Scientific Program

Conference Series Ltd invites all the participants across the globe to attend 3rd Antibodies and Bio Therapeutics Congress & B2B Las Vegas, Nevada, USA.

Day 1 :

Keynote Forum

William Whitford

GE Healthcare, USA

Keynote: Digital biomanufacturing supports mAb production

Time : 09:30-10:00

Conference Series Antibodies-2017 International Conference Keynote Speaker William Whitford photo
Biography:

William Whitford is Strategic Solutions Leader, GE Healthcare in Logan, UT with over 20 years’ experience in biotechnology product and process development. He joined the company as an R&D Leader developing products supporting protein biological and vaccine production in mammalian and invertebrate cell lines. Products he has commercialized include defined hybridoma and perfusion cell culture media, fed-batch supplements and aqueous lipid dispersions. An invited lecturer at international conferences, William has published over 250 articles, book chapters and patents in the bioproduction arena. He now enjoys such activities as serving on the Editorial Review Board for Bioprocess International.

Abstract:

The recent explosion in monitoring, analytics and new computing capabilities initiated the revolution we call digital bio-manufacturing. These, combined with such advances as artificial intelligence, automation and robotics are evolving our concept of manufacturing in general. The PredixTM system employed by GE is an example of how manufacturers can use continuous data acquisition, cloud technology and advanced analytics to provide a platform for the industrial internet. Changes are occurring from product development and factory operations to materials supply. These involve increased monitoring, data handling, connectivity, computer power, control algorithms and automation. DB promises such things as real-time optimization of the manufacturing process based on such highly valuable criteria as projected product quality and batch profitability. The IIoT, big data and the cloud now synergize with such initiatives as lean PPD, SCADA and DCS to advance our process control capabilities. The ability to elucidate metabolic bottlenecks in expression and PT processing combined with our heightened process control capability is opening new doors of opportunity in manufacturing efficiency and product quality. Advances in high throughput whole genomic RNA interference screening is advancing our process-control potential in mAb-relevant mammalian cell lines.

Keynote Forum

Robert M Stroud

University of California San Francisco, USA

Keynote: Engineering a 30-year-old intracellular channel: Twitching a voltage sensor with a taste of pharmacology

Time : 10:00-10:30

Conference Series Antibodies-2017 International Conference Keynote Speaker Robert M Stroud photo
Biography:

Robert M Stroud is Professor of Biochemistry and Biophysics, University of California in San Francisco. He focuses on the molecular basis for function of trans-membrane transporters and channels, and on structure-assisted drug discovery. He has contributed to fundamental mechanisms of receptor proteins, lipid-protein interactions, enzymes and protein-RNA recognition. He has obtained his BA and MA in Natural Sciences from the University of Cambridge (UK), his PhD is from University of London (JD Bernal). From a Post-doctoral and Professorship in Biological Chemistry at the California Institute of Technology, he came to UCSF. His research involves structural determination engineering and function of molecules and cells using X-ray crystallography, electron-cryo microscopy, computational simulations, spectroscopy, super-resolution optical microscopy. He is a Member of the National Academy of Sciences, a Fellow of the American Academy of Arts and Sciences, a Fellow of the Royal Society of Medicine (UK), Fellow and Former President of the US Biophysical Society.

Abstract:

Proteins in plasma membranes are subject to large trans-membrane potential of ~200,000 V/cm. This affects all such proteins. Some proteins harness voltage sensors to detect and respond to changes in potential. Such proteins are found also in internal organelles where they are oft en more intricately regulated. Hence by protein engineering it is possible to tune the responses to conducted ions and to voltage. I describe a channel in the TPC1 class 1, 2, 3. Because of its regulation by calcium ions, it can be restored to its closed resting state. A comparison between this and all other voltage sensors in full-length channels, which are generally activated at zero Volts, shows for the first time in a voltage sensitive channel how a voltage sensor responds, to effectively move charges across the membrane? A conformational transition indicates a mechanism that may be general. It accounts for the measured gating charge that defines how many charges must transition from one side to the other. Conformation changes lead to channel opening.

  • Antibody Engineering
    Antibodies: Medical Applications
    Immunotherapy and Immune Checkpoints
    Protein Engineering
    Genetic Engineering

Session Introduction

Ron L Martin

Nutrigenetics Unlimited, Inc., USA

Title: The gene expression evidence-base and its relevance to translational applications including prevention

Time : 10:30-10:55

Speaker
Biography:

Ron L Martin has received his BS and MS degrees in Food Science and Nutrition from Chapman University in Orange, California. He has worked for more than 35 years in the nutraceutical and food industries, including Hunt-Wesson/ConAgra, Plus Products, the William T Thompson Co., and the Nutrilite division of Alticor, where he served as a Senior Research Scientist in Nutrilite's New Concepts group. He has been a Professional Member of the Institute of Food Technologists (IFT) and is the President of Nutrigenetics Unlimited, Inc., which he founded in 2007. He is also one of the Founding Members of the International Society of Nutrigenetics/Nutrigenomics

Abstract:

Examples of the emerging gene expression evidence-base will be described, along with online tools for increasing both awareness and utility of the increasingly actionable information. This can be helpful to the entire spectrum of potential users, including students and other members of the public. Such tools are increasingly important as the body of literature continues to expand rapidly, making it progressively more difficult to identify and manage the evidence-base for making more fully informed choices. Online resources will be described, including the usefulness of standardized terminology which allows creation of subtopic listings for any given topic or for any given combination of topics – including for genes and gene variants. Beyond diagnosis and treatment alone, such approaches also allow identification and exploration of prevention opportunities, and as well as optimizations for both physical and mental health. Gene-environment examples include nutrition, pharmaceuticals, pollution, lifestyle, social environment, etc. Because nutrition applies to everyone without exception, it can become a useful introductory "archetype" for promoting greater engagement and greater genetics/health literacy.

Break: B2B, Networking & Refreshment Break 10:55-11:15 @ Foyer

Yuri L. Lyubchenko

University of Nebraska Medical Center, USA

Title: Nano-assembly of amyloid β peptide: Role of the hairpin fold

Time : 11:15-11:40

Speaker
Biography:

Yuri L Lyubchenko is a Professor of Pharmaceutical Sciences at the University of Nebraska Medical Center, USA. His research focuses on understanding fundamental mechanisms underlying health and disease, which are keys to developing new and more effective diagnostics and medications. This primarily basic research allows him not only to identify new drug targets for small molecule drugs, it also leads to development of the nanotools and methods to discover novel approaches for diagnostic, treatment and disease prevention and to more rapidly determine their effi cacy at the molecular level.

Abstract:

Statement of the Problem: Increasing evidence suggests that the self-assembly of amyloid β(Aβ) protein underlies the early onset of Alzheimer’s disease (AD). Given that small Aβ nano-assemblies (oligomers) are the most neurotoxic species, they have become the major target for the development of treatments and early diagnostic tools for AD. However, advances surrounding this are blocked by the lack of structure intrinsic to Aβ oligomers, as they are transient states of Aβ aggregation kinetics; making traditional structural approaches non-amenable. We have previously developed single-molecule approaches capable of probing of aμo dimers. Here, we extended our approaches to higher order oligomers. We hypothesized that the folding pattern of amyloid protein defines the aggregation pathway. 

Methodology & Theoretical Orientation: In this study, we tested this hypothesis using Aβ( 14-23) peptide in linear form and its tandem assembled in the hairpin-type shape. We combined two experimental approaches and molecular dynamics simulations to characterize molecular interactions and the stability of complexes between Aβ (14-23) hairpin and Aβ (14-23) monomer, as well as the interactions between two hairpins.

Findings: The lifetime measurements demonstrate that the Aβ (14-23) hairpin and a Aβ(14-23) monomer assemble in a very stable complex when compared with homologous ensembles. We measured the strength of hairpin-hairpin and hairpin-monomer interactions which demonstrated that the hairpin-monomer interaction is stronger compared with the hairpin-hairpin assembly; data that is fully in line with the lifetime measurements. Aggregation studies demonstrate that the Aβ(14-23) monomer formed fibrils and the hairpin formed spherical structures. However, their mixture formed neither fibrils nor spherical structures, but rather disk shaped nanostructures.

Conclusion & Significance: Overall, our study provides new insight into the role of the monomer structure on the self-assembly process that contributes to the formation of disease aggregates. Importantly, the developed experimental approaches and validation approaches for computational analyses are not limited to amyloid proteins, but can also be applied to other molecular systems.

Barry L Stoddard

Fred Hutchinson Cancer Research Center, USA

Title: De novo design and characterization of circular tandem repeat proteins

Time : 11:40-12:05

Speaker
Biography:

Barry L Stoddard has research interests in the structure, mechanism and engineering of proteins and enzymatic catalysts for basic research and biomedical applications. His lab conducts wide-ranging research on gene-specific endonucleases, the creation of suicide genes for biotech and medical applications, structural enzymology and protein engineering. He has coauthored over 160 research articles and reviews in these fields since 1990. In 2004, he was one of several recipients of the Newcomb Cleveland Prize from the American Association for the Advancement of Science (AAAS) for published work in the field of Protein Engineering.

Abstract:

De novo protein design offers access to regions of protein fold space that have not been sampled during natural evolution, regions that may contain solutions to novel structural and functional challenges. De novo design can also produce proteins that are highly robust and stable-favorable attributes for subsequent functionalization by rational means. We recently reported the development and experimental validation of a computational method for design of closed tandem repeat protein architectures (a closed repeat architecture is one in which the N- and C-termini are juxtaposed). We have used this method to create a set of alpha-helical solenoid repeat proteins whose bundle handedness is opposite that found in analogous natural proteins. These designed proteins have toroidal architectures with up to 24 repeats, variable thickness and central pore dimensions ranging from 0 to 60 Angstroms in diameter. We are now expanding the topological diversity of the repeat units to access new geometries and functionalizing the designed scaffolds to create reagents for biomedical and industrial applications. A motivating hypothesis behind this work is that designed will prove to be valuable protein scaffolds for protein array and display by virtue of their solubility, stability tunable symmetry, modularity, and self-reinforcing architectures.

Erik D Foehr

BioTether Sciences, USA

Title: BioTethering: A novel approach to engineering therapeutics proteins

Time : 12:05-12:30

Speaker
Biography:

Erik D Foehr is a Biotechnology Expert with over 15 years of research and development experience, resulting in numerous publications, patents and innovations that propel the advancement of science, medical treatment and patient care. He has received his PhD in Physiology and Biophysics at the University of California, Irvine and Post-Doctoral Fellowship at the Gladstone Institute of Virology and Immunology. He has a strong scientific background, research capabilities and business acumen that drive effective laboratory operations and project management. His work includes contributions in the areas of regulated bioanalysis and medical device characterization. He has conducted numerous developments, validation and testing studies using a variety of analytical approaches. He has proven leadership of bioanalysis research and drug development projects focused on increasing corporate brand, value proposition and impact to the industry.

Abstract:

Protein therapeutics is a dominant and fast growing sector of the $400 billion global pharmaceutical market. There are over 200 marketed protein pharmaceuticals. Major opportunities exist to bring to market the next generation of biopharmaceuticals. In the era of personalized medicine and targeted drugs (immuno-oncology), novel approaches to engineering proteins/peptides are highly desirable. BioTether Sciences develops enzyme, antibody and peptide therapeutics for cancer, infectious diseases, autoimmune diseases, rare diseases and other unmet medical needs. BioTether Sciences uses novel approaches to increase safety, efficacy and targeting of biopharmaceuticals. The technology involves the tethering of proteins with polymer linkers or using high affinity interactions between ligand-receptor and antibody-antigen. Innovator therapeutics can be greatly improved using this approach or novel tethered therapeutics may be developed. Examples and case studies of engineered proteins will be provided. Peglyation or other polymer linker is used to connect proteins and peptides together thereby increasing molecular weight and bulk. This reduces clearance by the kidney and greatly improves drug half-life. The increased vacancy improves receptor binding. Epitope masking improves specificity of targeting to enhance safety. These approaches have been applied to human growth hormone and therapeutic antibodies. For example, human growth hormone may only last for minutes in the circulation without protein engineering. In another application, an antibody is masked by a tethered antigen that improves targeting.

Speaker
Biography:

Dhanesh Gadre works in the Purification Process Sciences Department at MedImmune LLC, USA. He designs, develops and executes protein purification processes for the development of biopharmaceutical products such as humanized monoclonal antibodies and recombinant proteins. He performs purification and biophysical characterization and formulation of antibodies and protein reagents in response to the needs of several departments using protein biochemistry and chromatography principles and methods. He also has expertise in analytical biochemistry techniques such as HPLC, gel electrophoresis etc. He has Master of Science degree in Chemical Engineering from Syracuse University, USA.

Abstract:

Endotoxins are complex molecules and a significant impurity risk present in the downstream purification processes. Trace amounts of endotoxins can cause immune responses in humans resulting in fever or hypertensive shock. Endotoxins also interfere with the cell based activity assays, impacting the selection of biopharmaceutical drug candidates. For these reasons, it is very important for academic, research and development labs and manufacturing facilities to ensure that protein samples and products are free of endotoxins. During biopharmaceutical production, endotoxins are usually cleared during the downstream purification process. However, if endotoxins interact with a protein of interest through electrostatic or hydrophobic interactions, they can become difficult to remove. Triton X-100 has showed promise in breaking the endotoxins-proteins interactions. However, in some cases Triton X-100 becomes ineffective at breaking these interactions and is therefore unable to remove endotoxins. In this study, we were able to identify a wash condition on chromatography media using a non-ionic detergent octyl-β-D-1-thioglucopyranoside (OTG). This detergent wash can reduce endotoxins from protein solutions to lower levels than Triton X-100 with similar or better protein recovery. Different classes of proteins were bound to different modes of chromatography media and then washed with a variety of detergents. OTG showed the most promising data among these detergents in reducing endotoxin levels with high protein recovery. We examined the mechanism of action to determine why OTG showed better endotoxin clearance ability than Triton X-100. Triton X-100 affects only hydrophobic interactions but OTG can affect hydrophobic as well as electrostatic interactions between proteins and endotoxins. We also showed the impact of the robust OTG on research cell based assays.

Break: B2B, Meet the Exhibitor & Lunch Break 12:55-13:45 @ Renaissance III
Speaker
Biography:

Kelly Yan has over 17 years of biopharmaceutical experience in the field of protein biochemistry focusing on protein expression, purification and characterization. Her passion has been on difficult-to-produce proteins, from kinases with different phosphorylation status and activities to protein complexes with conformational changes and functions. She has received her MD degree in Shanghai, China before she started her graduate studies in SFSU and research in Charles Craik Lab in UCSF.

Abstract:

Midkine (MDK) belong to a newly evolving family of secreted neurotrophic and developmentally regulated heparin-binding molecules, which also includes pleiotrophin (PTN). Both proteins are involved in promoting neurite outgrowth. Various studies have shown that MDK is involved in cell mitogenesis, transformation, survival, migration and angiogenesis. Moreover, MDK is over-expressed in a number of human cancers; therefore it has become an attractive target for developing cancer therapeutics. Active recombinant human MDK (rhMDK) can be a critical reagent for cell assay development as well as generation of small molecule inhibitors or antibodies for therapeutic drug discovery. MDK is a cysteine-rich 13 kDa protein containing five disulfide bonds (C15-C39, C23-C48, C30-C52, C62-C94 and C72-C104). Expressing and purifying such a protein can be challenging, mainly because the disulfide bonds have to be properly formed to preserve its activity. Traditionally, proteins with disulfide bonds are purified under a denaturing environment and then re-folded. In this study, we expressed (rhMDK) in the Escherichia coli Origami 2 (DE3) strain, which carries a (trxB-/gor522-) double mutation. Soluble rhMDK was expressed at a high level in this strain and protein purification required only one step of heparin affinity chromatography followed by a size exclusion polishing step, without an inclusion body preparation and refolding process. Seven milligrams of rhMDK with high purity were obtained from a 3 L culture. All 10 cysteines were confirmed to be engaged in correct disulfide bond linkages by mass spectrometry analysis. Activity of purified rhMDK was confirmed by a neurite outgrowth assay using rat cerebellar granule cells. Our study demonstrated a very simple and useful technique for producing disulfide bonds-containing bioactive protein with high purity.

Speaker
Biography:

SooJin Han has completed her PhD from both KIST and Hanyang University in Korea and Post-doctoral studies from University of Alabama and University of Florida in USA. She has been working as a Scientist of the Department of Cell Culture and Fermentation Sciences at MedImmune, a Maryland based biotechnology development enterprise owned by AstraZeneca. She has much experience and great knowledge in mAb and non-mAb production in prokaryotic and eukaryotic expression platforms and published several papers in reputed journals.

Abstract:

Secretion of heterologous proteins into E. coli cell culture medium offers significant advantages for downstream processing over production as inclusion bodies; including cost and time savings and reduction of endotoxin. Signal peptides play an important role in targeting proteins for translocation across the cytoplasmic membrane to the periplasmic space and release into culture medium during the secretion process. Alpha toxinH35L (ATH35L) was selected as an antigen for vaccine development against S. aureus infections. It was successfully secreted into culture medium of E. coli by using bacterial signal peptides linked to the N-terminus of the protein. In order to improve the level of secreted ATH35L, we designed a series of novel signal peptides by swapping individual domains of modifying dsbA and pelB signal peptides and tested them in a fed-batch fermentation process. The data showed that some of the modified signal peptides improved the secretion efficiency of ATH35L compared with E. coli signal peptides from dsbA, pelB and phoA proteins. In particular, one of novel signal peptides improved the yield of secreted ATH35L by 4-fold in the fed-batch fermentation process and at the same time maintained the expected site for signal peptide cleavage. Potentially, these new novel signal peptides can be used to improve the secretion efficiency of other heterologous proteins in E. coli. Furthermore, analysis of the synthetic signal peptide amino acid sequences provides some insight into the sequence features within the signal peptide that influence secretion efficiency.

Speaker
Biography:

Andrew Lobashevsky MD,PhD, D(ABHI)- Diplomat of American Board of Histocompatibility and Immunogenetics is Associate Professor and Histocompatibility Laboratory Director at Department of Medicine of Indiana University and Clarian Health Partners Inc. Dr. Andrew Lobashevsky joined the IU Department of Medicine in December 2004, as the Directory of the Immunology-Histocompatibility Laboratory. Previously, Dr. Lobashevsky was the Co-director of the HLA Laboratory at the University of Alabama at Birmingham Transplant Center, the third largest center in the country. Dr. Lobashevsky received his medical education and postgraduate training at the Department of Immunology and Microbiology of Sechenov’s Medical Academy, Moscow, Russia. He had his post-doctoral training in cellular and molecular immunology at the University of Tennessee at Memphis, and received transplant immunology/histocompatibility training at the University of Alabama at Birmingham. The laboratory headed by Dr. Lobashevsky is focused on providing service for the solid organ transplant programs as well as the bone marrow transplant activities of the Hematology/Oncology division at IU.

Abstract:

In solid organ transplantations the graft outcomes critically depend on the degree of human leukocyte antigen (HLA) matching between the donor and recipient. Although the cellular component of the immune response to the transplanted tissue plays a key role, the contribution of antibodies should not be underestimated. Highly sensitive technologies such as solid-phase based single antigen assay allow to determine even low concentrations of donor specific antibodies and with a high degree of confidence to predict graft outcomes. Development of anti-HLA antibodies strictly depends on immunogenicity of mismatched HLAs. The latter is defined by fi ne epitope structure of HLA. Each HLA protein represents a linear sequence of amino acid residues (AAR) or triplets and the degree of mismatch is assessed as the number of triplets that are not shared between the donor and the recipient. Further investigations of the three dimensional structure of antibody-antigen complexes showed that HLA epitopes could be presented by a group of AARs that are not located beside one another, but rather represent a 3-Å to 5-Å radius patch. These patches have been defined as eplets. Some of eplets include short sequences of AARs, which are equivalent to triplets, whereas, others contain discontinuously located AAR. Further studies demonstrated that area of interaction between complementarity determining region of antibody and HLA is about 900 Å2 and comprises structural and functional epitopes. The former is responsible for binding, whereas the later determine strength of antigen-antibody interaction, which intern results to conformation changes of antibody and subsequent complement activation.

Speaker
Biography:

Itai Benhar is a full Professor, Vice Dean for Research at the George S Wise Faculty of Life Sciences, Tel-Aviv University, Israel. He received in 1992 a PhD in Molecular Biology from the Hebrew University, Israel and was a Post-doc 1992-1995 at NCI, NIH. He joined Tel-Aviv University as a tenure-track assistant professor in 1995 and is a full professor since 2008. He is an expert in the fields of antibody engineering, drug discovery and drug delivery with over the 20 years of being active in these fields, 92 peer reviewed articles and 11 patents.

Abstract:

Small interfering (si) RNAs can be used to silence disease-causing genes. However, their development as drugs has been limited mainly to knocking down liver gene expression, since delivery to other tissues requires development of a targeted delivery carrier. Modulating immune cells function using siRNAs holds great promise in advancing targeted therapies to many immune-related disorders including cancer, inflammation, autoimmunity, and viral infections. However, the ability to effectively knockdown gene expressing in leukocytes is still challenging. Here we present a modular platform to target specific cell types, exemplified here with immune cells, using siRNA loaded lipid nanoparticles (LNP) coated with oriented, targeting antibodies non covalently bound to a membrane-anchored lipoprotein that recognizes their Fc domain. Unlike chemically conjugated antibodies, these oriented antibodies maintain their high affinity and the LNPs avoid scavenging by Fc receptors on macrophages. A simple switch in 5 different targeting antibodies (against Ly6C, CD3, CD4, CD25 and Itgb7) redirected the LNP for exquisitely specific uptake in diverse leukocyte subsets in vivo and enabled specific knockdown in difficult-to-transfect CD4+ cells. Intravenously injected anti-Ly6C-coated LNP encapsulating TNF siRNAs were taken up selectively by Ly6C+ monocytes and activated tissue macrophages, suppressed TNF-alpha expression in the colon and ameliorated inflammatory bowel disease symptoms in a DSS-induced colitis mouse model, demonstrating the platform’s potential therapeutic utility. This approach opens new avenues for studying cell biology in vivo and potentially for a wide range of therapeutic applications in a cell-specific manner.

Anna V Hine

Aston University, UK

Title: ProxiMAX randomization: Precision antibody engineering

Time : 15:25-15:50

Speaker
Biography:

Anna V Hine has studied at the University of Manchester (UK) and Harvard Medical School. She is a Reader and Associate Dean Enterprise at Aston University (UK). In March 2013, she was named BBSRC Commercial Innovator of the Year 2013, for her work in transferring ‘ProxiMAX randomisation’ into SME Isogenica Ltd. A Molecular Biologist by training, she relishes interdisciplinary work.

Abstract:

ProxiMAX’ randomization is the technology that lies behind Isogenica’s Colibra™ offering. It is a defined saturation mutagenesis process that delivers precision control of both identity and relative ratio of amino acids at specified locations within a protein/antibody library. Thus unwanted amino acids such as cysteine and methionine can be eliminated from libraries because no constraints are imposed by the genetic code. Moreover, the process is non-degenerate, which means that encoding DNA libraries are as small as is physically possible. ProxiMAX relies on a process of saturation cycling comprising ligation, amplification and digestion for each cycle and is the science behind the commercial “Colibra™” technology. With achieved diversities of >99% (6 and 11 saturated codons) and the potential to generate libraries of up to 1014 components, we contest that ProxiMAX randomization is a vital tool in engineering antibody libraries of the highest quality. Th is presentation will examine the development of the ProxiMAX process and give examples of antibody libraries created to date.

Ernesto J Fuentes

University of Iowa, USA

Title: Design of PDZ domain specificity

Time : 15:50-16:15

Speaker
Biography:

Ernesto J Fuentes began his Post-graduate education at the University of Dayton (Dayton, Ohio) with a Master's degree in Developmental Biology. He has obtained a PhD degree in Biochemistry from University of Illinois under the mentorship of Dr. A Joshua Wand. He has pursed Post-doctoral training at the University of North Carolina in the areas of protein NMR dynamics with Dr. Andrew Lee and Rho-family GTPase function with Dr. Channing Der. His current research broadly focuses on elucidating the molecular mechanisms that regulate signal transduction. Our recent work has focused on two systems: the regulation of Rho-family GTPase signal transduction and bacterial two component systems.

Abstract:

Molecular recognition is critical for the function and regulation of signal transduction in all cell types. PSD-95/Dlg1/ZO-1 (PDZ) domains are among the most abundant protein-protein interaction modules in the human proteome, commonly found in multi-domain signal scaffolding proteins. PDZ domains typically recognize a variety of short amino acid motifs, including the C-termini and internal peptide sequences of partner proteins. How PDZ domains accommodate these diverse interaction partners while providing specificity remains poorly understood. The overall goal of our studies is to define the molecular basis underlying PDZ domain specificity towards its known ligands. In this presentation, I will discuss recent studies that reveal, how specificity is obtained and rationally altered in model PDZ domains that bind C-terminal and internal peptide sequences? In addition, I will provide examples for how conformational dynamics and structure both contribute to molecular recognition of altered PDZ proteins? These studies have important implications for the evolution, design and regulation of protein-ligand interactions.

Break: B2B, Meet the Exhibitor, Networking & Refreshment Break 16:15-16:45 @ Foyer
Speaker
Biography:

James Endrizzi, founder of SuperNova C, is a Ph.D. chemist and X-ray crystallographer based in Missoula, Montana. He specializes in the use X-ray crystallography combined with biochemical, biophysical, and bioinformatic approaches to unveil structure-based molecular mechanisms for macromolecular function. With a B.S. in Chemistry & Biology from the University of Minnesota and a doctorate from Eugene, Oregon, James went on to a post-doctoral position in the Alber lab at UCBerkeley, where the newly commissioned tunable, synchrotron beam 8.3.1 was available at the Atomic Light Source for many Friday and Saturday nights spent experimenting on various crystals. After 9 years at Berkeley, he went on to research stints at UC Davis (4 years),, Montana State University (3 ½ years) and the Hormel Institute, giving 15 talks, presenting over 20 posters and publishing 23 papers along the way before founding SuperNova C, a company devoted to providing vitamin C as mineral ascorbates.

Abstract:

Aspartate transcarbamoylase (ATCase) has been featured in many biochemistry textbooks as an example of allosteric enzyme regulation. It catalyzes the fi rst step of pyrimidine nucleotide biosynthesis and is feedback inhibited by cytidine triphosphate (CTP), which is the end product of the pathway. ATCase exhibits cooperative substrate binding to its catalytic subunits and allosteric binding of nucleotides to its regulatory subunits. Opposing inhibition by CTP and activation by adenosine triphosphate (ATP) promote homeostasis of purine and pyrimidine nucleotides. Regulation of ATCase activity has been interpreted as a ligand promoted change in the equilibrium between inactive (taut) and active (relaxed) states largely consistent with a two-state allosteric model. The structure of the ligand-free enzyme represents the inactive, taut state and the structure of ATCase bound to a bisubstrate analog (N-phosponacetyl-L-aspartate; PALA) has been proposed to represent the active, relaxed state. However, there is little evidence that the PALA-bound enzyme is structurally similar to the active conformation(s) in the absence of substrates. A novel approach to define the structure of the ligand free relaxed, or “activated” state is to use mutant enzymes that destabilize the taut state and shift the allosteric equilibrium toward the relaxed-state conformation(s). In contrast to the wild-type enzyme, which exhibits a more compact global conformation and sigmoidal enzyme kinetics, the mutants are more expanded and display hyperbolic kinetics characteristic of non-cooperative enzymes. Thus, we tested the hypothesis that ATCase is activated through the modulation of flexibility by determining the X-ray crystal structures of several activated-state mutants. We obtained crystals of three ATCase mutants in multiple crystal forms, collected X-ray diffraction data on the various forms and solved and refined dozens thus far. A variety of tertiary and quaternary structures in activated-state mutant enzymes support the hypothesis that these structures represent metastable states accessed by an ensemble of activated-state conformations..

Seth McDonald

Illumina, Inc., USA

Title: Improved illumina sequencing by polymerase engineering

Time : 17:10-17:35

Biography:

Seth McDonald received his PhD in Biochemistry from the University of Utah where he studied the structure and biological implications of the interaction of the essential Spn1 and Spt6 transcription factors in Saccharomyces cerevisiae. In his Post-doc at Colorado State University, continuing his use of x-ray crystallography and enzyme kinetics, he engineered fast and accurate RNA polymerases from small RNA viruses that effectively restrict viral genome variations within the given population required for infection and survival. At Illumina, he has made significant contributions to the structural and functional understanding of how polymerases incorporate nucleotides modified on the base and sugar. He is the assay development and structural biology lead in the protein engineering group where he continues to contribute to the ongoing enzyme engineering projects within Illumina.

Abstract:

The sequencing-by-synthesis (SBS) chemistry commercialized by Illumina has been a key enabler of massively parallel nextgeneration sequencing, which in turn has dramatically reduced the cost of sequencing human genomes and resulted in personalized medicine initiatives and population sequencing efforts throughout much of the world. The capacity of engineered archaeal family B DNA polymerases (pols) to incorporate modifi ed nucleotides has been exploited by researchers throughout academia and biotechnology, including Illumina’s SBS chemistry. Nucleotides with a 3’-reversible terminator and fluorophores attached via the base serve as the basis for base calling in Illumina NGS platforms. Pols deployed on the fi rst Genome Analyzer™ instruments had poor incorporation effi ciency of these modifi ed nucleotides. Through our engineering eff orts, we have facilitated a significant reduction in chemistry time while improving sequencing data quality and enabling over 300 bp read lengths. The critical goal of our research is to engineer pols with enhanced selectivity, fidelity, and incorporation efficiency of modified nucleotides to enable faster sequencing turnaround time and boost adoption of NGS technologies in the clinic.

Xiaoying Zhang

Northwest Agriculture and Forestry University, China

Title: Immunological detection of antibiotic residues using antibody engineering

Time : 17:35-18:00

Speaker
Biography:

Xiaoying Zhang is Professor at Northwest Agricultural and Forestry University (elite university in the national 985 and 211 programs), China since 2008. He is graduated from China Pharmaceutical University (undergraduate study in Pharmacology) and Charité Medical School (Post-graduate and PhD in Pharmacology) of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Germany. Before and during his PhD study, he has worked in WHO- headquarters Geneva and Robert- Koch Institute Berlin. He was the nominee for the outstanding Young Scientist Award, China (2015). His major research interests are: antibody engineering, immunoassay and rapid detection/diagnosis, nature product research and pharmacological study. He has contributed 100+ publications (from which 80+ international publications). He is holding three Chinese patents for his innovative research accomplishments. He has also edited fi ve books (four– Chinese and one in English) and contributed three book chapters in the books of International publishers. He has been appointed as editor and/or reviewer in more than 40 international journals and serves as Member of Board of Examiner in universities from China, Germany, India and Pakistan.

Abstract:

Residues of antibiotics in the aquatic environment and in food of animal origin represent a major concern, as prolonged exposure to antibiotics is a serious health hazard, related to both the side effects of prolonged use and the risk of developing bacterial resistance to various antibiotics. Given the low levels of the antibiotics residues in complex matrices, the development of sensitive analytical methods represents a major challenge. We report the current state of the art in the determination of antibiotics based antibody engineering and immunoassays. First, diversified antibody generation platforms/methods promote the screening and assessment of high qualified antibodies. Second, various immunoassays simplify and empower the immune-detection applications for different biomedical purposes. Third, there are tremendous academic motivations and industrial needs in the detection/evaluation of antibiotics by using antibodies and immunoassays. There are diversified antibody generation platforms/methods (e.g., Polyclonal antibody (pAb), Cell engineering antibodies—hybridoma technology, recombinant antibdy (rAb)). Different species of antibodies e.g. chicken antibody (polyclonal IgY, monoclonal IgY), camel antibodies, and rabbit antibodies have been widely used. The recent advancements in antibody engineering permit to produce chicken mIgY quickly and inexpensively by phage display technology, because chicken possess only one functional immunoglobulin heavy chain variable region (VH) gene and one light chain variable region (VL) gene. The purpose of our study is to prove that the mIgY could be used for immunological detection and diagnosis, for screening and validating of biomarkers and drug targets. There is a huge demand for the detection reagent of antibiotic residues on the market, and it gradually becomes the intrinsic driving force of the development of industry. Diversified antibody generation platforms/methods promote the screening and assessment of high qualified antibodies. Various immunoassays simplify and empower the immune-detection applications for different biomedical purposes.

Speaker
Biography:

Amirali Kia has completed his Bachelor’s and Master’s working on the application of various artifi cial intelligence algorithms in computational fl uid mechanics. He then started his PhD at Stanford University where his focus was on high performance computing and developing fast algorithms with applications in computational biology. He started his career at Illumina by joining Protein Engineering group to apply machine learning algorithms to enzyme engineering. He is now a senior manager at Illumina in the Computational and Applied Biology Department, leading Deep Learning group. The focus of his team is to bring new AI algorithms to genomics and Illumina’s internal technology.

Abstract:

Statement of the Problem: Next generation sequencing has opened new doors into genomics. Recent advances in sequencing technology have enabled researchers to sequence genomes with unprecedented accuracy and speed. However, preparing DNA sequencing libraries still remains as a challenging step. Transposase-based library preparation off ers a simple, fast, scalable, and flexible solution. However, when compared to other (ligation-based) library preparation methods, transposases show some insertion bias that aff ects the fi nal DNA sequencing library.

Methodology: We applied various machine learning algorithms to random mutagenesis libraries of transposases to efficiently design new mutant transposases with less insertion bias.

Findings: We present the discovery of a mutant transposase (Tn5-059) with low GC insertion bias and AT dropout. Libraries prepared by this mutant have good uniformity of genome coverage as well. Tn5-059 also shows low sensitivity to the amount of input DNA. In addition, this enzyme shows effi cient performance in DNA sequencing libraries for open chromatin profiling.

Conclusion & Signifi cance: DNA input tolerance together with superior coverage uniformity are two important factors to be considered in DNA sequencing library preparation. Tn5-059 successfully delivers on both of these aspects, which leads to less sequencing volume and lower sequencing cost. We discuss the importance of choosing a correct assay as well as the importance of filtering data based on the biology behind the assay.

  • Young Researchers Forum

Session Introduction

Steven Alexander Mann

Indiana University, USA

Title: PD-L1 assays: An update on FDA-approvals and uses with companion immunotherapies

Time : 18:25-18:45

Speaker
Biography:

Steven Alexander Mann received his Medical degree in 2015 from the University of Alabama, School of Medicine and is currently a Pathology Resident at the Indiana University School of Medicine. He is actively involved in managing molecular requests for his current institution and is a Member of the College of American Pathologists Engaged Leaders Network. He has multiple peer-reviewed publications on a variety of topics including a recent review of PD-L1 immunohistochemistry for genitourinary tumors and a chapter on biomarkers for hepatic and pancreatobiliary malignancies

Abstract:

Novel immunotherapies have recently demonstrated significant objective responses in many advanced malignancies. The standard therapy of some of these cancers had remained unchanged for decades despite poor outcomes. Immune checkpoint inhibitors, in particular those for the programmed cell death protein (PD-1) pathway, have bought great progress for the care of these patients in the past decade. Currently, the most clinically significant ligand of PD-1 is the programmed cell death ligand-1 (PD-L1). When this pair binds, cytotoxic T cell function is inhibited. Many types of tumors can express PD-L1 as a mechanism to evade the antitumor response of the immune system. By the end of 2016, the United States Food and Drug Administration (FDA) had approved four different immune checkpoint inhibitors for use in a total of six different types of malignancies. Many of these drug-indication combinations have an FDA-approved companion PD-L1 immunohistochemistry assay available. These PD-L1 assays are used to predict therapeutic responses and in some cases tumor PD-L1 positivity is a prerequisite for initiating therapy. Each of these assays uses a different anti-PD-L1 clone. The use of these antibodies and the interpretation of tumor PD-L1 expression are continually evolving, as well as the understanding of the overall impact of this information. Knowledge of the current state of these rapidly evolving drugs and assays will prepare researchers, manufacturers and clinicians for future applications of these promising antibodies.

Speaker
Biography:

Estefania Arevalo-Tristancho is currently pursuing Master’s degree in Design and Process Management with emphasis on Bioprocesses.

Abstract:

Enzymes of microbial origin have been proven to be useful in different fields such as medicine and the food industry. L-asparaginase, of microbial origin is an amidohydrolase enzyme, which catalyzes the conversion of L-asparagine to aspartate and ammonium cation. L-asparaginase is known as an anti-cancer agent, which prevents the proliferation of tumor cells by decreasing the level of asparagine in the blood. Th is enzyme has been shown to be a form of treatment for acute lymphocytic leukemia (ALL), extracted from E. coli and Erwinia chrysanthemi, which have a high commercial value and multiple side effects due to L-glutaminase activity produced by them. In this research, 25 Streptomyces isolated from the Arauca River bank (Equatorial zone, Colombia) with L-asparaginase activity were found by the Nessler method. Studies have shown that Streptomyces, in addition of being a source of easy access and production, produce L-asparaginase with less or none L-glutaminase activity. Streptomyces isolated were identified morphologically and molecularly. Plackett-Burman design established that at 30 ºC, 200 rpm, pH 7, lactose (1%) and malt extract/asparagine (0.15%) were the best conditions for the fermentation of the isolates and enzyme production. This enzyme secreted in the medium will be purifyed by dialysis, lyophilization and ion exchange chromatography to establish a mathematical model to simulate the effect of the substrate, pH and temperature on L-asparaginase activity.

Speaker
Biography:

Estefania Arevalo-Tristancho is currently pursuing Master’s degree in Design and Process Management with emphasis on Bioprocesses.

Abstract:

Enzymes of microbial origin have been proven to be useful in different fields such as medicine and the food industry. L-asparaginase, of microbial origin is an amidohydrolase enzyme, which catalyzes the conversion of L-asparagine to aspartate and ammonium cation. L-asparaginase is known as an anti-cancer agent, which prevents the proliferation of tumor cells by decreasing the level of asparagine in the blood. Th is enzyme has been shown to be a form of treatment for acute lymphocytic leukemia (ALL), extracted from E. coli and Erwinia chrysanthemi, which have a high commercial value and multiple side effects due to L-glutaminase activity produced by them. In this research, 25 Streptomyces isolated from the Arauca River bank (Equatorial zone, Colombia) with L-asparaginase activity were found by the Nessler method. Studies have shown that Streptomyces, in addition of being a source of easy access and production, produce L-asparaginase with less or none L-glutaminase activity. Streptomyces isolated were identified morphologically and molecularly. Plackett-Burman design established that at 30 ºC, 200 rpm, pH 7, lactose (1%) and malt extract/asparagine (0.15%) were the best conditions for the fermentation of the isolates and enzyme production. This enzyme secreted in the medium will be purifyed by dialysis, lyophilization and ion exchange chromatography to establish a mathematical model to simulate the effect of the substrate, pH and temperature on L-asparaginase activity.