home   genetic news   bioinformatics   biotechnology   literature   journals   ethics   positions   events   sitemap
 
  HUM-MOLGEN -> Events -> Meetings and Conferences  
 

Protein Therapeutics Discovery & Development

 
  August 05, 2008  
     
 
GTCbio, San Diego, CA
September 8-9, 2008


Day 1 - Monday, Sept 8, 2008

   

7:00

Registration & Breakfast

   

7:55

Chairman's Opening Remarks

   

  8:00

[KEYNOTE PRESENTATION]

 

Challenges and Opportunities for Peptide Biologics

 

Michael R. Hanley, Ph.D., Chief Scientific Officer, Amylin Pharmaceuticals, Inc

   
   

Session I - Therapeutic Drug Design

   

 8:45

Identifying Novel Receptor-Ligand Interactions using SPR, BLI and Protein Microarrays

 

Lino Gonzalez, Ph.D., Scientist, Protein Chemistry, Genentech, Inc.

   
 

Now that the human genome is complete, one of the next steps is to understand the interactions between all transmembrane and secreted proteins, which represent about one-third of all human genes. This knowledge would greatly aid in characterizing the biological function of many receptors/ligand pairs and their potential as therapeutic targets. We concentrate on receptors of the immune system, in particular the immunoglobulin receptor family. Surprisingly, a significant number of these receptors still remain orphans. In order to identify candidate partners, we have utilized Genentech's SPDI (Secreted Protein Discovery Initiative) protein library, consisting of over 1,000 purified, secreted proteins. We have used surface plasmon resonance (SPR) and Bio-layer interferometry (BLI) technology to screen hundreds of interactions per day. To date, this work has led to the identification of several novel interactions that are now being evaluated for their therapeutic potential. The first was an interaction between the B and T Lymphocyte Attenuator (BTLA) and the tumor necrosis factor receptor Herpes Virus Entry Mediator (HVEM). We have recently increased our screening capacity to the thousands of interactions per day by generating protein microarrays from the SPDI library. We are using these protein arrays to identify novel receptor interactions with the hope that they may become future therapeutic targets for fighting disease.
Attendees will benefit from:
* Highlight technologies and methodologies for screening extracellular protein interactions
* Development of secreted protein microarrays and analysis of potential hits
* Methods for validating extracellular protein interactions
* Examples of functionally validated hits will be presented

   

9:15

Coengagement of B cell Receptor and FcgRIIb Inhibitory Receptor via an Fc-Engineered Antibody: A New Strategy to Suppress B Cell Function in Autoimmune Diseases

 

David Szymkowski, Ph.D., Senior Director, Biotherapeutics, Xencor

   
 

B cell activation can be inhibited by coengagement of B cell receptor (BCR) and inhibitory receptor FcγRIIb, which occurs physiologically via binding of immune complexes of IgGs and their cognate antigens. To mimic the inhibitory effect of immune complexes, we engineered a novel therapeutic antibody that coengages BCR and FcγRIIb with high affinity. The Fc domain of a humanized anti-CD19 antibody was engineered to have over 400-fold increased binding affinity for FcγRIIb, with decreased binding to activating Fcγ receptors. Such "IIb enhanced" (IIbE) variants stimulated the SHIP-mediated FcγRIIb signaling cascade, and strongly inhibited BCR-induced calcium mobilization and ex vivo proliferation of activated human primary B cells. Preliminary studies also suggest that XmAb™5871, our clinical candidate IIbE antibody, blocks B cell-mediated immune responses in huPBL-SCID mouse models. The new therapeutic strategy described here inhibits B cell function by mimicking the physiological negative feedback mechanism induced by immune complex coengaging FcγRIIb and BCR. XmAb™5871 represents an alternative therapeutic antibody to the clinically validated B cell depletion strategy, and raises the possibility of suppressing autoimmunity without compromising long-term host immunity.
Benefits:
1) The talk will introduce a novel therapeutic approach that is grounded in well-established biology (e.g., the phenomenon of B cell negative regulation has been accepted for over one hundred years).
2) The talk will describe the cutting-edge antibody Fc engineering which was essential to success. (Antibodies with native Fc domains, including other therapeutic antibodies, do not stimulate this regulatory pathway).
3) The B cell is now a clinically validated target in oncology and in autoimmune disease, yet "IIbE" represents a fresh approach with a completely different mechanism than conventional B cell depleting therapeutics.
4) This talk will present a case study of early R&D through to candidate selection of XmAb™5871, a humanized and high-affinity (Fv and Fc) antibody with a novel mechanism of action.

   

9:45

Recombinant Protein Design for the Generation of Particle-Bound and Targeted Therapeutics Using Natural and Synthetic Vesicles as Delivery Vehicles

 

Alain Delcayre, Ph.D., Director of Research, BN ImmunoTherapeutics

   
 

The pharmacodynamic properties of therapeutics are critical parameters that contribute to drug efficacy. They have significant bearing on the determination of treatment regimen and doses and consequently also affect benefit/safety ratio as well as cost of treatment. Drug features leading to improved efficacy include particle-bound formulation, which can increase the stability /half life of compounds, and targeted delivery. In addition to improving efficacy, these features may contribute to achieve clinical benefits using lower drug doses, thereby potentially limiting drug-related side effects. We have developed a technology called Exosome Display that enable the manipulation of the protein content of naturally occurring exosomal vesicles as well as the efficient loading of synthetic vesicles with therapeutic protein and/or ligand for targeted delivery of therapeutic compounds. The features and advantages of this technology include the following:
• Vesicles bearing chimeric proteins containing the C1C2 domain of Lactadherin are generated with proteins presented on the vesicle surface
• The C1C2 domain-fusion partners may be any protein such as antigens, cytokine, co-stimulatory molecules, receptor ligand…
• When expressed in non-mammary cells, the C1C2 domain of Lactadherin targets chimeric protein expression to exosomal vesicles released in the extracellular milieu
• Purified C1C2-chimeric proteins exhibit high binding capacity to synthetic lipid surfaces of liposomes
• Particle-bound multimeric forms of protein therapeutic may be co-localized on the same vesicles with enhancing factors and/or ligand for targeted delivery
• Full-length membrane drug targets such as GPCR can be displayed on recombinant exosomes for use in drug screening.
Exosome display yields novel compounds with attributes and properties that can be used in a broad field of technology and medical applications. It notably leads to a novel strategy for vaccine design which will be described.

   

10:15

Networking and Refreshment Break

   

10:45

Phylomers: Synthetic Peptides Derived from Biodiverse Gene Fragment Libraries, for Blocking Targets Inside and Outside Cells

 

Karen Kroeger, Ph.D., Senior Research Scientist, Phylogica Ltd

   
 

Phylomers are a new class of peptide derived from genomic fragments of biodiverse archael and bacterial species. These peptides can have high affinities and high specificity for targets, even before affinity maturation or optimization. Phylomer peptides can also exhibit encouraging functional hit-rates, when compared to randomly derived peptides, possibly due to an evolutionary selection of natural sequences for structure and stability. Phylomer libraries have been made in yeast two hybrid, phage display and mammalian expression vectors. Peptides have been identified from such Phylomer libraries, with diverse activities, ranging from antimicrobial to mimetics of discontinuous allergen epitopes. Synthesised Phylomers directed against intracellular targets have been shown to function in animal models of ischemia and wound healing of severe burns, when fused to a protein transduction domain. Data showing Phylomer peptide blockade of an intracellular signalling adaptor in a Toll Like Receptor pathway will be presented. A series of Phylomers targeting CD40 Ligand (CD40L) will also be described, which are able to block CD40L mediated biological functions. Results addressing the challenges of immunogenicity and pharmacokinetics in the development of peptide therapeutics will be shown.
Attendee will benefit from:
1. Overcoming the hit-rate problem in screening peptide libraries for functional inhibitors
2. Delivering functional peptides into cells in vivo using protein transduction
3. Extending half life of peptides in human serum using retroinverso synthesis
4. Finding peptide sequences which are unencumbered by patents around targets or antibodies
5. Accessing a diverse array of target classes and target epitopes

   

11:15

Maximizing Protein Yield in Heterologous Expression Systems

 

Mark Welch, Ph.D., Senior Scientist, DNA 2.0, Inc.

   
 

Maximizing protein yield in heterologous expression systems is critical for efficient production of high value protein products and can be essential for generating sufficient material for structure based drug design and functional studies. While much work has been done in expression vector and host strain development, surprisingly little has until now been accomplished in the area of gene sequence optimization. In an NSF funded project, DNA2.0 has used its gene synthesis capacity to created large systematically varied gene sets for an antibody fragment (in collaboration with OncoMed Pharm. Inc.) and a DNA polymerase. In both datasets, the amino acid sequence is kept constant and only silent mutations are tested. Protein expression levels changes for up to two orders of magnitude are seen in E. coli and the variables responsible for the changes are identified. Our data show that codon bias is a strong determinant of expression level. However, the codons preferred for increased expression are distinctly different from the most common codons. Together with Prof. Bob Stroud’s UCSF laboratory, we have also synthesized and expressed a systematically varied large set of a clinically relevant protein structure target. The correlation between codon bias and protein expression in yeast is discussed.

   

11:45

Effect of Carbohydrates on Biological Properties of Protein Therapeutics

 

Rachael Brake, Senior Research Scientist, Amgen

   
 

Therapeutic proteins have revolutionized the treatment of many diseases, but low activity or rapid clearance can often limit their utility. New approaches have been taken to design drugs with enhanced in vivo activity and half-life to reduce injection frequency, increase convenience, and improve patient compliance. One recently used approach is that of glycolengineering, whereby the protein-associated carbohydrates on a recombinant protein are altered to introduce N-linked glycosylation motifs. This technology has been applied to a recombinant form of human erythropoietin and has successfully validated the approach of glycoengineering rHuEPO (darbopoetin alfa). This and other examples of glycoengineered proteins will be discussed.

Benefits to attendees:
1. Understand carbohydrate modification
2. Facilitate an understanding of Phamacokinetic properties of therapeutic proteins
3. Observe the impact of (darbopoetin alfa) Aranesp to patient care
4. Determine the applicability of glycoengineering to other proteins
5. Determine a research strategy to investigate glycoenginering

   

12:15

Interfering with RAS-Effector Protein Interactions Inside Cells

 

Terry H. Rabbitts, Ph.D., Leeds Institute of Molecular Medicine, St. James's University Hospital

   
 

RAS family proteins have pivotal roles as molecular switches to transmit a variety of extracellular signals to the nucleus controlling cell growth, differentiation, migration and survival via the interaction with a range of effectors molecules. RAS mutations, impairing the GTPase activity and consequently causing accumulation of active GTP-bound RAS, are found in up to 30% of human cancers. Growing insight in RAS signal transduction reveals that more than ten proteins with distinct functions (including PI3K, RAF and RALGEF) have been identified as RAS effectors, directly interacting with RAS to constitute different downstream signalling pathways. The mutation of the RAS family members are examples of mutations that can be classified as genotype-specific targets for therapy. Despite many years of research on RAS mutations and their effects in cancer, attempts to develop drugs directed to RAS have proven ineffective, and the crucial protein interactions between RAS and effectors like PI3K remain elusive targets because it is generally assumed that protein-protein interactions will not be “druggable” as the interaction surfaces are large and usually featureless. We have developed methods to target antibody fragments as first generation leads to block protein-protein interactions in cells and used RAS-effector binding as a model system. Our technique called intracellular antibody capture (IAC) has been used select single variable domain antibody fragments (anti-RAS VH) that specifically bind to activated GTP-bound RAS and prevent RAS-dependent tumourigenesis in pre-clinical models. The crystal structure a RAS-VH complex reveals that the anti-RAS VH binds to the conformationally variant switch regions of RAS, where its effectors including PI3K, RAF and RALGDS also bind. This anti-RAS interfering antibody inhibits lung tumourigenesis in RAS-dependent pre-clinical models. Thus anti-RAS VH has potential use in biotherapy as a macromolecular drug (macrodrug) since as it functions by a different mechanism from anti-RAS reagents currently in clinical trials. Our findings have implications for use of interfering single domain antibody fragments to drug the “undruggable” protein-protein interactions in cancers. Furthermore, detailed structural and mutational studies of the RAS-VH interface could lead to the prediction of chemical shapes that may facilitate the synthesis of small molecules with parallel properties to the interfering macrodrugs.

   

12:45

Lunch on your own

Session II - Novel Developments in Protein Based Therapeutics

   

2:15

Oral Presentations from Submitted Abstracts

 

Submit your abstract by August 8 to be considered.

   

2:45

High Throughput Protein Production and Screening in vitro and in vivo

 

Steve Doberstein, Ph.D., Vice President, Research , FivePrime Therapeutics, LLC

   
 

We have built a high-throughput platform for genome-wide screening, discovery and early preclinical development of protein therapeutics for oncology. We combine advances in full length cDNA cloning technology, proprietary high-throughput protein expression technologies, advanced automated cell-based screening, and rapid in vivo testing of leads to discover proteins with potential for clinical utility. The FivePrime process has revealed both completely novel proteins with medically relevant pharmacology and uncovered new utility for previously described proteins. We have recently expanded the use of our unique protein library to comprehensively match orphan ligands with receptors, and to determine the best tumor-host interactions to inhibit in cancer models in vivo. This platform has yielded multiple lead compounds in oncology, the first of which will enter the clinic in 2008.

   

3:15

Development of Novel Therapeutic Antibodies for the Treatment of Sepsis

 

Xiang Yang Tan, Principal Scientist, Lab Head, Wyeth Pharmaceuticals

   
 

Sepsis is an inflammatory response to an infection or stimulus that becomes amplified and dysregulated. Despite recent market entries and continually improving hospital care, sepsis remains a significant unmet medical need. More than 2 million cases of sepsis occur each year in the U.S., Europe and Japan, with estimated annual costs of $17 billion and mortality rates ranging from 20-50%. Safe and well-tolerated therapeutic agents that could reduce the progression from early sepsis to severe sepsis or septic shock, and thereby improve survival, could provide a break-through in sepsis therapy.
The receptor for advanced glycation end products (RAGE) is a cell-surface member of the immunoglobulin super-family, which binds with multiple ligands, such as HMGB1, A-beta and S100. RAGE was originally identified by its ability to bind advanced glycation end-products (AGEs) that are produced by glycoxidation of proteins. RAGE and its ligands are elevated in a number of disease settings such as sepsis, atherosclerosis and vascular complications secondary to diabetes. Data generated at Wyeth indicates that RAGE may play an important role in the pathogenesis of sepsis. Homozygous RAGE null mice (RAGE-/-) showed a significant degree of protection from the lethal effects of cecal ligation and puncture (CLP), a murine model of sepsis, when compared to parental, wild type mice. In addition, an anti-RAGE antibody designated XT-M4 was generated in Wyeth. XT-M4 recognized RAGE with high affinity and blocked RAGE and ligands interactions. XT-M4 showed efficacy in the CLP model when delivered at the time of surgery or delayed to 24 hours post-CLP. In a second model, inhibition of RAGE or deletion of RAGE did not disrupt the host mechanism or clearance of microbial pathogens. These studies demonstrated that RAGE is involved in the septic response and an anti-RAGE antibody can improve survival in polymicrobial sepsis in mice.
XT-M4 was fully humanized to decrease any potential immunogenicity and manufacturability issues. The humanized XT-M4 retained full binding activity and was administered in a prophylactic study in mouse CLP. A single intravenous dose protected about 90% of mice seven days post-CLP.
Highlights of the presentation:
- Sepsis remains a significant unmet medical need with mortality rates ranging from 20-50%.
- The receptor for advanced glycation end-products (RAGE) plays an important role in the pathogenesis of sepsis.
- RAGE antibody XT-M4 showed significant protection of the mice from the lethal effects of a murine model of sepsis, cecal ligation and puncture (CLP).
- XT-M4 was fully humanized to decrease any potential immunogenicity and manufacturability issues.

   

3:45

Networking and Refreshment Break

   

4:15

A Clinically Validated Method for Generating Best-in-Class Antibody Therapeutics

 

Mark R. Alfenito, Ph.D., Head of Technology Licensing, KaloBios Pharmaceuticals, Inc.

   
 

Therapeutic proteins, --even “fully human” antibodies— show some level of immunogenicity. As early as possible in the development of the drug candidate, the goal is therefore to minimize the costly risk of seeing immunogenicity in the clinic. We believe that human germline and near-germline antibody segments can provide repertoires with which to build such low immunogenicity antibodies. Humaneering™ is a new method of engineering antibodies with these high value qualities, as well as with other important and frequently overlooked characteristics (e.g., germline, or near-germline, high affinity, good expression level, good down stream processing characteristic, etc.). Humaneering™ is not based on other proprietary methods. Two Humaneered™ antibodies have now completed Phase I clinical trials. Recent data will be shown on methods and results.

   

4:45

Amyloid as a New Challenge in Protein Based Therapeutics

 

Ludmilla Morazova-Roche, Department of Medical Biochemistry and Biophysics, Umeå University, Sweden

   
 

Significant advances in therapeutic applications of proteins and peptides have brought new challenges in the field of drug development, among which an ordered protein aggregation or amyloid formation plays an important part. In contrast to amorphous aggregation, the amyloid self-association of polypeptides results in the structurally organized protein polymers known as amyloid fibrils. Due to extensive research in protein folding and amyloid diseases amyloid self-assembly has recently emerged as a universal phenomenon. The amyloid represents a new generic structure characterized by cross-β-sheet formation in its core, which implies that any polypeptide can adopt this conformation under amyloid-prone conditions. Some widely-used biopharmaceuticals such as insulin, glucagon, amylin and calcitonin have been shown to form amyloids and this list may be significantly extended upon further research. Compared to soluble precursor proteins and amorphous aggregates amyloids gain new properties such as remarkable stability and protease resistance, polymorphism, self-propagation via seeding and cross-seeding, cytotoxicity and induced immunogenicity. Some of them can be hazardous in biopharmaceutical applications. The causes of amyloid aggregation and strategies for its prevention which utilize the current knowledge of amyloid properties, structure-based design principles and protein chemistry are discussed using insulin, lysozyme, de novo protein albebetin and Ab peptide as model systems. Once these challenges are met and amyloid formation is avoided or reversed, these will lead to safer and surer pharmaceuticals.

   

5:15

Second Generation Affibody-Molecules and Novel Albumin-Binding Technology

 

Maria Åkerman, Ph.D., Project Manager, Affibody

   

5:45

Poster Session & Networking Reception

   

Day 2 - Tuesday, Sept 9, 2008

 

Top of the page

7:00

Continental Breakfast

   

7:55

Chairman's Review of Day One

   

Session III - Discovery to Production: Protein Formulation and Process Development

   

8:00

cIEF Analysis of Monoclonal Antibodies with High-Resolution and Reproducibility

 

Ingrid D. Cruzado-Park, Senior Development Scientist, Beckman Coulter, Inc

   
 

During development and manufacture of therapeutic monoclonal antibodies (MAbs), characterization of protein glycosylation is critical. Glycoforms of purified MAbs are usually separated by differences in isoelectric point (pI). Capillary Isoelectric Focusing (cIEF) is a separation technique that has been successful in generating glycosylation profiles of MAbs. Faster separations can be achieved with cIEF than with IEF-gel electrophoresis. When compared with Imaging IEF systems, cIEF provides higher resolution due to the use of longer capillaries. However, cIEF is an inherently complex technique and understanding it is essential to achieving success. For example, the use of a protein solubilizer is critical in preventing MAbs aggregation and precipitation during focusing. In addition, the pI measurement is more accurate and precise when using synthetic peptides as pI markers rather than using proteins. The use of stabilizers can reduce distortions in the pH gradient and improve resolution. Optimization of the ampholyte volume, focusing time and amount of stabilizers is essential in achieving the resolution required. This presentation will describe how to achieve maximum resolution and reproducibility in cIEF. In addition, it will provide guidelines for the development of cIEF methods.
Attendees will:
• Learn how to characterize therapeutic antibodies by cIEF.
• Find out the key steps in cIEF method development.
• Realize that high resolution and reproducibility can indeed be achieved in cIEF.

   

8:30

High Concentration Subcutaneous Monoclonal Antibody Formulations

 

Larry Brown, Sc.D. Chief Technology Officer, VP Research, Baxter Healthcare

   
 

Therapeutic monoclonal antibodies (mAbs) are one of the fastest growing segments of the pharmaceutical market. A therapeutic challenge for the mAb market is that most of these drugs are delivered intravenously due to high dosing requirements often in the several hundred-milligram range. A critical medical need in the formulation of this drug class is finding an alternative to the IV route of administration. We have incorporated monoclonal and polyclonal antibodies into our PROMAXX microsphere formulation technology. Microspheres were formulated by mixing monoclonal anti-factor VIII and anti-CD34 antibodies, with water-soluble polymer solutions, such as PEG or Poloxamer. The solution was cooled below ambient room temperature for microsphere formation. The microsphere formation process showed no impact on antibody integrity, as demonstrated by fluorescence spectroscopy and by size exclusion chromatography. Light scattering measurements of particle size showed monodispersed mAb microspheres with a diameter of 1.92 microns. X-ray powder diffraction showed that the protein in the microsphere was amorphous. Microsphere suspensions at concentrations up to 300 mg/mL, were shown to be injectable through 26 gauge needle sizes in vitro and in vivo. Studies of mAb microspheres and solutions delivered by the subcutaneous route showed similar pharmacokinetics.
• Novel monoclonal antibody microspheres suspensions formulated.
• Stability of the monoclonal antibodies demonstrated.
• Injectability through small bore 26 G needles demonstrated.
• Suspension and solution pharmacokinetics were similar.
• Technology aimed at transforming delivery from intravenous to the subcutaneous administration route.

   

9:00

Enabling Efficient Upstream Commercial Development of an Escherichia Coli-Based Process Leveraged by the Guidance of Manufacturability Limits

 

Henry Lin, Ph.D., Scientist, Process Development, Amgen

   
 

As numerous microbially-derived products mature in the pipeline, reducing timeline and cost are critical to successful commercialization. Manufacturing aspects beyond the clinical setting need be considered, which often fall outside the scope of a platform process. Relying on process improvement to unilaterally meet launch demand creates strain on development and heightens risks. Commercial development strategies that take into account quality, manufacturing fit, facility capacity, and process robustness early on help reduce cost, maintain focus and streamline development. In addition, these strategies mitigate the risks for meeting commercial demand. Such philosophy coupled with design of experiments for optimization was applied to the commercial development of a fermentation process that produced therapeutic proteins as inclusion bodies in E. coli. Results of a successful development that not only considers optimizing production, but also the commercial manufacturability will be discussed.
1. Presents a commercial philosophy that facilitates development, and saves time and cost to market
2. Commercialization and manufacturability considerations to know beforehand with an eye on market launch
3. How to guide sound process development with facility fit in mind from start to finish?
4. How to plan optimization experiments by leveraging clinical or platform process data
5. How to meet market demand by balancing solution between scientific innovation and the use of facility capacity?

   

9:30

From Laboratory to Phase I/II Cancer Trials with Recombinant Biotherapeutics

 

Berend Tolner, Ph.D., Senior Post-doctoral Scientist, UCL Cancer Institute, University College London

   
 

Bioprocess development and GMP (Good Manufacturing Practice) production is a key component for advancing biologics toward the clinic1. Academic Institutions often have the essential know how for production of their successful research biologicals and can benefit from developing their own ‘Artisan’ GMP production to facilitate Phase I/II clinical trials with these products. We describe how such a production facility can be created and illustrate the pathway from laboratory to phase I/II cancer trials with a recombinant antibody-based bio-therapeutic produced in the Pichia pastoris fermentation system. The case study product has been used safely in patients. The procedure has generic application as it has been successfully used for production of scFvs, diabodies, and recombinant antigen.

   

10:00

Analysis and Characterization of PEGylated Recombinant Therapeutic Proteins

 

Jifeng Zhang, Ph.D., Principle Scientist, Amgen, Inc.

   

10:30

Networking and Refreshment Break

   

Session IV - Overcoming Challenges in Developing Protein Based Drugs

   

11:00

Safety First: Standardizing ex vivo Immunotoxicity and Immunogenicity Testing

 

Dominic Eisinger, Ph.D., Director, Strategic Development, RulesBasedMedicine

   
 

Protein therapeutics presents many unique advantages and challenges. One particular challenge of protein therapeutics is the potential for immunotoxicity as exemplified by the 2006 TeGenero incident. The ex vivo cell culture of isolated peripheral blood mononuclear cells (PBMCs) has been the standard means for monitoring the body’s immune system. Unfortunately, the isolation and analysis of PBMCs is prone to variability and requires skilled personnel in addition to a specialized cell culture laboratory. Moreover, PBMCs are an artificial system that loses key immune signaling elements emanating from other cell types such as granulocytes and platelets.
TruCulture™ is the first fully closed whole blood culture system that allows one to avoid the problems inherent in PBMCs or other blood culture methods especially in the clinical trial setting. Data will be presented demonstrating the utility of this simple ex vivo system when combined with a quantitative multiplex read-out of key immune signaling mediators.
Benefits:
• Introduces a new analytical technology of immuntoxicity and immunogenicity monitoring
• Highlights the use of multiplex Biomarker monitoring from discovery through development and into clinical trails.
• Highlights innovation for improving drug development and saftey

   

11:30

BioMarkers for Immunogenicity using HLA, EpiMatrix and iTEM -That’s Easy!

 

Annie De Groot, CEO/CSO, Epivax, Inc.

   
 

Immunogenicity elicited by protein therapeutics can cause serious side effects in humans. We affirm that immunogenicity can be predicted and patients who are at increased risk of developing adverse immune responses can be identified using HLA typing and existing bioinformatics tools.
We have used HLA typing in conjunction with EpiMatrix, an in-silico epitope-mapping tool to predetermine the immunogenicity of biological therapeutics. In a recent published case, we identified promiscuous epitopes (“EpiBars”) in the C-terminal region of a recombinant fusion protein (FPX) consisting of two identical, biologically active, peptides attached to human Fc fragment. On administration of FPX in 76 healthy human subjects, 37% developed antibodies after a single injection; immune responses were limited to individuals who had the HLA that could present the immunogenic regions of the peptide. A memory T-cell response against the carboxy-terminus of the peptide was both predicted and observed. As predicted by iTEM (individualized T cell epitope measure), HLA-haplotype DRB1*0701/1501 was associated with the highest T-cell and antibody response.
This is one of many examples that confirm that in-silico prediction can be successfully used to identify Class II restricted T-cell epitopes within therapeutic proteins and predict immunogenicity in humans. For monoclonal antibodies, a close correlation between silico immunogenicity assessment and the confirmed immunogenicity of monoclonal antibodies in the clinic has also been documented.
Lastly, tolerance can be induced to protein therapeutics by exploiting the body’s own tolerance induction systems. We have successfully reduced the immunogenicity (allergenicity) of dust mite lysate, smallpox immunogen, and a number of other immunogenic proteins, both in vivo and in vivo (in HLA transgenic mice), using a novel T reg “adjuvant”.
BioMarkers for immunogenicity will improve clinical trial success rates and lead to safer biological therapeutics. What’s important is that these tools are available now – companies that implement them will have a distinct advantage over their competitors. This presentation will illustrate the use of these readily available tools to pre-determine immunogenicity. Techniques for salvaging immunogenic therapeutics will also be addressed.

   

12:00

Progress of Mass Spectrometry Bioanalysis for Proteins in Drug Development

 

Qin Ji, Ph.D., Senior Director of Bioanalytical Services, Covance

   
 

The determination of protein concentrations in plasma samples often provides essential information in biomedical research, clinical diagnostics, and pharmaceutical discovery and development. Binding assays such as ELISA determine protein concentrations by using specific antigen or antibody reagents. Concurrently, mass spectrometric technology is becoming a promising complemen

 
 
Organized by: GTCbio
Invited Speakers: Michael Hanley
VP, Discovery Research & CSO
Amylin Pharmaceuticals
 
Deadline for Abstracts: August 8, 2008
 
Registration:

Register by August 8 for a 10% Discount. 

E-mail: raniah@gtcbio.com
 
   
 
home   genetic news   bioinformatics   biotechnology   literature   journals   ethics   positions   events   sitemap
 
 
 

Generated by meetings and positions 5.0 by Kai Garlipp
WWW: Kai Garlipp, Frank S. Zollmann.
7.0 © 1995- HUM-MOLGEN. All rights reserved. Liability, Copyright and Imprint.