4, Yechezkel Barenholz1 and Amiram Goldblum2*

1Laboratory of Membrane and Liposome Research, Department of Biochemistry, IMRIC, The Hebrew University - Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem, Israel2Molecular Modeling and Drug Design Laboratory, Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel3European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL–EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, United Kingdom; 4The Laboratory for Molecular Modeling, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC, USA

AbstractRemote drug loading into nano-liposomes is in most cases the best method for achieving high concentrations

of active pharmaceutical ingredients (API) per nano-liposome that enable therapeutically viable API-loaded nano-liposomes, referred to as nano-drugs. This approach also enables controlled drug release. Recently, we constructed

Page 18: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 18

computational models to identify APIs that can achieve the desired high concentrations in nano-liposomes by remote loading. Towards that goal, we used our in–house “Iterative Stochastic Elimination” (ISE) algorithm. ISE is a heuristic which finds solutions to extremely complex combinatorial problems. We model and predict API suitability for nano-liposomal delivery by fixing the main experimental conditions: liposome lipid composition and size to be similar to those of Doxil® liposomes. On that basis, we add a prediction of drug leakage from the nano-liposomes during storage. The “load and leak” models were used to screen two large molecular databases in search of candidate APIs for delivery by nano-liposomes. The distribution of positive instances in both loading and leakage models was similar in the two databases screened. The screening process identified 667 molecules that were positives by both loading and leakage models (i.e., both high-loading and stable). Among them, 318 molecules received a high score in both properties and of these, 67 are FDA-approved drugs. This group of molecules, having diverse pharmacological activities, may be the basis for future liposomal drug development.

BiographyDr. Amiram Goldblum studied Chemistry and Physics followed by a Ph.D. in Organic Chemistry at the

Hebrew University of Jerusalem (With Prof. R. Mechoulam). He was subsequently a postdoctoral fellow at the Institute for Physico-Chemical Biology in Paris, studying Quantum Biochemistry (with Profs. Alberte and Bernard Pullman), at Pomona College, Claremont, CAL studying Quantitative Structure-Activity Relations (with Prof. Corwin Hansch) and at Stanford University, Palo Alto, CAL, studying computational reaction mechanisms of enzymes (with Dr. Gilda Loew). Goldblum is currently head of the Molecular Modeling and Drug Design unit at the Institute for Drug Research of the Hebrew University of Jerusalem.

Privileged structures: a versatile concept to explore new chemical space with medicinal chemistry utility

Gerhard Müller1, Brian Kuijpers1, Jorg Benningshof1*, Koen Hekking1, Dagmar Stumpfe2, Antonio de la Vega de León2, Norbert Furtmann2, Dilyana Dimova2 and Jürgen Bajorath2

1Mercachem bv, Kerkenbos 1013, 6546 BB Nijmegen, Netherlands 2Department of Life Science Informatics, B-IT, LIMES Program Unit Chemical Biology and Medicinal Chemistry, Rheinische Freidrich-Wilhelms-Universität, Dahlmannstrasse, Bonn, Germany

AbstractMatching synthetically accessible chemical space with biological target space is one of the core activities of

medicinal chemistry. The content of today’s compound collections is a reflection of target families that have been addressed in the past, while chemical libraries are a reflection of the number and type of chemical reactions pursued in a 2 week chemistry/biology cycle time typical for lead finding campaigns. Hence, there is increasing evidence that currently pursued compound space does not match with wide areas of biological target space pharmaceutical industry will focus on in the future. Within our design and synthesis approaches we embark into a systematic exploration of spiro-cyclic systems, as well as macrocycles encompassing ring sizes from 8 to 17 ring atoms. Results of a systematic cheminformatics analysis of the bioactive compound space will be presented, followed by structure-based designs and chemical synthesis of novel spiro-cyclic and macrocyclic ring topologies. The focus on spiro-cyclic systems is based on a strong correlation with the GPCR target family, while we will show that macrocyclic systems are capable of disrupting complex protein-protein interactions, exemplified by IL-17A and Mcl-1 antagonists. In pursuit of this concept, we achieve an optimal balance between novelty and proximity to bioactive compound space, i.e. resemblance of e.g. peptide secondary structure elements, and increased 3D skeletal complexity. We consider this as a significant contribution to unlock the chemical accessible spiro-cyclic and macrocyclic ring system space that is often inaccessible in lead finding and lead optimization campaigns due to the underlying chemical complexity.

Page 19: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 19

Characterizing the human norovirus nucleotydylation: a potential target for intervention?

Brent E. Korba

Department of Microbiology and Immunology, Georgetown University Medical Center, Washington, DC, USA

AbstractThe VPg protein of human Norovirus (hNoV), which is covalently attached to the 5’-end of the viral genome,

is a multi-functional protein required for virus replication. VPg is used to prime viral replication and recruits cellular transcription initiation proteins. Nucleotidylylation of hNoV VPg by its polymerase at Tyr27 is obligatory to attach VPg to viral RNA. In in vitro reactions, we have demonstrated that the un-cleaved hNoV precursor protein, ProPol (NS5-6) was 100-fold more efficient in catalyzing VPg nucleotidylylation than the mature viral polymerase (Pol, NS6), suggesting a specific role for this long-lived, intracellular viral precursor protein. We identified several positively charged amino acids in the first 20 amino acids of the amino terminal end of hNoV VPg that regulate its nucleotidylylation, likely though several potential mechanisms. We provided the first evidence that VPg directly binds NTPs and that inhibition of binding inhibits nucleotidylylation. Alanine substitution of multiple positively-charged amino acids within the first 13 amino acids of VPg is required to inhibit nucleotidylylation or NTP binding. By contrast, a single substitution of Lys20 abolished nucleotidylylation (but not NTP binding). Since hNoV VPg nucleotidylylation is performed by a viral polymerase, we show that a previously identified non-nucleoside inhibitor of the hNoV polymerase can inhibit nucleotidylylation, indicating that VPg nucleotidylylation is a potential antiviral target and that polymerase inhibitors can potentially affect two essential hNoV processes.

BiographyProf. Brent Korba has been a faculty member of the Department of Microbiology and Immunology in

the Georgetown University Medical Center since 1984, with expertise in early-stage preclinical drug discovery (enzymatic and tissue culture-based assays, pre-clinical animal models). He has conducted antiviral testing contracts for the NIAID/NIH since 1990, focusing on HBV, HCV, and Noroviruses. Dr. Korba has published 118 peer-reviewed papers, 18 book chapters and 64 conference abstracts, holds several patents, is an active consultant and past member of several scientific advisory boards in the pharmaceutical field, has been on editorial boards of several journals, and a member of several grant study sections for the NIH and ACS.

Novel treatment strategies for smooth muscle disorders: targeting Kv7 potassium channels

Kenneth L. Byron*, Lyubov I. Brueggemann, Leanne L. Cribbs and Matthias MajetschakLoyola University Chicago, Stritch School of Medicine, Maywood, IL, USA

Abstract Smooth muscle cells provide crucial contractile functions in vascular and lung tissues, controlling vascular

and airway resistance, respectively. The contractile state of smooth muscle cells is largely determined by their electrical excitability, which is in turn influenced by the activity of potassium channels. The activity of potassium channels sustains smooth muscle cell membrane hyperpolarization, reducing cellular excitability and thereby promoting smooth muscle relaxation. Our research has indicated an important role for Kv7 (KCNQ) voltage-gated potassium channels in the regulation of the excitability of vascular and airway smooth muscle cells. We have detected expression of multiple Kv7 channel subtypes and demonstrated that a number of clinically used drugs, some of which were developed to target Kv7 channels in other tissues, exert robust effects on the smooth muscle Kv7 channels. Functional studies in tissues and in vivo have indicated that Kv7 channel activators and inhibitors have the ability to relax and contract smooth muscle, respectively, suggesting a wide range of novel applications for the pharmacological tool set. Taking advantage of these functional effects, we have recently provided evidence supporting novel therapeutic uses for Kv7 channel activators in the treatment of cerebral vasospasm and asthma. In addition, our recent findings support a therapeutic benefit of the use of Kv7 channel blockers in the treatment of hemorrhagic shock.

Page 20: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 20

BiographyDr. Kenneth L. Byron earned a BA in Natural Sciences from Johns Hopkins University and a Ph.D. in Cell

Physiology from the University of Chicago. After 3 years as a Wellcome Trust Fellow in the Department of Pharmacology at Cambridge University in the U.K., he joined the faculty at Loyola University Chicago where he has advanced in rank to his current position as Professor of Molecular Pharmacology & Therapeutics. For the past 24 years, Dr. Byron has directed an internationally renowned extramurally funded research program at Loyola, focusing on calcium signaling and regulation of ion channels in vascular and airway smooth muscle.

Antiplasmodial activity and cytotoxicity of lapachol and synthetic related naphthoquinones

Tatiane Freitas Borgati, Maria Fernanda Alves do Nascimento and Alaíde Braga de Oliveira*Faculdade de Farmácia, Departamento de Produtos Farmacêuticos, Universidade Federal de Minas Gerais - UFMG, Brazil

Abstract Malaria is caused by protozoa of the Plasmodium genus, being endemic in 91 tropical and sub-tropical

countries. In Brazil, malaria is endemic in the Amazonia where the main agents are P. vivax and P. falciparum, the first one being the prevalent. Resistance of the parasite to the available antimalarial drugs is a serious problem (WHO, 2016) and the quest for new antimalarials is of great relevance. In Brazil, plant species belonging to the genera Handroanthus and Tabebuia (Bignoniaceae family) are sources of naphthoquinones such as lapachol that disclose an wide spectrum of bioactivities what has motivated the synthesis of a series of eight analogues that were assayed in vitro against chloroquine resistant P. falciparum (W2 strain) and for cytotoxicity in HepG2 cell cultures. The synthesized naphthoquinones were more potent than lapachol (IC50 206.38 µmol.L-1). Lapachol, that was clinically used to treat malaria during the second world war due to the scarcity of quinine, disclosed low activity (IC50 206.38±3.64 µmol.L-1). Furthermore, lapachol was a lead molecule for the development of atovaquone, a naphthoquinone that was recently introduced in malaria chemotherapeutics, although resistance was already observed. Our results encourage to pursue the optimization of naphtoquinone structures in the quest of potential antimalarial. Financial support: CNPq, FAPEMIG

BiographyDr. Alaide Braga de Oliveira is an Emeritus Professor and a Senior Researcher at Universidade Federal de

Minas Gerais – UFMG – in Belo Horizonte, state of Minas Gerais, Brazil. She is a member of the Academia Brasileira de Ciências and a Research Fellow of the CNPq (National Research Council). Since the beginning of her academic carrier, her major research interests were in the Phytochemistry of plants from the Brazilian Flora and in the two last decades she has concentrated her efforts on the investigation of the biological effects of medicinal plants aiming to identify bioactive compounds as well as the synthesis of biologically active natural products and analogues. The presently reported results are part of T. F. Borgati Ph.D. Thesis with the collaboration of M. F. A. Nascimento for the biossays.

Page 21: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 21

Systemic, targeted nanotherapies for the treatment of neuroinflammation in CNS disorders: from chemistry to clinical translation

Kannan RangaramanujamArnall Patz Distinguished Professor of Ophthalmology, Co-director, Center for Nanomedicine, Johns Hopkins School of Medicine, Baltimore, MD, USA

AbstractNeuroinflammation, mediated by activated microglia and astrocytes, plays a key role in the pathogenesis of

cerebral palsy (CP), autism, and most other debilitating central nervous system (CNS) disorders. ‘Appropriate’, targeted manipulation of neuroinflammation can bring novel approaches for treated diseases, increasing the drug efficacy and decreasing the side effects (a major issue with many CNS therapeutics). However, targeted delivery of drugs to specific cells in the CNS is a challenge. To address this, we take advantage of the unique, intrinsic, pathology-dependent, brain uptake of dendrimers (with no targeting moieties) in more than 20 disease models of CNS and retinal disorders. Upon systemic administration, hydroxyl-terminal poly (amidoamine) dendrimers localize selectively in activated microglia and astrocytes in animals with CP. Building on these findings, we have designed and synthesized dendrimer-drug conjugates which have shown significant promise for many brain and ocular disorders. Two examples of this approach of targeted, systemic therapy for neuroinflammation, one for pediatric brain injury and one for age-related macular degeneration, representing two opposite ends of the age spectrum, will be presented. Delivering drugs to injured glia cells also allows us to understand their role in repair and recovery following brain injury, with broad implications in developing novel therapies for CNS disorders, a growing health problem worldwide, and yet an unmet need. These dendrimer-drug conjugates are undergoing commercialization and clinical translation.

A rapid-evolving small RNA based immune system targeting avian leukosis virus in chickens

Xin Zhiguo Li * and Yu Huining SunCenter for  RNA  Biology: From Genome to  Therapeutics, Department of Biochemistry and Biophysics, Department of Urology, University of Rochester Medical Center, Rochester, NY, USA

Abstract Vertebrate genomes harbor a large number of pre-existing retroviral invaders, and face a never-ending stream

of new retroviral endogenization. The challenge of controlling endogenous retroviruses (ERVs) is formidable, as ERVs can remain infectious and can recombine with other viruses or host genes to evolve into new viruses. The conflict between ERVs and their hosts is especially brutal in germ cells where the propagation of ERVs will be inherited, requiring the host to distinguish non-self from self-genetic elements. The defense mechanisms essential for animal fertility from worms to humans, are PIWI-interacting RNAs (piRNAs), which protect the integrity of the germ-line genome by targeting ERVs through base-pair complementarity. However, we are just beginning to understand how the host keeps up the arms race with ever-changing ERVs. Here, we described new piRNA acquisition in vertebrates, in which chickens hijack a pre-existing provirus for piRNA production to defend against Avian leukosis virus (ALV), employing a strategy similar to the prokaryotic CRISPR-Cas system. The provirus produces anti-viral piRNAs in a domestic egg-laying breed, but does not do so in the Red Jungle Fowl, the undomesticated wild chickens. Our study identified previously unrecognized mechanisms enabling the host to rapidly evolve its piRNA repertoire and target ERVs specifically.

BiographyDr. Xin Zhiguo Li has completed his Ph.D. with Bik Tye and John Schimenti from Cornell University

and postdoctoral studies with Philip Zamore from HHMI/University of Massachusetts Medical School. He is an assistant professor at University of Rochester Medical center. Research in Li lab is at the interface between reproductive Biology and RNA biology.  Li laboratory is currently focusing on piRNAs to define how RNAs protect genome integrity and shape information flow across generations.

Page 22: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

2November 10

Friday

Page 23: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Technical Session III

Hydralazine induces stress resistance and extends lifespan in C. elegans via Nrf2/SKN-1 pathway

Hamid Mirzaei1*, Esmaeil Dehghan1, Yiqiang Zhang2, Bahar Saremi1, Sivaramakrishna Yadavali1, Amirmansoor Hakimi1, Maryam Dehghani1, Mohammad Goodarzi1, Xiaoqin Tu1, Scott Robertson3, Rueyling Lin3 and Asish Chudhuri1

1Department of Biochemistry, UT Southwestern Medical Center, Dallas, TX, USA2Greehey Children’s Cancer Research Institute, UT Science Center at San Antonio, San Antonio, TX, USA3Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX, USA

Abstract

Oxidative stress increases gradually with aging and steadily diminish the cell’s ability to maintain homeostasis. Nuclear factor (erythroid-derived 2)-like 2 and its C. elegans ortholog, SKN-1, are transcription factors that play a pivotal role in the oxidative stress response, cellular homeostasis, and lifespan. But like other defense systems, the Nrf2-mediated stress response is compromised in aging and neurodegenerative diseases. In this study we provide evidence that hydralazine, a drug used for treatment of hypertension, is a bona fide activator of the Nrf2/SKN-1 pathway. We demonstrate that hydralazine protects Alzheimer’s disease model cells and C. elegans from chemical stressors linked to neurodegenerative diseases. We also show that hydralazine extends lifespan and health in C. elegans. Hydralazine is an FDA approved drug; therefore, we suggest it is an excellent candidate for clinical trials for treatment of age-related disorders. Hydralazine may also offer general health benefits for the aging population.

Biography

Dr. Hamid Mirzaei’s research is focused on finding the target of novel and FDA approved compounds using a combination of proteomics, computational biology and biochemistry. Many FDA approved drugs are currently in use without clear understanding of their mechanism of action. On the other hand there are quite a few well-characterized natural products with unknown targets. Dr. Mirzaei uses systems biology to understand the drug’s mechanism of action by identifying the target of the drugs and their cellular and organismal phenotypes.

Can lipid nanoparticles improve intestinal absorption?

Maria Mendes1,2, Helder Soares3, Luís Arnaut3, João Sousa1,2, Alberto Pais3 and Carla Vitorino1,2*

1Pharmacometrics Group of the Centre for Neurosciences and Cell Biology (CNC), University of Coimbra, Portugal2Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Portugal3Department of Chemistry, University of Coimbra, Portugal

Abstract

Lipid nanoparticles and their multiple designs have been considered appealing nanocarrier systems. Bringing the benefits of these nanosystems together with conventional coating technology clearly results in product differentiation.

This work aimed at developing an innovative solid dosage form for oral administration based on tableting nanostructured lipid carriers (NLC), coated with conventional polymer agents. A dual-step development approach was performed, firstly, focused on the optimization of the NLC scale-up, and subsequently, on the conversion of NLC into tablets. NLC dispersions co-encapsulating olanzapine and simvastatin (Combo-NLC) were produced by high pressure homogenization, and evaluated in terms of scalability, drying procedure, tableting and performance from in vitro release, cytotoxicity and intestinal permeability stand points.

Factorial design indicated that the scaling-up of the NLC production is clearly feasible. The chosen formulation

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 23

Page 24: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

presented a particle size of 156±4 nm, with good stability and drug loading properties. Spray-drying was the method selected to obtain dry particles, not only because it consists of a single step procedure, but also because it facilitates the coating process of NLC with different polymers. Modified NLC formulations with the polymers allowed obtaining distinct release mechanisms, comprising immediate, delayed and prolonged release. Sureteric: Combo-NLC provided a low cytotoxicity profile, along with a ca. 12-fold OL/3-fold SV higher intestinal permeability, compared to those obtained with commercial tablets.

Such findings can be ascribed to drug protection and control over release promoted by NLC, supporting them as a versatile platform able to be modified according to the intended needs.

Acknowledgements

The authors acknowledge Fundação para a Ciência e a Tecnologia (FCT), Portuguese Agency for Scientific Research, for financial support through the Research Project POCI-01-0145-FEDER-016648, the project PEst-UID/NEU/04539/2013, and COMPETE (Ref. POCI-01-0145-FEDER-007440). The Coimbra Chemistry Centre is supported by FCT, through the Project PEst-OE/QUI/UI0313/2014 and POCI-01-0145-FEDER-007630. Maria Mendes acknowledges the Ph.D. research Grant SFRH/BD/133996/2017 assigned by FCT. Carla Vitorino also acknowledges Fundação Luso-Americana para o Desenvolvimento for the financial support at the CSDD2017 attendance.

Biography

Dr. Carla Vitorino received her Ph.D. degree in Pharmacy, Pharmaceutical Technology, in 2013, from the University of Coimbra, Portugal. Currently, she is an invited Assistant Professor at the Faculty of Pharmacy, University of Coimbra and a member of the Pharmacometrics Group of the Centre for Neurosciences and Cell Biology of the same University. She has developed work concerning permeation enhancement strategies for transdermal and oral drug delivery, published in high-impact peer reviewed journals. She is now involved in a project, aimed at developing nanomedicines as therapeutic surrogates to treat glioblastoma. Her research interests are Nanotechnology, Controlled-Release, and Novel Drug Delivery Systems.

A trisaccharide analog that acts as a TLR-4 agonist: a potential vaccine adjuvant?

Cecilia H. Marzabadi1*, Vikram Basava1 and Constantine Bitsaktsis2

1Department of Chemistry & Biochemistry, Seton Hall University, NJ, USA2Department of Biological Sciences, Seton Hall University, NJ, USA

Abstract

A novel small molecule carbohydrate has been synthesized and its immunomodulatory effects in cells has been studied. Pro-inflammatory cytokine secretion (TNF-α and IL-6) was observed in Raw 264.7 macrophages and in PECs from C57BL6 mice (4-6 weeks). The mechanism of innate signaling was studied in TLR2 and TLR4 ko C57 BL6 PECs and was shown to be dependent on TLR4 and not TLR2. Using flow cytometry, competitive binding to TLR4 in the presence of added E. coli LPS showed that the new analog binds to TLR4 at the same site as LPS and binding is dose-dependent. Downstream signaling from TLR4 was further studied using MyD88 knockout mice.-Structural modification of the lead compound-is ongoing to assess the efficacy of structural changes on the biological activity and signaling pathways.

Biography

Prof. Cecilia H. Marzabadi is a synthetic organic chemist and currently serves as the chairperson for the Department of Chemistry & Biochemistry at Seton Hall University. Her research areas are in the synthesis of carbohydrate-based small molecule therapeutics, in particular, for cancer, CNS and immunological applications. In addition, she is interested in increasing and diversifying the STEM workforce and has done research looking at the reasons for the low numbers of women at higher levels and more prestigious institution.

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 24

Page 25: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 25

Tissue models for drug screening against cancer stem cells

Esmaiel Jabbari

University of South Carolina, SC, USA

Abstract

Increasing evidence suggests that the tumor heterogeneity is rooted in the existence of a very small sub-population of stem-like cells (CSCs) within the population of tumor cells. Since cancer takes many years to grow, it is important to develop in vitro models to study tumor initiation and progression. In an effort to understand the role of CSCs in cancer relapse and drug resistance, my laboratory has merged material synthesis, microscale technologies, and cancer biology to develop an engineered 3D tissue model for selection and enrichment of CSCs as a platform technology to investigate the role of individual factors in the tumor microenvironment on CSC maintenance, self-renewal, and differentiation. I will demonstrate in my seminar that this platform 3D tissue culture technology is potentially useful in drug screening and personalized cancer treatment.

Biography

Dr. Esmaiel Jabbari is Tenured Full Professor of Chemical Engineering at the University of South Carolina. He directs the Tissue Engineering and Drug Delivery Laboratory which specializes on the design of 3D Tissue models to study the effect of micro-environmental factors on the fate of stem cells. He has received numerous awards for his research program including Berton Rahn Award in 2012, Stephen Milam Award in 2008, and election to the College of Fellows of AIMBE in 2013. He is the author of >250 refereed articles and he has mentored >130 scholars. He serves as Academic Editor for PLOS ONE.

The essential role of pharmacology in discovery translational research

Giora Z. Feuerstein

Temple University, Philadelphia, Pennsylvania, USACellphire, Inc, Rockville, MD, USA

Abstract

The process whereby medicines are discovered and developed has been dramatically transformed over the past decades due to wealth of basic scientific knowledge, technology advances and systematic transformation of regulatory and clinical trials practices. However, at the start of the 21st century delivery of innovative drugs for diseases that afflict billions of people across countries remain a daunting task marred by high attrition rate (>95%). This reality steadily erodes the traditional pharmaceutical industry business model and stalls needed innovative medicine for leading causes of morbidity and mortality: cardiovascular- metabolic, respiratory, neurodegenerative and cancer. Much has been written and debated on the obstacles the pharmaceutical industry face in its efforts to “outpace” the growing disease burden to the detriment of the national health care services and the patients. In analysis of the complex reasons that lead to this dire situation. Intrinsic factors in pharmaceutical discovery phases point to correctable measures – pharmacology, which is virtually mal-practiced across the pre-critical stages of drug validation likely for financial burden that detailed pharmacological studies require and prolongation of the development timeline that such practices command. This presentation aims to illuminate on principles of target validation, compound-target interaction and translational pharmacology principles in transitioning candidate compounds into clinical practices. The teaching of this presentation should help scientists interested in engaging in drug discovery-practices to secure a higher rate of success.

Biography

Dr. Giora Z. Feuerstein is a graduate of the Hadassah Medical School in Jerusalem Israel where is also received his MSc degree in Pharmacology. Following a Fogarty fellowship at the NIH, Bethesda Maryland where he trained in Clinical Sciences research (focused on the central and peripheral adrenergic system) Dr. Feuerstein

Page 26: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

joined the USUHS to become the Professor (Research) and head of the Neurobiology Research labs where is studies the role of adrenergic and peptidergic neurotransmitters in adaptation to cardiovascular stress (shock) and CNS ischemia/trauma. Dr. Feuerstein then joined the Pharmaceutical Industry where he stayed for 25 years) holding executive positions (GSK, Merck, Wyeth/Pfizer) in cardiovascular and hemostasis drug discovery and development. At GSK he lead the discovery and pre-clinical development of carvedilol (COREG) - the first third “generation beta-blocker” registered for treatment of heart failure. Over his time in the Industry Dr. Feuerstein contributed to numerous programs that transitioned to clinical development and has received several distinguished awards. At Wyeth/Pfizer, Dr. Feuerstein served as the Head of discovery ‘Translational Medicine’ and Biomarkers development with global responsibilities. After retirement from the Pharmaceutical Industry, Dr. Feuerstein served as Chief, Medical and Technical officer for the “Medical Counter Measures” program for the USG/DoD for 6 years (2011-2016). At the present time, Dr. Feuerstein serves as Chief Medical and Technical Officer at Debina Diagnostics Inc., Pennsylvania focusing on Bio-Engineering of NanoDiamond Particles for diverse medical utilities. Dr. Feuerstein lists over 700 scientific publications in professional and 15 patents, has served on numerous professional editorial boards and grant study sections.

Delineating ligand binding to the human cytochrome P450 3A4

Maria Kontoyianni*, Christina Hayes, Brett Lacy, Brittany Schuchman and Phillip Kilver

Department of Pharmaceutical Sciences, Southern Illinois University, Edwardsville, IL, USA

Abstract

Drug-drug interactions are critical in drug development because they potentially lead to reduced bioavailability or toxicity of a clinical agent. The major site of drug-drug interactions is the P450 system, with the 3A4 isoform (CYP3A4) being responsible for the biotransformation of more than one-third of currently marketed drugs. Thus, co-administration of a therapeutic that is normally metabolized (substrate) by CYP3A4 with an inhibitor can prolong its plasma levels, whereas CYP3A4 inhibition can be beneficial when metabolism results in toxic or carcinogenic metabolites. CYP3A4 is malleable, and can accommodate up to three ligands in its active site. Furthermore, some drugs are CYP3A4 substrates, others behave as inhibitors, while quite a few can be substrates and inhibitors. We carried out ensemble-docking experiments of substrate and inhibitor libraries with a modified Induced Fit Docking methodology and a number of scoring functions. Enzyme conformations included three currently available crystal CYP3A4 structures. All docking experiments were performed in duplicates with and without inclusion of crystallographic waters. Generated poses were assessed based on accuracy of site of metabolism prediction (substrates), and/or feasibility of the drug-CYP3A4 generated complexes (inhibitors). Our analyses suggest that certain residues make favorable interactions with bound ligands, while the presence of waters does not lead to improved docking performance which is dependent upon the macromolecular conformation and the size of the ligand. We will present our docking results along with findings from molecular dynamics simulations on ligand-free and ligated CYP3A34 structures.

Biography

Dr. Maria Kontoyianni holds a Ph.D. in computational chemistry from the University of North Carolina, Chapel Hill, and is an associate professor of pharmaceutical sciences at Southern Illinois University Edwarsdville. Before joining the SIUE School of Pharmacy, she worked in Research & Development of Fortune 500 companies such as Johnson & Johnson and Procter & Gamble. Dr. Kontoyianni’s interests involve the development of computational methodologies to understand ligand recognition, and implementation of processes to accelerate drug discovery. She sits on the editorial board of several journals, holds seven patents, and is panelist for the European Union large scale grant application proposals.

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 26

Page 27: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 27

Neutrophil gelatinase-associated lipocal in urine as a non-clinical safety biomarker for acute kidney injury

Kiyonori Kai*, Takashi Yamaguchi, Kumi Honda and Kazuhiko Mori

Medical Safety Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo

Abstract

Neutrophil gelatinase-associated lipocal (NGAL), a member of the lipocalin superfamily, is synthesized in renal tubular, intestinal, hepatic, and pulmonary tissue. In both mouse and rat models of acute kidney injury (AKI), NGAL has been shown to be detectable within an hour after onset of injury. Experimental studies demonstrated that NGAL gene expression was markedly and immediately up-regulated after renal ischemia/reperfusion injury in mice. Urinary NGAL is actively investigated for the prediction of AKI in a number of clinical settings including interventional trials for AKI, the diagnosis and management of cardiorenal syndrome. In the initial biomarker Predictive Safety Testing Consortium qualification submission, a set of 7 urinary biomarkers was qualified for preclinical use in rats to monitor AKI. The NAGL was additionally proposed as a next generation urinary biomarker and it was approved by FDA and EMA in 2014. For non-clinical toxicity study, interspecies evaluation of renal toxicity is necessary for adequate safety assessment of drugs. Gentamicin, an aminoglycoside antibiotic, was subcutaneously injected to rats, dogs or monkeys for approximately 2 or 3 weeks, and blood biochemistry, urinary biomarkers measurement including NGAL, and histopathological examination were performed. Degeneration and/or necrosis of proximal tubular epithelium were histopathologically observed in all species, and urinary NGAL markedly increased relative to serum creatinine or urinary nitrogen. Urinary NGAL is considered to be a sensitive biomarker in gentamicin-induced AKI models in rats, dogs or monkeys.

Biography

Dr. Kiyonori Kai is the senior director of clinical and anatomical pathology group for Medical Safety Research Laboratories, Tokyo, Japan. Dr. Kai is also Veterinarian, and advisory committee member of Japanese Society of Veterinary Pathology ( JCVP) and Japanese Society of Toxicologic Pathology ( JSTP). He is also Diplomate of JCVP, JSTP and Japanese Society of Toxicology. Dr. Kai is also a dog working group member of International Harmonization of Nomenclature and Diagnostic Criteria (INHAND).

High levels of serum lipid peroxides and protein carbonyls in mild dengue fever verses severe dengue

Sunil Premawansa1*, Maheshi Mapalagamage2, Gayani Premawansa3, Sharmila Thillainathan3, K. Karunayokiny3, Tharanga Fernando3, Dharshan De Silva4 and Shiroma Handunnetti2

1Department of Zoology and Environmental Sciences, University of Colombo, Sri Lanka2Institute of Biochemistry, Molecular Biology & Biotechnology, University of Colombo, Sri Lanka3North Colombo Teaching Hospital, Ragama, Sri Lanka4Genetech Research Institute, Colombo, Sri Lanka

Abstract

Oxidative stress arises due to the imbalance between pro-oxidants and anti-oxidants and it is a major cause of pathogenesis in dengue severity. Structure and function of lipids and proteins are damaged after reacting with pro-oxidants forming lipid peroxides (LP) and protein carbonyls (PC). Severe dengue hemorrhagic fever (DHF) and dengue fever (DF) patients admitted to Colombo North Teaching Hospital, Sri Lanka were recruited for the assessment of LP, antioxidant capacity (AOC) and PC using FOX-2 assay, ABTS decolorization assay and a commercially available kit respectively in patients at the time of admission (DHFA & DFA), critical stage (DHFC), discharge (DHFD & DFD), convalescence after 30 days (DHF-con and DF-con) and in healthy controls (HC).

DFA patients recorded significantly higher levels of LP (p=0.027) and PC (p<0.001) compared to DHFA. AOC in HC was significantly higher compared to different stages of dengue patients (p<0.001). These results

Page 28: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

show more damage of lipids and proteins in DFA patients and this effect may be due to more production of pro-oxidants such as reactive nitrogen and oxygen species in DFA patients. Accordingly it is evident that high levels of pro-oxidants are produced at early stage of the mild dengue (DF) infection, which may be involved in reducing the viral load effectively whereas at severe dengue, less production of the pro-oxidants at early disease stage may lead to high viral load resulting severity. Reduction of AOC in dengue patients compared to healthy controls also indicates the utilization of antioxidants to neutralize pro-oxidants produced in dengue patients.

Biography

Dr. Sunil Premawansa obtained his Ph.D. in Immunology and Molecular Biology of malaria from Faculty of Medicine, University of Colombo. He worked on transmission blocking vaccines of Plasmodium vivax malaria and immunopathogenesis and immunogenetics of P. vivax and P. falciparum malaria. For this work he worked as a post-doctoral fellow at University of Edinburgh, University of Oxford, UK and Pasteur Institute, France. He currently works on immunopathogenesis of dengue severity and leptospirosis, specifically studying the mechanisms for plasma leakage in dengue. He works as a Senior Professor at Faculty of Science, University of Colombo and teaches Immunology and Molecular Biology.

Advanced machine learning to boost the accuracy of scoring function

Simon Wang

Molecular Modeling and Drug Discovery Core Laboratory for District of Columbia Center for AIDS Research (DC CFAR), Department of Pharmaceutical Sciences, Howard University College of Pharmacy, Washington, DC, USA

Abstract

Structure-based virtual screening (SBVS) has become an indispensable technique for hit identification at the early stage of drug discovery. However, the accuracy of current scoring functions is not high enough to confer success to every targets thus remains to be improved. Precedently, we developed binary pose filter (PF) using knowledge derived from the protein-ligand interface of single X-ray structure of specific target. This novel approach had been validated as an effective way to improve ligand enrichment. Continuing from it, we attempt to incorporate protein flexibility from multiple crystal structures of the same target to build PF ensemble (PFE). To this end, we constructed a comprehensive data set to meet the requirement of ensemble modeling and validation, which contains 10 diverse targets, 118 well-prepared X-ray structures of protein-ligand complexes and large benchmarking actives/decoys sets. Notably, we designed our unique workflow of two-layer classifiers based on the concept of ensemble modeling and applied it to building PFEs for all targets. Via extensive benchmarking studies, we demonstrated that (1) PFE coupled with Chemgauss4 improves significantly the early enrichment of Chemgauss4 alone; (2) It shows greater consistency in boosting early enrichment and displays larger overall enrichment than the prior PF. In addition, we analyzed pairwise topological similarity among cognate ligands used to construct PFE and found that it is the higher chemical diversity enriched led to its improved performance. Taken together, the current outcome indicates that the incorporation of protein flexibility via ensemble modeling is able to enhance the screening competence of SBVS scoring functions.

Biography

Dr. Simon Wang is currently a tenured associate professor at the Department of Pharmaceutical Sciences, Howard University College of Pharmacy (HU COP). He also directs the Molecular Modeling and Drug Discovery Core Laboratory for District of Columbia Developmental Center for AIDS Research (DC D-CFAR). Dr. Wang has about 20 years of experience in structure-based drug design (SBDD), cheminformatics, fragment-based drug discovery (FBDD), high-throughput screening (HTS), molecular recognition, biomolecular simulation, and modern medicinal chemistry. He has published over 40 peer-reviewed research articles, reviews, book chapters and patents, as well as about 150 conference abstracts.

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 28

Page 29: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 29

Anti-cancer effects of marijuana and hemp

Wasana K. Sumanasekera

Sullivan University College of Pharmacy, Louisville, KY, USA

Abstract

Marijuana (Cannabis sativa) is a derivative from the hemp plant and is an addictive compound. Cannabis composed of several constituents including Δ9–tetrahydrocannabinol (Δ9-THC), cannabidiol (CBD), and cannabinol (CBN). While Δ9-THC is responsible for the mind altering properties, the non-psychoactive component, CBD is high in therapeutic value. Despite its’ adverse effects, the therapeutic value of cannabis can’t be ignored. Some US states legally allow cannabis for the personnel medical use. Medical cannabis can be used as a therapeutic option for many diseases including neuropathic pain, epilepsy, and cancer. However, cannabis use is controversial due to the side effects.

Hemp has the same genus and species name as marijuana but it contained fewer amounts of mind altering constituents, and increased amount of beneficial components such as CBD. Our laboratory is interested in studying the anti-cancer activity of CBD as well as hemp extract. Anti-cancer activity of cannabidiol (CBD) against ovarian cancer was detected using caspase-3 ELISA. In brief, A2780 human ovarian cancer cells (OCCs) were treated with medium alone (negative control), 5% ethanol (second negative control), Staurosporin (positive control apoptosis inducer), or different concentrations of CBD (sigma) for 24 hours. Then the media overlaying the OCCs were removed and 250 pg protein samples from each treatment group was subjected to Caspase-3 Elisa (Boster Bio.). OCC death and morphological changes due to CBD- induced apoptosis was noted. CBD-induced apoptosis was evident by the caspase-3 release. In addition to CBD, we are currently studying the anti-cancer effects of KY grown hemp extract.

Biography

Dr. Wasana K. Sumanasekera is a native of Sri-Lanka. She migrated to the US in 1991 after finishing her B.S. degree. Upon completion of 2 M.S. degrees, Dr. Sumanasekera obtained her Ph. D. in Molecular Toxicology from Pennsylvania State University. Then she pursued a post-doctoral career and a fellowship from American Heart Association. Currently she works as an associate professor of Pharmaceutical Sciences, in the College of Pharmacy, Sullivan University. She is involved in multi-disciplinary research in the areas of Toxicology / Substance Abuse, Cell and Molecular Biology, and Cardiovascular Physiology. She has published several articles in leading peer-reviewed journals.

Page 30: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Technical Session IV

Discovery of novel reactivators for DFP-inhibited acetylcholinesterase using in silico pharmacophore modeling

A.K. Bhattacharjee1*, E. Marek2, H.T. Le2, R. Ratcliffe2, J. C. DeMar2, D. Pervitsky2 and R.K. Gordon2

1Department of Microbiology & Immunology, Georgetown University School of Medicine, Washington DC, USA2Walter Reed Army Institute of Research, Maryland, USA

Abstract

Organophosphorus (OP) compounds such as tabun, soman, DFP, sarin, cyclosarin and pesticides (paraoxon, chlorpyrifos, tetraethyl pyrophosphate: TEPP) are highly toxic group of compounds. Tabun (GA) is the most toxic and developing antidotes is a challenging problem. However, since tabun is a G–simulator, less toxic G-simulators like DFP can be used for experimental analysis to discover new reactivators. Applying the rational on our previously reported in silico pharmacophore model (Chem Res & Toxicol, 2010, 23, 26-36) for reaction efficacy of oximes against tabun, we discovered 17 novel non-oxime reactivators of DFP-inhibited AChE through virtual screening of an in-house database and two commercial databases, Maybridge and ChemNavigator. All these non-oximes contained a nucleophile group in lieu of the oxime moiety, none was reported before to have potent reactivation efficacy. Efficacy (kr) of all the 17 non-oximes were found to be within 10-fold of 2-PAM in an eel DFP-inhibited AChE in vitro assay. One the non-oximes showed in vivo efficacy comparable to 2-PAM against brain symptoms for DFP-induced neuropathology in guinea pigs. In addition two other non-oximes showed promising results with no major differences in brain restoration of AChE activity compared to 2-PAM after DFP exposure. Since a serious loss of cholinergic function in the central nerves system contributes significantly to the cognitive symptoms associated with AD and other advanced age related brain diseases, our results on competitive OP-inhibited AChE reactivation and inhibition behavior of non-oximes may be useful further studies for development of neurologic therapeutics.

Biography

Dr. Apurba K. Bhattacharjee is an Adjunct Professor at the GUMC, Washington, DC, USA. His research is in the field of Computer Aided Drug Design (CADD) that includes quantum chemistry, pharmacophore modeling, and virtual screening of compound databases for discovery of novel bioactive agents. His past works involved discovery programs of antimalarials, antileishmanials, insect repellents and counter agents against organo-phosphorus (OP) poisoning at WRAIR. After Ph.D. in Chemistry from NEHU (India), he did postdoc at the IToDyS, University Paris 7, France with Professor J.-E. Dubois in CADD. His current interest is antiviral discovery targeting specifically Zika and dengue viruses. He has authored and coauthored 127 peer reviewed international publications including four book chapters and five patents.

Study design and analysis for multistage clinical proteomic studies

Irene Suilan Zeng

University of Auckland, New Zealand

Abstract

The presentation will discuss the design issues in clinical proteomics studies and provides specific suggestions for addressing these questions when using the standard guidelines for the planning. Author provides extensions for designs in clinical proteomics studies from the current guidelines such as STROBE-ME.

Two methods for the sample size estimation will be introduced. The first method is used for the planning of a clinical proteomic study at the discovery or verification stage; the second method is proposed for the systematic planning of a multi-stage design.

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 30

Page 31: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 31

The second part of the presentation introduces three approaches to analyzing the clinical proteomic study and provides analyses for the two case studies of clinical proteomic discoveries.

Biography

Dr. Irene Zeng is a postdoctoral fellow working in the Statistics department, University of Auckland. Dr Zeng’s research interests are statistical methods in proteomics and inter-omics, biomedicine,  clinical and community research. She has worked as a mathematical statistician in Statistics New Zealand, biostatistician in Auckland city hospital (2005-2008), Center for Clinical Research Middlemore hospital and a senior biostatistician of Counties Manuka district health board (2008-2016). Irene’s master and doctorate research investigated statistical methods for biomarker and proteomic studies. Dr. Zeng has also founded the i-StatDome® online datalab (onlinedatalab.org.nz) with her computer science colleague in 2015.

Discovery of novel small molecules and targets for neurodegenerative diseases through yeast based phenotypic screening

B. A. Pandya*, M. Lucas, I. Wrona, K. Ozboya, P. Tivitmahaisoon, D. Kouvchinov, D. Tardiff, J. Piotrowski, B. Vincent, C.Y. Chung, V. Khurana, B. L. Bourdonnec, R. Scannevin and K. Rhodes

Yumanity Therapeutics, Cambridge, MA, USA

Abstract

Neurodegenerative diseases (i.e. Parkinson’s disease, Alzheimer’s disease, and Dementia with Lewy Bodies) are critical unmet medical needs that are projected to grow in impact as the global population ages. Traditional drug discovery approaches have yet to produce disease modifying treatments. A common biological etiology of many of these diseases involves the misfolding of endogenous proteins. Pioneering work in the Lindquist labs1 has demonstrated that disease relevant toxicity caused by protein misfolding may be modeled in yeast cells.

Yumanity’s proprietary discovery platforms synergize yeast based phenotypic screening and target identification with patient derived stem cells to find novel biological targets and new chemical leads. Initial efforts of our target agnostic high throughput phenotypic screens have yielded small molecule hits that are being advanced to central nervous system active leads. Herein we will describe the Yumanity’s discovery platform along with preliminary in vitro and in vivo data of our small molecule leads.

Biography

Dr. Bhaumik Pandya is a Ph.D. medicinal chemist, with 10 years of experience across various disease areas and organizational sizes. He has managed and mentored scientists internally and externally. His contributions has come in the form of individual designer/synthesizer as well as project team leader. The impact of his work has been captured either in peer-reviewed journal articles, invited lectureships or patents.

Page 32: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Novel phosphatidylinositol 4-kinase IIIβ inhibitors as potential therapeutics

Radim Nencka1*, Ivana Mejdrová1, Dominika Chalupská1, Pavla Plačková1, Christin Müller2, Michal Šála1, Adriana Baumlová1, Hubert Hřebabecký1, Eliška Procházková1, Milan Dejmek1, Dmytro Strunin1, Jan Weber1, Gary Lee3, Helena Mertlíková-Kaiserová1, John Ziebuhr2, Gabriel Birkus3 and Evzen Boura1

1Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Gilead Sciences & IOCB Research Centre, Czech Republic2Institute of Medical Virology, Justus Liebig University Giessen, Germany3Gilead Sciences, Inc., Foster City, CA, USA

Abstract

Phosphatidylinositol 4-kinase IIIβ (PI4KB) is indispensable for replication of various positive-sense single stranded RNA viruses (or (+)ssRNA) viruses, which hijack this cellular enzyme to remodel plasmatic membranes inside infected cells and to set up functional replication machinery on such modified membrane structures. Therefore, inhibition of this particular PI4K isoform leads to arrest of viral replication. In our pilot study, we focused on a thorough structure-activity study starting from a HTS hit compound and identified a series of selective PI4KB inhibitors. One of these derivatives was co-crystalized with the target enzyme. Subsequently, we prepared a series of new derivatives, which were rationally designed based on two distinct structural types of inhibitors that were both shown to bind in the ATP binding site of this enzyme. The obtained “hybrid” structures excel not only in outstanding inhibitory activity, but also proved to be highly selective to PI4KB in comparison with other protein and lipid kinases. Thus, our prototype compounds exert low nanomolar or even subnanomolar activities against PI4KB without significant inhibition in wide kinome screen at 1µM. Our crystallographic studies unveiled the exact position of the side chains and explain their extensive contribution to the inhibitory activity. The obtained inhibitors were tested in a panel of (+)ssRNA viruses and proved to possess profound antiviral effect against hepatitis C virus (HCV), human rhinovirus (HRV) and coxsackievirus B3 (CVB3).

Biography

Dr. Radim Nencka is a group leader at the IOCB Prague. He is also a lecturer at Palacký University, Olomouc. He got his master’s degree in pharmacy from Faculty of Pharmacy, Charles University - his thesis was done at University of Crete, Heraklion, Greece (Prof. Manolis Stratakis). He obtained Ph.D. in organic chemistry from Faculty of Science, Charles University, Prague (Dr. Hubert Hřebabecký, Prof. Antonín Holý group at IOCB Prague) and spent his postdoctoral stay in Laboratory of Medicinal Chemistry, University of Ghent, Belgium (Prof. Serge Van Calenbergh). His main research interest is synthesis of novel compounds for treatment of viral diseases.

Herbal remedies for mandibular fracture healing

Shadab Mohammad

Department of Oral and Maxillofacial Surgery, King George’s Medical University, Lucknow, Uttar Pradesh, India

Abstract

Purpose: When a bone is fractured it is usually necessary to employ a mechanical means to reduce and maintain the fragments in position. However, healing of the fracture is governed by biological principles, with which the mechanical measures must be coordinated to the end, such that a satisfactory bony union and restoration of form and function are obtained. We have studied the effect of Cissus quadrangularis (Harjor) and Ocimum sanctum (Tulsi), in the healing of mandibular fractures.

Materials and Methods: A total of 29 cases having a fracture in the body of the mandible were included in the study and divided into three groups. Groups A and B were treated with Ocimum sanctum and Cissus quadrangularis, respectively, and fracture healing was assessed with biochemical markers and the bite force. Group C was the control group.

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 32

Page 33: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 33

Results: The period of immobilization was the lowest in the Group A followed by Group B. A significant increase in alkaline phosphatase and serum calcium was seen in Group B. The tensile strength in terms of the biting force was the maximum in cases of Group B.

Conclusion: We conclude that Cissus quadrangularis and Ocimum sanctum help in fracture healing, and use of such traditional drugs will be a breakthrough in the management and early mobilization of facial fractures.

Key words: Alkaline phosphatase, Cissus quadrangularis, Ocimum sanctum

Chemoecology guided discovery of drug leads from South China Sea marine invertebrates

Yue-Wei Guo

Shanghai Institute of Material Medica, Chinese Academy of Sciences, Shanghai, China

Abstract

Natural products have played a significant role in the drug discovery process throughout the last hundreds years. It is generally accepted that there are two ways to explore the pharmaceutical potentialities of natural products. The first way is so called bioassay guided isolation of bioactive natural products; the second one is so called random screening methodology. In fact, every procedure could be only partially satisfactory. Apart the above mentioned two solutions, an alternative way could be a good choice through studying the compounds that really play a biological role in the organism where they are present. This could be the starting point to discover other biological potentialities. Of course, to perform studies like these one needs a careful selection of promising biological systems and, also, the close collaboration among chemists, biologists and pharmacologists.

Trying to follow this bio-chemical approach some years ago we started to investigate marine nudibranchs that are extremely interesting from an ecological point of view. In fact, these mollusks are completely devoid of the mechanical protection of the shell. But, in spite of this apparent vulnerability, they are rarely victims of predators. This is due to a series of defensive strategies that include the use of chemicals that either derive from their food habits or are biosynthesized “de novo” by themselves.

In this lecture we will report the recent chemical studies on opisthobranch molluscs collected from South China Sea. All work has been performed in close collaboration with marine biologists who have correctly submitted the biological problems to the chemical analysis, and with pharmacologists who have carried out bioassay based on the clue provided through chemoecology studies.

Biography

Dr. Yue-Wei Guo has completed his Ph.D from Naples University and postdoctoral studies from both Istituto di Chimica Biomoleculare-CNR, Italy and Hokaido University, Japan. He is the Professor of Shanghai Institute of Materia Medica-CAS. He has published more than 390 papers in reputed journals and serving as editorial board members of several reputed national/international journals. 

Page 34: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

A chemist’s view of animal disease models for drug screening

Takashi Tsukamoto

Johns Hopkins School of Medicine, Baltimore, MD, USA

Abstract

The credibility of efficacy data obtained from animal disease models has been the subject of much debate lately, driven by increasing concerns over their inability to predict clinical outcome. The challenge of replicating human diseases in animals has never been greater as our unmet medical needs shift to a broad spectrum of more complex and poorly understood diseases. Yet, we continue to use animal disease models as the key experiments for selecting clinical candidates. The prevailing thoughts on this paradox appear to be “There is no better alternative”. There may be another way out of this, namely, skipping them altogether. With the right approach in place, this may not be as ludicrous as it sounds.

Biography

Dr. Takashi Tsukamoto, Ph.D. is Associate Professor of Neurology at Johns Hopkins University and the Director of Medicinal Chemistry at Johns Hopkins Drug Discovery Program. Dr. Tsukamoto received his Ph.D. degree in Chemistry from Tokyo Institute of Technology and pursued postdoctoral studies in Medicinal Chemistry at the University of Michigan. Prior to joining Johns Hopkins, Dr. Tsukamoto has held a variety of research positions in the pharmaceutical industry including Guilford Pharmaceuticals, MGI Pharma, and the Eisai Research Institute. During the course of his career, he has served as a lead medicinal chemist in a number of drug discovery projects.

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 34

Page 35: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 35

Young Researchers Forum

Modulating supramolecular nanostructures cross-linked by hyaluronic acid derivatives for drug delivery

Tânia. F .G. G. Cova*, Sandra. C. C. Nunes and Alberto. A. C. C Pais

CQC, Department of Chemistry, University of Coimbra, Coimbra, Portugal

Abstract

Host-guest interactions have been extensively explored for developing nanostructures to improve solubilization, transportation and targeting of therapeutic agents [1]. Supramolecular assemblies based on hyaluronic acid derivatives and stabilized by host-guest interactions have been constructed to be used drug delivery applications [2].

Hyaluronic acid (HA) is considered an attractive building block that provides a promising platform to encapsulate/conjugate several drugs and to construct nanostructures with superior properties and tailored functionality, by incorporating, for example, nanoparticles within the respective network [3]. It has been hypothesized that the cellular uptake and efficiency of tumor-targeted drug delivery may increase using HA-based nanostructures as drug carriers, thus fostering new formulation opportunities [4].

In this work the free energy patterns associated to the formation of supramolecular assemblies cross-linked by hyaluronic acid derivatives, bearing monomeric β-cyclodextrin and adamantane moieties, are investigated by molecular dynamics simulations and potential of mean force calculations. The free energy, enthalpy, and entropy changes along the host-guest association coordinate are evaluated for a set of four model systems with varying structural complexity. The estimated stability of the inclusion complexes is highly affected by the fit and orientation of the guest into the cavity, the size and nature of the guest moieties and by dessolvation effects. A comprehensive picture of structural and thermodynamic motifs is provided, in which water contributions appear to govern binding stability. The modulation of the network interaction sites can be achieved imposing amphiphilic substituents of different sizes, with direct transposition for the design of improved properties in supramolecular structures based on these complexes.

[1] T.F.G.G. Cova, S.C.C. Nunes, A.A.C.C. Pais, Phys. Chem. Chem. Phys. 19 (2017) 5209-5221.

[2] L.K. Widjaja,  M. Bora, P.N.  Chan,  V. Lipik,  T.T. Wong,  S. S. Venkatraman. Hyaluronic acid-based nanocomposite hydrogels for ocular drug delivery applications. J. Biomed. Mater. Res. A. 102 (2014) 3056-65.

[3] S. Merino, C. Martín, K, Kostarelos, M. Prato, E. Vázquez, Nanocomposite Hydrogels: 3D Polymer–Nanoparticle Synergies for On-Demand Drug Delivery. ACS Nano 9 (2015) 4686–4697.

[4] G. Saravanakumar, V.G. Deepagan, R. Jayakumar, J.H Park, Hyaluronic acid-based conjugates for tumor-targeted drug delivery and imaging. J. Biomed. Nanotechnol. 10 (2014) 17-31.

Extended release of venlafaxine hydrochloride using montmorillonite based excipients as a delivery vehicle

Shilpa Jain* and Monika Datta

Department of Chemistry, University of Delhi, Delhi, India

Abstract

Patients suffering from depression generally require long - term treatment using anti-depressants. Venlafaxine hydrochloride (VF) is one of the most effective third generation drug. This highly water soluble drug has a short steady state elimination half-life of about 4 hours, due to which this drug needs to be administrated several times

Page 36: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

in a day in order to maintain its required concentration in the blood plasma. The drug is available in the market as immediate-release and the extended release formulations do not extend the time period long enough to cover the recommended requirement.

The present work discusses the outcomes of a new class of drug delivery vehicles using Montmorillonite (Mt) as excipient. The product/s with highest encapsulation efficiency was selected for the in-vitro drug release studies. The cumulative release of VF in the gastric fluid for the pure drug, Mt-VF complex, Organo Mt–VF complex, Mt-PLGA-VF nanocomposites and Mt-VF- Alginate beads were found to be 100% (in 5.5h), 83% (in 12h), 63 (in 12h), 62% (in 12h), 13% (in 12) respectively. The cumulative release of VF in the intestinal fluid for the pure drug, Mt-VF complex, Organo Mt –VF complex, Mt-PLGA- VF nanocomposites and Mt-VF- Alginate beads were found to be 100% (in 3.5h), 50% (in 12h), 45% (in 12h), 100% (in 12h), 16% (in 12h) respectively. Thus, there is possibility of making formulation/s using Mt based excipient which is likely to provide with better patient compliance as repeated intake (every 3-4 hours) of drug will not be necessary.

Biography

Dr. Shilpa Jain has obtained her Ph.D. degree in chemistry in 2015 from department of chemistry, University of Delhi, India. She has been awarded with senior and junior research fellowship by CSIR to pursue Ph. D. and all India post graduate scholarship to pursue master’s degree in Chemistry. She has published 5 papers as first author in reputed international journals. She has presented several research papers in national and international conferences in India. She is the recipient of Young Scientist Award of Indian Chemical Society in 2014. She is currently working as an assistant professor at Shivaji College, University of Delhi, India.

Bioanalytical method development, validation & pharmacokinetics study of 5-fluorouracil loaded nanoparticles

Saurabh Srivastava

Department of Oral & Maxillofacial Surgery, King George’s Medical University, India

Abstract

Background: 5-fluorouracil (5FU) is a chemotherapeutic agent against different types of cancer.5FU loaded with nanotechnology can enhance efficacy over conventional drawback of 5-FU, such as short half-life, toxicity, low bioavailability and non-selective action. Pharmacokinetic profile of this advanced nano-formulation is needed to correlate with overall ADME (absorption, distribution, metabolism and excretion) process.

Purpose of the study: In this study, development of the HPLC-UV method & validation perform in expression of specificity, precision, sensitivity, accuracy and stability of the developed 5-fluorouracil nanoparticles (5-FUNPs) and correlates & collects the valuable pharmacokinetics data.

Methodology: 5-FUNPs formulated with polymer poly lactic co-glycolic acid with Oil-in-water/solvent evaporation. Characterizations of nanoformulation performed which included particle Size, stability studies. Analytical method developed and validated from HPLC-UV and applied to pharmacokinetic parameters.

Results: The calibration curve plotted for 5-FUNPs was linear at 267 nm. The lower limit for the quantification found 10.13ng/mL. The size of 5-FUNPs was between 137 + 0.97 to 193 + 0.93 nm & zeta potential between 0.27 + 0.08 to 0.29 + 0.07mv on the side of positively charged. The highest peak for drug concentration, Cmax was 3.235±0.78 mg/L at highest time point, Tmax 7.21±2.52 hrs. The AUC (0-96) & AUC (0-∞) showed 8.89±4.98 mg/L-h and 9.57±3.77 mg/L-h respectively and t1/2 was 22.98±3.73 hrs.

Conclusion: The results show a simple, specific, sensitive and stable HPLC-UV method for the quantitative determination of 5-FUNPs in plasma and successfully applied to the pharmacokinetic study after oral administration in rats.

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 36

Page 37: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 37

The facial-Re(CO)3 core based luminophores as non-toxic bio-imaging agents

Deepak Gupta1*, Vikash Singh2 and Biprajit Sarkar3

1Department of Chemistry, Bhaskaracharya College of Applied Sciences, University of Delhi, India2Zentrum für Infektionsmedizin, Institut für Mikrobiologie und Tierseuchen, Freie Universität Berlin, Germany 3Institut für Chemie und Biochemie, Anorganische Chemie, Freie Universität Berlin, Germany

Abstract

Organometallic coordination complexes are fast gaining attention as potential multimodal compounds for biological applications such as anticancer-agents, cell-imaging and pharmaceutical agents. Particularly, the rhenium-carbonyl complexes have come-up as biocompatible luminescent probes for organelle specific cell-imaging applications. A series of dinuclear facial-Re(CO)3 based transition metal complexes have been demonstrated as luminophores for live cell-imaging. These complexes can be synthesized by one pot synthetic methodologies in high yield and purity. The characterization of these complexes was done by FTIR, 1H NMR, ESI-MS and elemental analysis. These compounds exhibit properties such as high solubility, high molar extinction coefficients in the visible region (ε = 40000−50000 M-1cm-1), room temperature luminescence, kinetic inertness, photo-stability, large Stock’s shift, high cellular uptake and low cell-toxicity which are favorable for cell-imaging applications. Herein, we present a series of amino-quinonoid ligands containing facial-Re(CO)3 based luminescent, dinuclear metal complexes that show strong visible light absorption ability and are capable of selectively co-localize into the lysosomes of HeLa cells.

Biography

Dr. Deepak Gupta has obtained his Ph.D. degree in the field of supramolecular chemistry based on facial-rhenium carbonyl complexes and metallacycles from Department of Chemistry, University of Delhi on a Council of Scientific and industrial Research, Govt. of India funding in 2014. He is a recipient of the renowned DAAD fellowship to work in Germany. He has published ten research papers in the international journals of repute. Presently, he is working on his first major research project funded by the Department of Science and Technology, Govt. of India under the Young Scientist Scheme.

Drug addiction and oral health complications in India

Sana Farooqui

Department of Oral & Maxillofacial Surgery, King George’s Medical University, India

Abstract Oral Health complications related with drug abuse are increasing on a rapid pace in India. Drug use gives birth

to number of Oral health complications such as periodontal diseases, Oral precancer, oral cancer, bruxism, tooth loss etc. Not only has this but it also given lodgement to several social and psychological problems such as violence, anxiety, depression and stress. The habit of drug addiction has posed a great threat to mankind as it destroys the individual both physically and psychologically. There are several reasons held responsible for drug addiction. A person becomes addicted to it in order to alleviate from distress and anxiety. Sometimes due to the several social issues such as poverty, unemployment, unrest when a person does not find anyway in order to cope from such problems then he turns towards addiction. In India drugs such as Opiates, Cocaine, Nicotine, and Cannabis are currently used. These drugs are highly addictive and harmful and the fact remains very clear that once a man start its use then he becomes habitual of taking it and so it becomes difficult for him to get free from it. Published evidences on drug addiction and health issues emphasizes that drug addiction is just a sickness and through certain preventive measures such as counseling of addicts, support of family and friends and governmental policies this problem can be curbed to a great extent This paper aims to review oral health complications associated with drug use, factors associated for its use and focuses on preventive strategies which support the addicts in changing their attitude towards more positive and healthy lifestyles. Keywords: Opiates, Cocaine, Nicotine, Cannabis.

Page 38: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

3November 11

Saturday

Page 39: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 39

Technical Session V

High throughput screening identifies an anti-ebola lead compound

Ryan P. Bennett1*, Ryan A. Stewart1, Elena N. Postnikova2, Húanyíng Zhou2, Shuīqìng Yú2, Julie Dyall2, Laura Bollinger2, Peter B. Jahrling2, Jens H. Kuhn2 and Harold C. Smith1, 3

1OyaGen, Inc., Rochester, NY, USA2IRF-Frederick/DCR/NIAID/NIH, MD, USA3University of Rochester Medical Center School of Medicine and Dentistry, Rochester, NY, USA

Abstract

High throughput screening (HTS) of libraries of drug-like compounds requires a primary assay as a screening method and a critical path of methods for selecting on-target antiviral compounds with low cytotoxicity that work through a known mechanism of action. To that end, an essential viral function is the ideal drug target. We targeted the Ebola virus (EBOV) matrix protein (VP40) because (i) it is required for EBOV particle production, (ii) it is the most abundantly expressed of the seven EBOV genome-encoded proteins, (iii) it is required as a cytoplasmic protein to regulate viral genome transcripts, (iv) the VP40 homodimer crystal structure is known, and (v) VP40 protein-protein interaction domains have been determined. Using our screening methods, we identified a compound we call “Oya1” that inhibits EBOV VP40 localization to membranes where VP40 needs to localize to form EBOV particles. Secondary assays revealed that Oya1 also causes a decrease in EBOV virion-like particle (VLP) release from cells. In addition, Oya1 also blocked EBOV RNA-dependent RNA polymerase (L) activity in an EBOV minigenome reporter assay. Other viral polymerases were also effected as Oya1 effectively suppressed the activity of EBOV, LASV, MERS-CoV, and VACV at IC50 concentrations ranging from 0.01–0.09 µM with the therapeutic indexes ranging from 0.62–53.8. These findings suggest that Oya1 has dual targeting capacity against both EBOV VP40 and the L protein (along with other viral polymerases), therefore making it a highly attractive potential medical countermeasure.

Biography

For the past ten years, Dr. Ryan P. Bennett have been the lead scientist at the biotech company OyaGen, Inc. In this capacity, Dr. Bennett oversee assay development for HTS for various therapeutic targets from compound libraries. Dr. Bennett have developed and optimized various secondary screens and functional endpoint analyses to validate hits for which leads are currently in preclinical development. Third party contract relationships for medicinal chemistry, live virus studies, ADMET and animal testing for lead-to-drug candidate development are also critical aspects that Dr. Bennett coordinate.

Hypoxia inducible factor 1: a urinary biomarker of kidney disease

Movafagh S1*, Raj D2, Sanaei-Ardekani M3, Bhatia D1, Vo K4, Mahmoudieh M4, Rahman R3, Kim EH3 and Harralson A1

1Department of Pharmacogenomics, Bernard J. Dunn School of Pharmacy, Shenandoah University, Ashburn, VA, USA2Department of Nephrology, George Washington University Division of Kidney Diseases and Hypertension, Washington, DC, USA3Kidney and Hypertension Specialists, Manassas, VA, USA4Shenandoah University, Bernard J. Dunn School of Pharmacy, Ashburn, VA, USA

Abstract

Identifying non-invasive biomarkers of kidney disease is valuable for diagnostic and therapeutic purposes. Hypoxia Inducible Factor 1 (HIF-1) expression is known to be elevated in the kidneys in several renal disease pathologies. Elevation of HIF-1a gene and protein expression has been shown to be protective in acute kidney injury, however, long term HIF-1a elevation has been associated with tubular interstitial fibrosis. Furthermore,

Page 40: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 40

HIF-1a enhancing drug therapy is currently in clinical trials for treatment of anemia of chronic kidney disease. To avoid detrimental effects of prolong HIF-1a exposure, assessment of baseline and post therapy HIF-1a renal expression is warranted. In this study, we proposed that urinary HIF-1a mRNA level measurement may be a suitable index for expression of this protein in Chronic Kidney Disease (CKD). We quantified and compared HIF-1a mRNA levels from urinary pellets of CKD (stages III-V) and healthy subjects. We found that HIF-1a mRNA is readily quantifiable in urine pellets and its expression was significantly higher in CKD patients compared with healthy adults. To assure that urinary HIF-1a mRNA is of kidney origin, we examined co-localization of HIF-1a mRNA with kidney specific markers in urine cells. We showed that the urinary HIF-1a mRNA comes primarily from cells of renal origin and its levels are correlated with mRNA of kidney specific protein. Our data suggest that urinary HIF-1a mRNA is a potential biomarker in CKD and can be non-invasively assessed in patients for diagnostic and therapeutic purposes.

Biography

Dr. Shahrzad Movafagh is an associate professor of pharmacology and pharmacogenomics at Shenandoah University School of pharmacy and INOVA center for personalized health (ICPH). Dr. Movafagh obtained her Pharm.D. degree from University of California San Francisco with an emphasis on pharmacogenomics. She further obtained her Ph.D. in pharmacology from Georgetown University Medical Center. During her training she also received an NRSA postdoctoral fellowship in pharmacology from NIH. She has been a faculty member at Shenandoah University for 7 years. The focus of her research has been studying the effects of hypoxic signaling in cardiac and renal pathology and therapeutics.

Neointimal Caveolin‐1 loss may in part facilitate metabolic shift in pulmonary arterial hypertension

R. Mathew*, J. Huang, W.H. Frishman and M.H. Gewitz

Department of Pediatrics and Medicine, New York Medical College, Valhalla, NY, USA

Abstract

Recent studies indicate that the metabolic shift may have a pivotal role in the irreversibility of PAH. We have previously reported loss of endothelial caveolin‐1 (cav‐1) followed by enhanced expression of cav‐1 in the smooth muscle cells (SMC) and subsequent neointima formation in human PAH. We have further shown that by subjecting monocrotaline (M)‐treated rats to hypoxia (H), the PAH progression is accelerated, leading to neointima formation with scant cav‐1, and near normal eNOS expression. We hypothesized that the scant cav‐1 and normal eNOS expression in the neointima leads to oxidative stress. Nrf2 activated during oxidative stress induces anti‐oxidants and facilitates aerobic glycolysis providing “survival milieu” for proliferating cells. We examined male Sprague‐Dawley rats (Controls, M and M + H groups). At 4 weeks, M group revealed significant PAH and right ventricular hypertrophy compared with C; and a further increase in PA pressure in M+H gr. Cav‐1 and eNOS expression was significantly reduced in the M gr, but not in M+H group. In the M+H gr only 8% of arteries exhibited endothelial cav‐1 and 61% revealed enhanced cav‐1 expression in SMC; but the neointimal cells exhibited scant cav‐1 expression. The expression of Nrf2, MnSOD and Glut1 were significantly increased in the M+H gr. The cav‐1 loss and near normal eNOS in neointimal cells lead to oxidative stress, resulting in the activation of Nrf2 which induces MnSOD; thus, creating an “antioxidant milieu”. Glut1 increases glucose uptake and participates in aerobic glycolysis, thus, facilitates the survival of the proliferating cells.

Biography

Dr. R. Mathew received MBBS degree from All India Institute of Medical Sciences, New Delhi; trained in Pediatrics and Pediatric Cardiology in UK (Hospital for Sick Children, London; Brompton Hosp, London). Pediatric Cardiology Fellowship at Boston Children’s Hospital under Dr. Alexander Nadas). She started basic science research in mid‐career. Her main focus has been pulmonary hypertension. She is currently at New York Medical College NY, as an Associate Professor in Pediatrics and Physiology.

Page 41: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 41

P-coumaric acid, main active compound isolated from Bambusae Caulis in Taeniam suppresses cigarette smoke–induced lung inflammation

Woogyeong Kim and Jinju Kim*

Department of Korean Physiology, College of Pharmacy, Kyung Hee University, Kyungheedae-ro, Dongdaemun-gu, Seoul, Republic of Korea

Abstract

Cigarette smoke (CS) is a major risk factor for pulmonary inflammatory diseases. Our previous study demonstrated that a Bambusae Caulis in Taeniam inhibits CS–induced pulmonary and intestinal inflammation. p-coumaric acid (p-CA) is the main active compound isolated from Bambusae Caulis in Taeniam which has been reported to possess anti-oxidant, anti-mutagenic, anti-ulcer and anti-inflammatory activities. However, therapeutic effect of p-CA on CS-induced lung inflammation has not been reported. In present study, we investigated the effect of p-CA on CS-induced lung inflammatory response. C57BL/6 mice were treated with p-CA (5 and 10 mg/kg) or roflumilast (ROF) via oral injection 2 h before CS expose 5 days per week for 4 weeks. The body weight and the number of macrophages and neutrophils in bronchoalveolar lavage fluid (BALF) were measured. The levels of relative inflammatory factors including IL-1ß, IL-6, TNF-α and MCP-1 were confirmed by ELISA. The lung histological changes were examined by hematoxylin and eosin (H&E) staining. Also, the mRNA expressions of MCP-1, MIP-3α and GCP-2 and the protein levels of MMP-3, MMP-9 and MMP-12 were evaluated by real-time PCR or Western blotting. Our results showed that p-CA attenuated CS-induced lung inflammation with inhibited the macrophages and neutrophils infiltration and an amount of IL-1ß, IL-6, TNF-α and MCP-1 in BALF. In addition, p-CA decreased inflammatory cells infiltration and airspace enlargement into lung tissue. Furthermore, mRNA and protein levels were down-regulated in p-CA treated mice. In conclusion, these data’s demonstrated that p-CA suppressed against CS-induced lung inflammation.

Stability and activity of nano-crystalized Camptothecin

Honglei Zhan, Tina Jagtiani and James Liang*

Department of Biomedical Engineering, Chemistry, and Biological Sciences, Charles V. Schaefer School of Engineering and Sciences, Stevens Institute of Technology, Hoboken, NJ, USA

Abstract

Camptothecin (CPT) is a potential anticancer agent against a variety of cancers. However, its clinical trials were hampered due to the drug’s poor aqueous solubility, instability and unexpected side effects. To combat these problems, the most outstanding approach is the preparation of drug nanocrystals, which improves the dissolution rate suggested by the Noyes-Whitney equation and the nano-scaled nature enables it to take good advantage of EPR effect. However, because of the lack of target specificity, CPT nanocrystals exhibited lower cellular uptake and showed limited therapeutic efficacy towards specific types of tumors. We functionalized CPT nanocrystals through spontaneous polymerization. Besides the added benefits attributed to the polymer layer such as the improved dissolution rate and stability of CPT, the polymer coating provided a versatile platform for introducing secondary components for diverse functional applications. This newly prepared formulation had dramatically improved stability, dispersion property, dissolution rate and showed targeting capability to biomarker over-expressed cancer cells.

Biography

Dr. James Liang is a Professor at the Department of Biomedical Engineering, Chemistry, and Biological Sciences, Stevens Institute of Technology. Previously, Dr. Liang served as professor at Tsinghua University (Beijing) and Associate Research Professor at University of Michigan (Ann Arbor). Prof. Liang has over 100 peer-reviewed publications and 16 patents. The research interests of Prof. Liang are in functional materials, drug delivery, and bioactive peptides.

Page 42: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 42

The clinical implementation of molecular biomarkers in glioma

Jessica L. Fleming*, Erica Hlavin Bell and Arnab Chakravarti

Department of Radiation Oncology, Comprehensive Cancer Center-Arthur G. James Cancer Hospital, The Ohio State University, OH, USA

Abstract

Gliomas are the most common primary malignant brain tumor, affecting around 20,000 individuals each year in the United States. While gliomas only account for a little over 1% of all cancers diagnosed each year, the survival times for these patients are dismal. Historically, gliomas were categorized and treated based on histopathological and grading criteria. Recently the World Health Organization updated the classification system for gliomas incorporating molecular biomarkers such as IDH mutation and 1p19q co-deletion status. This has boosted us into the era of precision medicine for glioma patients. While these advancements mark a turning point in the management of gliomas, there is still a great need to further stratify patients into clinically relevant risk groups and importantly into specific treatment groups in order to increase patient survival times. The objective of our research is to identify novel prognostic and predictive molecular biomarkers in glioma and develop clinically relevant models that are useful to both patients and physicians in making treatment decisions. We performed high-throughput molecular profiling on clinical trial biospecimen from the NRG Oncology cooperative group and identified individual molecular biomarkers as well as multi-marker models that improved upon existing glioma classification systems. We were also able to address questions regarding treatment due to the well-annotated clinical data from these biospecimen. With this information, patients will have more accurate survival probabilities and physicians will be able to objectively guide treatment decisions for individual patients.

R01CA108633, 1RC2CA148190, U10CA180850-01, 1R01CA169368, Brain Tumor Funders Collaborative Grant, OSU CCC Award

Biography

Dr. Jessica Fleming is a senior postdoctoral researcher in the laboratory of Dr. Arnab Chakravarti in the department of Radiation Oncology at the Ohio State University. She received her B.S. in Biology from Ashland University in 2006 and her Ph.D in Molecular, Cellular, and Developmental Biology from the Ohio State University in 2012. Currently her research is focused on identifying prognostic and predictive molecular biomarkers in glioma that can be clinically utilized. This molecular information also enables our group to study mechanisms of treatment resistance and aggressive cancer phenotypes, allowing us to identify and validate putative therapeutic targets.

Restoring FMRP expression: an alternative treatment strategy for fragile X syndrome

Daman Kumari1*, Manju Swaroop2, Wei Zheng2 and Karen Usdin1

1Laboratory of Cell and Molecular Biology, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA 2Therapeutics for Rare and Neglected Diseases, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA

Abstract

Fragile X syndrome (FXS), a leading form of inherited cognitive disability, is caused by the expansion of a CGG-repeat tract in the 5’-untranslated region of the Fragile X Mental Retardation-1 (FMR1) gene to >200 repeats. This causes transcriptional silencing and the loss of the FMR1 protein product, FMRP. There is no cure for FXS. Current efforts for drug development rely on normalizing aberrant signaling pathways. Given that FMRP functions in many pathways, restoring FMRP expression in FXS patients may be an approach to ameliorating disease symptoms that is also worth pursuing. We use two complementary strategies to identify compounds that are capable of restoring FMR1 gene expression in FXS. The first strategy is to try to elucidate the mechanism of FMR1 gene silencing so that specific genes and/or pathways can be targeted for gene reactivation. This work suggests that

Page 43: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 43

it might be possible to block FMR1 silencing specifically, thus avoiding the side effects that would result from global inhibition of gene silencing. The second strategy uses unbiased high throughput screening (HTS) and a sensitive and quantitative assay for FMRP detection that we have developed to identify compounds that increase FMRP expression. Pilot screens with FXS neural stem cells identified a small number of compounds that modestly increased FMRP expression. Screens of larger compound libraries might identify suitable lead compounds for FMR1 gene reactivation in patients.

Biography

Dr. Daman Kumari is a Staff Scientist in the Laboratory of Cell and Molecular Biology at the National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health. Dr. Kumari received her Ph.D. in 1997 from the Jawaharlal Nehru University in New Delhi, India, and postdoctoral training at the National Institutes of Health. Her research goal is to elucidate the mechanism of repeat mediated FMR1 gene silencing in fragile X syndrome which will help design treatment strategies based on restoration of gene expression.

Synthesis of ofornine mimics from natural product l-vasicine as anti-hypertensive agents

Abdul Rouf1*, Mushtaq A. Aga2, Brijesh Kumar2, Sheikh Rayees2, Gurdarshan Singh2 and Subhash C. Taneja2

1Department of Chemistry, University of Florida, Gainesville, FL, USA2Bio-organic Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, J&K, India

Abstract

Hypertension is a multifactorial, mutagenic vascular disorder and a major risk factor for many other cardiovascular diseases1. According to a WHO report, hypertension is prevalent in one out of three adults worldwide2 and is one of the most common chronic disorders3,4. It is a serious health problem in developed as well as developing countries. Although several effective pharmacological agents are commercially available for its treatment,5 the problems of cardiovascular disease related to hypertension still persist6.

In this report, a library of vasodilator Ofornine mimics was synthesized from l-vasicine which was isolated from Adhatoda vasica. The synthesized analogs were studied for the structure-activity relationship and in vivo screening for anti-hypertensive action in Wistar rats. Most of the analogs were observed to possess anti-hypertensive effect; however, the duration of the effect was variable and mostly transient. One of the analogs named as (S)-(3-hydroxypyrrolidin-1-yl)(2-(pyridin-4-ylamino)phenyl)methanone showed a significant decrease in blood pressure in a dose dependent manner whose maximal response lowered to 79.29±4.26 mmHg of SBP and 62.55±2.9 mmHg of DBP at 10 mg/kg intravenous dose. Further, the significant anti-hypertensive effect of this analog lasted for about 2.5 h at 10 mg/kg dose. The SAR studies clearly showed that the amide, hydroxyl and pyridine ring play an important role in the activity.

O

N

NH

NN

NOH

R

l-vasicineAdhatoda vasica

O

N

HO

N NN

R

Page 44: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 44

Biography

Dr. Abdul Rouf was born in India. He completed his Ph.D. from CSIR-Indian Institute of Integrative Medicine, Jammu, India, in Organic Chemistry. He is currently working as a Postdoctoral Research Associate in the University of Florida, USA and is working on the synthesis of polyketides and small molecule pheromones and hormones isolated from C. elegans.

(Endnotes)

1 Akinlua, J. T.; Meakin, R.; Umar, A. M.; Freemantle, N. PLoS One 2015, 10, e0140021.

2 (a) Katki, K. A.; Dipette, D. J. 2006, 138-145; (b) The World health statistics 2012 report, WHO, 2012.

3 Akinlua, J. T.; Meakin, R.; Umar, A. M.; Freemantle, N. PLoS One 2015, 10, e0140021.

4 Schutte, A. E.; Van Rooyen, J. M.; Huisman, H. W.; Kruger, H. S.; Malan, N. T.; De Ridder, J. H. J. Hum. Hypertens. 2003, 17, 29-35.

5 Phillips, M.I. Hypertension 2001, 38, 543-548.

6 Lape, H.; DeFelice, A.; Bailey, D.; Pharmacologist (Abstr.) 1983, 25, 101.

Synthesis and biological evaluation of some novel N-methylenebenzenamine derivatives as selective acetylcholinesterase inhibitors to improve learning and memory

Sushant Kumar Shrivastava*, Pavan Srivastava, TVR Upendra and Prabhash Nath Tripathi

Department of Pharmaceutics, Indian Institute of Technology (Banaras Hindu University), Varanasi, UP, India

Abstract

Series of some 3,5-dimethoxy-N-Methylenebenzenamine and 4-(methyleneamino)benzoic acid derivatives comprising of N-Methylenebenzenamine nucleus were designed, synthesized and assessed for their acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) inhibitory activity thereby improving learning and memory in rats. The IC50 values of all derivatives on AChE and BChE were determined, where selective inhibitions of all compounds were observed against AChE in comparison with standard donepezil. The enzyme kinetic study of compound 4 revealed an uncompetitive inhibition of AChE. The docking studies of the most active compound 4 confirmed its interaction with the important active-site gorge residues Phe330 and Trp279 responsible for its high affinity for AChE. The lacking of the BChE inhibition can be considered due to a wider gorge binding site and absence of important aromatic amino acids. The compound 4 elicited a considerable decrease in escape latency and was comparable to donepezil via Spatial Reference Memory (SRM) and Spatial Working Memory (SWM) in-vivo models. Thus, our results support the cognition-enhancing potential of compound 4. Ex vivo study confirmed their brain AChE inhibition abilities.

Biography

Prof. S. K. Shrivastava has around 23 years’ experience in teaching and research, as a researcher, he has contributed a lot in the field of synthetic medicinal chemistry, drug design and molecular modeling and published around 73 research papers in international and national journals. Some national and international awards and honors are to his credit. His h-index and i10-index are 13 and 18, respectively. Presently, Prof S. K. Shrivastava is involved in synthesizing some novel molecules (NCE) to treat neurodegenerative and inflammatory disorders. He has completed projects having rational significance.

Page 45: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 45

Consensus comparative modeling to construct structural models of human C-C chemokine receptor 4 (CCR4) to discover small-molecule therapeutics to treat cancer metastasis

Jeffery J. Bullock* and Xiang S. Wang

Department of Pharmaceutical Science, Chinese Institute, Howard University, Washington, D.C., USA

Abstract

Cancer can spread rapidly throughout the human body, either locally to nearby normal tissues or regionally to lymph nodes and distant organs. Human CC chemokine receptor 4 (CCR4) plays fundamental roles in the development, homeostasis, and function of the immune system, and has profound effects on endothelial cells involved in angiogenesis or angiostasis. A defucosylated, humanized monoclonal antibody targeting CCR4, Mogamulizumab, has been approved in Japan for the treatment of adult T-cell leukemia-lymphoma (ATL). Considering the high cost of antibody treatment, there is an urgent need to develop small-molecule based, anti-CCR4 therapeutics. Comparative Modeling has become a powerful tool to address the issue of lack of crystal structures for important drug targets such as human CCR4. Through a new algorithm in Maestro 11, we constructed consensus homology models of CCR4 using multiple templates with success. Initially, the FASTA amino acid sequence for human CCR4 was loaded and pair-wise alignment was conducted. A BLAST query was submitted to NCBI PDB to find homologs, and returned 4MBSas a potential template. The query based on multiple sequence alignment resulted in our second template of 5T1A. Our consensus comparative models obtained from different templates meet above stereo-chemical criteria specified, while a premium binding pocket was identified consisting of Lys191, Glu283, Thr259, 195, 284, 247, 257 and Phe182. The new consensus comparative modeling algorithm provides an option toward more accurate determination of both the global structure and binding sites for human CCR4, which can facilitate the discovery of small-molecule CCR4 antagonists.

In vitro efficacy of a novel RNA-dependent RNA polymerase inhibitor against a broad range of RNA viruses

Elena N. Postnikova1*, Ryan P. Bennett2, Huànyīng Zhōu1, Yíngyún Caì1, Shuīqìng Yú1, Julie Dyall1, Laura Bollinger1, Peter B. Jahrling1, Jens H. Kuhn1 and Harold C. Smith2, 3

1IRF-Frederick/DCR/NIAID/NIH, MD, USA2OyaGen, Inc., NY, USA3University of Rochester Medical Center School of Medicine and Dentistry, Rochester, NY, USA

Abstract

Nucleoside analogs have successfully been used to treat a number of serious infections caused by pathogenic RNA viruses, including hepatitis C virus. Typically, nucleoside analogs are phosphorylated once inside the cells by cellular kinases, thereby mimicking precursors for nucleic acid biosynthesis. As a result, viral RNA-dependent RNA polymerases incorporate the analog into nascent RNA chains, often causing chain termination. High throughput screening for antagonists of Ebola virion assembly identified a new nucleoside analog (Oya1) that is currently under development at OyaGen. Oya1 was evaluated in vitro for cytotoxicity and efficacy against cowpox virus (CPXV), vaccinia virus (VACV), Ebola virus (EBOV), Marburg virus (MARV), Lassa virus (LASV), Middle East respiratory syndrome coronavirus (MERS-CoV), and human immunodeficiency virus 1 (HIV-1). Huh7 cells, monocyte-derived macrophages, and NR596 cells were exposed to Oya1 for 1 h prior to virus exposure. Infection rates were measured by IFA or eGFP expression. The percentage of virus-infected cells was measured by high-content imaging. Cytosine β-D-arabinofuranoside, cidofovir, and toremifene citrate were used as positive controls. The new compound effectively suppressed the activity of EBOV, LASV, MERS-CoV, and VACV at IC50 concentrations ranging from 0.01–0.09 µM. CC50 concentrations ranged from 0.04–0.6 µM and the therapeutic index ranged from 0.62–53.8. The compound had modest effects against CPXV and MARV and no effect against HIV-1.

Biography

Currently, Dr. Postnikova’s work focuses on BSL-4 viral pathogens and includes developing and optimizing

Posters Presentations

Page 46: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 46

assays for antiviral compound validation and high-content imaging. Over the course of her career, she created culture collections for microorganisms for developing mycoherbicides against poppy opium, she collaborated with CIMMYT to screen wheat cultivars, and participated in project design to utilize NGS methods as a diagnostic tool at the USDA. Dr. Postnikova also initiated projects with the former Soviet Union bioweapons research institutes as part of the US Cooperative Threat Reduction program to counter biological and chemical terrorism and reduce proliferation threats.

Transition metal and photocatalyst-free perfluoroalkylation reactions of organic sulfides with relevant biological activity

Damian E. Yerien*, Sebastian Barata-Vallejo, M.L. Uhrig and Al Postigo

Departamento de Química Orgánica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina

Abstract

Perfluoroalkyl sulfides have found wide applications, such as in liquid crystal displays, for oleophobic surface coatings, as well as in the synthesis and development of potential hypotensive agents for clinical use containing SRF moieties such as Losartan derivatives (Dup 753).

Particularly, the case of pefluoroalkylated thiosugar derivatives, where the sulfur atoms in thioaldoses make the interglycosidic bonding stable to glycosidases. The incorporation of a hydrophobic chain as aglycone, such as a perfluorinated residue, could have a high impact in protein–carbohydrate recognition events which take place between sugars and glycosidases, glycosyltransferases and also lectins. For these reasons, fluorinated thiosugars have a great potential as tools in the field of Glycobiology. To that effect, we proceeded to studying the S-perfluoroalkylation of thiocarbohydrates. The reactions of the sugar thioaldoses 2,3,4-tri-O-acetyl-1-thio-β-D-glucuronic acid methyl ester and 2,3,4,6-tetra-O-acetyl-1-thio-β-D-galactopyranose under our standard reaction conditions, afford 75 and 83 % yields of SC4F9 -substituted products, respectively. The disaccharide 2´,3´,4´,6´,2,3,6-hepta-O-acetyl-1-thio-β-D-cellobiose affords product in 50 %. However, the reaction of galactoside-disulfide (1,1´-bis-(2,3,4,6-tetra-O-acetyl-1-thio-β-D-galactopyranose)) did not afford any S-C4F9 substitution product.

In this work, we will show the serendipitous

discovery of a radical initiation event from visible light irradiation of a complex between TMEDA and I2 (i.e. :[(TMEDA) I.I3]) which triggers an electron-catalyzed process towards the synthesis of perfluoroalkyl sulfides of biological potential in improved substitution yields. We will also discuss the applications to such perfluorinated thiosugar derivatives.

Biography

Damian E. Yerien was born in Buenos Aires (Argentina) and holds a degree in Biochemistry (2014). He is currently a Ph.D. student at the University of Buenos Aires, studying radical fluorination and fluoroalkylation reactions in heterogeneous and microheterogeneous media under the direction of Professor Dr Alberto Postigo. He is a holder of a National Research Council (Conicet) scholarship at the Faculty of Pharmacy and Biochemistry- University of Buenos Aires.

Ferumoxytol nanoparticles enable imaging of brain tumor-associated neuroinflammation

Heather L. McConnell*, Daniel L. Schwartz, Cymon N. Kersch, Leslie L. Muldoon and Edward A. Neuwelt

Oregon Health and Science University, Portland, OR, USA

Abstract

In the brain tumor microenvironment, non-neoplastic inflammatory cells can influence the survival, proliferation and infiltrative activity of cancer cells. The ultrasmall superparamagnetic iron oxide nanoparticle ferumoxytol can be used as a contrast agent for magnetic resonance imaging (MRI) of brain vasculature, intracerebral tumors and neurological lesions. Ferumoxytol leaks across the blood-brain barrier in a rat model of acute neuroinflammation 24h after administration and MRI signal changes correlate with ferumoxytol uptake by macrophages, microglia and activated astrocytes. The purpose of this study was to test the hypothesis that delayed MRI would demonstrate uptake of ferumoxytol by inflammatory cells in and around the tumor in rat models of intracerebral metastasis. Immune-compromised nude rats received intracerebral implantation of human metastatic cancer cells including H460 non-small cell lung carcinoma and HCC1954 breast carcinoma. Post-ferumoxytol T1-weighted scans showed pockets of hypointensity dispersed throughout the tumor and, occasionally, a distinct rim of signal dropout at the tumor border, combined with hyperintensity extending

Page 47: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 47

1-2 mm around the tumor mass. Post-ferumoxytol T2-weighted scans showed mottled signal dropout throughout the tumor, while T2* scans showed intense hypointensity throughout the tumor mass extending into surrounding brain. Immunohistochemistry demonstrated ferumoxytol in macrophages within necrotic areas of the tumor and astrocytes in peritumoral brain, with no staining in tumor cells. We conclude that delayed ferumoxytol MRI provides a biomarker of the inflammatory component of intracerebral tumors.

Biography

Heather is currently a Ph.D. student in the Neuroscience Graduate Program at the Oregon Health and Science University in Portland, Oregon. She holds a Master’s degree in Clinical Research and is interested in translational biomedical science and science communication, particularly in the context of neuropathology.

A genome-wide screening of antifungal drug target genes in fission yeast

Sol Lee¹*, Miyoung Nam¹, Ah-Reum Lee¹, Ji Ahn Choe¹, Hye Jin Lee¹, Dong-Uk Kim² and Kwang-Lae Hoe¹

¹New Drug Discovery and Development, Chungnam National University, Republic of Korea²Aging Research Center, Republic of Korea

Abstract

Major antifungal drugs, selectively targets ergosterol biosynthesis by inhibiting the enzyme squalene epoxidase and consequently disturbs fungal cell membrane. To search novel targets of antifungal drug, we performed a genome-wide screening using drug-induced haploinsufficiency screening in ~5,000 heterozygote Schizosaccharomyces pombe (S. pombe) gene deletion library. we identified antifungal drug-hypersensitive mutants using both microarray and a plate confirmation assay. The drug-sensitive mutants were tested for sensitivity to lyticase, representative hypersensitive mutants were identified, and suggesting that these mutants were defective in cell wall integrity and add ergosterol to hypersensitivity mutants abrogated growth inhibition by antifungal drug.

Biography

Sol Lee is a Ph.D. student in Graduate School of New Drug Discovery at Chungnam National University studying under Professor Kwang-Lae Hoe.

She performed a genome-wide screening in ~5,000 heterozygote S. pombe gene deletion library. When new target of drugs are found, it is used for cell line experiment or assay.

Protein kinase selectivity analysis based on the topological water networks

Myeong Hwi Lee*, Hye Ree Yoon, Kwang-eun Choi and Nam Sook Kang

Graduate School of New Drug Discovery and Development, Chungnam National University, Republic of Korea

Abstract

Protein kinases are associated with several diseases including various cancers and inflammatory diseases. Accordingly, kinases have emerged as potential targets for the drug discovery and development. They generally cause selectivity problems when designing ligands that bind ATP-binding sites using ATP as the same substrate. Furthermore, it’s difficult to define or compare the characteristics of kinases in which the ATP binding site has a similar sequence in addition to a highly conserved structure. To solve this problem, we attempted to analyze topological water network (TWN) in ATP binding site of protein kinases. In TWN analysis, molecular dynamics (MD) simulation was initially carried out on kinase in aqueous environment and then water molecules forming the hydrogen-bonded network in the binding site were selected and analyzed. The positions and frequencies of water networks were considered in the analysis. Herein, we describe the TWN analysis results based on some case studies, IRAK4 and DYRK1A. These results revealed that TWN analysis could be exploited for the drug discovery and repositioning.

Biography

Myeong Hwi Lee received her bachelor’s degree in chemistry as well as biochemistry from Chungnam National University. Then, she completed her master’s degree in the field of molecular modeling and cheminformatics at the graduate school of new drug discovery and development, Chungnam National University. Currently, she is pursuing Ph.D. degree in the same department under the guidance of Prof. Nam Sook Kang. Her research work mainly focuses on the computer-aided drug design.

Page 48: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 48

Enhancement of expression level and stability of hEC-SOD3 by conjugating human serum albumin

Eun Yeong SONG1*, Jong-Woo Jeong1, Joo Young GO1, Tae-Yoon Kim2, Tae- Sung Koo1 and Sungsub KIM1

1Graduate School of New Drug Discovery and Development, Chungnam National University, Korea2College of Medicine, The Catholic University, Korea

Abstract

Reactive oxygen species induce oxidative stress leading to cellular damage and a variety of human diseases. To remove these reactive oxygen species, cellular antioxidant system exists. Among them, extracellular superoxide dismutase (SOD3) is an antioxidant enzyme with excellent efficacy against asthma and excessive inflammatory reaction, and thus is expected as a strong biomedicine candidate substances. However, there are many difficulties in obtaining large amounts of active recombinant human SOD3 (hEC-SOD3).

Previous studies have shown that hEC-SOD3 is mostly composed of an inactive apo-enzyme when overexpressed in HEK293 cell and hEC-SOD3 rapidly lost its activity after purification, but the activity was maintained when metal ions, BSA (Bovine serum albumin) and PEG (Polyethylene glycol) was added. Furthermore, since the plasma half-life of hEC-SOD3 is as short as 1.54h, it is expected that the intact use of hEC-SOD as a medicine is limited.

Here, we prolonged half-life by fusing human serum albumin (HSA) to the N-term of hEC-SOD3. The expression level of HSA-conjugated SOD3 was higher than hEC-SOD3 in HEK293 cell and its anti-oxidative activity was similar to that of hEC-SOD3. In addition, the activity was maintained for over 14 days without addition of BSA or PEG, and the plasma half-life was extended to 13.3 hours. HSA-conjugated SOD3, developed through this study, may help overcome clinical barriers to drug development of hEC-SOD3 as a bio-drug.

Biography

Eun Yeong SONG is a Ph.D student at Graduate school of New Drug Discovery and Development, Chungnam national university, Korea. She is studying about protein drug and antibody drug. And her main experiments are DNA, RNA, and Protein work.

In vitro glucuronidation of icaritin by human liver microsomes, human intestine microsomes and expressed UDP-glucuronosyltransferase enzymes: contribution of individual enzymes, reaction kinetics and activity correlation

Zhihong Yao1,2*, Zifei Qin1-3, Xiaodan Hong1,2, Yi Dai1,2, Baojian Wu1-3 and Xinsheng Yao1-3

1College of Pharmacy, Jinan University, China 2Guangdong Provincial Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, China3Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, China

Abstract

Icaritin is a natural polyphenol with many biological effects. The present study aimed to characterize icaritin glucuronidation by pooled human liver microsomes (HLM) and pooled human intestine microsomes (HIM), and to determine the contribution of individual UDP-glucuronosyltrans-ferase (UGT) enzyme to icaritin glucuronidation. Glucuronidation rates were determined by incubating icaritin with uridine diphosphate glucuronic acid (UDPGA)-supplemented microsomes. Kinetic parameters were derived by appropriate model fitting. Relative activity factors (RAF) and activity correlation analysis were performed to identify the main UGT isoforms. Kinetic profiling revealed that formation of icaritin-3-O-glucuronide (G1) and icaritin-7-O-glucuronide (G2) in HLM well modeled by the substrate inhibition equation, whereas they followed the classical Michaelis-Menten kinetics in HIM. UGT1A3, 1A7, 1A8, 1A9 and 2B7 were mainly responsible for catalyzing the formation of G1 and G2. Icaritin 3-O-glucuronidation (G1) was significantly correlated with chenodeoxycholic acid (CDCA) glucuronidation (r = 0.787, p = 0.002), propofol glucuronidation (r = 0.661, p = 0.019) and zidovudine (AZT) glucuronidation (r = 0.805, p = 0.002), respectively. Similarly, icaritin 7-O-glucuronidation (G2) was also correlated with CDCA glucuronidation (r = 0.640, p = 0.025), propofol glucuronidation (r = 0.592, p = 0.043) and AZT glucuronidation (r = 0.661, p = 0.019), respectively. Furthermore, UGT1A3, 1A9 and 2B7 contributed 37.5, 33.8 and 21.3% for G1 in pooled HLM, respectively. Also, UGT1A3, 1A9 and 2B7 contributed 34.3%, 20.0% and 8.6% for G2 in pooled HLM, respectively. Take altogether, icaritin was subjected to significant glucuronidation, wherein UGT1A3, 1A7, 1A8, 1A9 and 2B7 were main contributing enzymes.

Page 49: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 49

Biography

Prof. Zhihong Yao focuses on bioactive component discovery based on Pharmacokinetics and Metabolism of Traditional Chinese Medicine prescription (TCMP) by LC-MS. Her interests is to discover the bioactive components of TCMP according to their pharmacokinetic character and metabolism pathways, and try to illustrate component-component interaction in complicated TCMP system. All these researches were supported by dozen of funds like National Natural Science Foundation and have been published in more than 40 papers on international and national academic journals (half of them embodied in SCI journal, while most of them published as first author/co-first author/corresponding author/co-corresponding author).

Identification of ESE-1 as a tumor suppressor in lung cancer

Zhiyuan Lou*, Taekyu Ha, Yanrui Xu and Seong-Ho Lee

Department of Nutrition and Food Science, College of Agriculture and Natural Resources, University of Maryland, College Park, MD,USA

Abstract

Lung cancer is most life-threatening cancer in the world due to poor prognosis and resistance to therapy. Targeted therapy using effective molecular targets is urgently required for prevention and therapy. Epithelial Specific ETS-1 (ESE-1) is an epithelial specific member of ETS transcription factor family. Although the association of ESE-1 with cancer was posed in several types of cancer model, the role of ESE-1 during lung cancer development was still unknown. In the current study, we observed that ESE-1 expression repressed the anchorage-independent growth of human non-small cell lung cancer (NSCLC) cells. In addition, ESE-1 expression repressed invasion and migration of human NSCLC cells. ESE-1 expression was repressed by treatment of TGF-β, and this decrease is associated with transcriptional downregulation of ESE-1 gene during the epithelial mesenchymal transition (EMT). Here we identify ESE-1 as a tumor repressor in tumorigenesis and metastasis of lung cancer.

Validated LC-MS/MS method for recombinant human SOD3 (rhSOD3) in mouse plasma and its application to pharmacokinetic study

Jong-Woo Jeong* and Tae-Sung Koo

Graduate School of New Drug Discovery and Development, Chungnam National University, Korea

Abstract

A simple and robust LC-MS/MS method was developed and validated for quantification of rhSOD3 in mouse plasma. Before the analysis, plasma samples were purified by using Ni-NTA magnetic bead and digested by recombinant porcine trypsin to produce signature peptide for detection. Then, sample digests were simply cleaned-up with acidified methanol containing 3% of formic acid. Chromatographic separation was achieved utilizing the Agilent ZORBAX C18 reverse phase column with a binary mobile phase consisting of A: 10 mM ammonium formate, 0.1% formic acid, and 5% acetonitrile and B: 10 mM ammonium formate, 0.1% formic acid, and 95% acetonitrile as a gradient eluent, at a flow rate of 0.3 mL/min. Analytes were detected using positive ion MRM mode. The precursor to product ions (Q1 → Q3) selected for SOD3 signature peptide (AVVVHAGEDDLGR) and IS peptide (AVVVHGGDDDLGR) were m/z 669 → 969 and 655 → 941, respectively. The developed method was validated according to the requirements of FDA guidance. The established method showed linearity for rhSOD3 in the range 5 to 1,000 µg/mL with a correlation co-efficient >0.996. The results for the inter-day and intra-day accuracy (<9.17%) and precision (<9.56%) of quality control samples were within the acceptable range. Stability test indicated rhSOD3 was stable up to 180 days. Significant matrix effect was not observed. Consequently, the analytical method was successfully applied to a pharmacokinetic study of SOD3 in male ICR mice.

Biography

Jong-Woo Jeong is a PhD student and research assistant of Lab. of pharmacokinetics in Graduate school of new drug discovery and development (GSDD), Chungnam national university. Mr Jeong graduated from Kangwon national university in 2011, majoring in biology. And then, Mr Jeong has started MS/PhD integrated course in Chungnam national university majoring in Pharmacokinetics since 2013.

Page 50: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

United Scientific Group (USG), an expert-driven initiative led by the editors asso-ciation and the advisory board which includes academicians, researchers, and in-

dustry leaders across various fields of research. USG provides broad range of services in the fields of science and technology including publishing, conducting world class scientific events, and holding highly interactive and proficient world forums.

About United Scientific Group

USG Editors Association

Founding President Founding Vice-President

Kenneth Blum, Ph.D., DHLUniversity of Florida, USA

Sayon Roy, Ph.D., FARVOBoston University, USA

The scientific industry involved in networking, organizing meetings and publishing scholarly journals is increasing constantly in order to meet the ever changing demands of emerging new concepts and subjects in different fields of science. Rigorous, meticulous policies and guidelines are essential to maintain the highest standards of scientific excellence. USG is fortunate to have the United Scientific Group Editors Association (USGEA) that serves this role. USGEA is an association of United Scientific Group Journals editors with diverse backgrounds and professional experience, who seek to foster cooperation and communication among editors, improve editorial standards, promotes the concept of self-criticism, self-regulation in scholarly publishing, and encourage research on the editorial principles and practices of publishing.

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 50

Page 51: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

Founding Members

Ahmad Salehi, MD, Ph.D.Stanford Medical School

USA

Rajendra Badgaiyan, MDWright State University

USA

Jin Jun Luo, MD, Ph.D.Temple University

USA

Dawei Li, Ph.D.Shanghai Jiao Tong

University, China

Mun Yhung Jung, Ph.D.Woosuk University

South Korea

Reza Hakkak, Ph.D.University of Arkansas for Medical Sciences, USA

Clinical Sciences & Drug Discovery (CSDD-2017) | November 9-11, 2017 | Washington, DC, USA 51

Page 52: CLINICAL SCIENCES DRUG DISCOVERY...Ongoing debates and controversies whether inflammation is protective (‘friend’) for preventing cancer or it is a cause of carcinogenesis (‘foe’)

We wish to see you at CSDD-2018

t

# 8105, Rasor Blvd - Suite #112, PLANO, TX 75024, USAPh: +1-408-426-4832, +1-408-426-4833; Toll Free: +1-844-395-4102; Fax: +1-408-426-4869

Email: [email protected]: www.unitedscientificgroup.com/conferences/csdd/

UNITED Scientific Group