Call for Abstract

4th Antibodies, Antibiotics and Bio Therapeutics Congress: R&D, Market and B2B, will be organized around the theme “Novel Research and Therapeutic Challenges in Antibodies and Antibiotics”

Antibodies-2018 is comprised of keynote and speakers sessions on latest cutting edge research designed to offer comprehensive global discussions that address current issues in Antibodies-2018

Submit your abstract to any of the mentioned tracks.

Register now for the conference by choosing an appropriate package suitable to you.

Antibody engineering is a great tool for improving antibody functions and immunogenicity improvement. Engineered therapeutic antibodies are better for affinity maturation, specifically by improving on-rate of the antibody binding affinities. The need to overcome the immunogenicity problem of rodent antibodies in clinical practise has resulted in a plethora of strategies to isolate human antibodies.

  • Track 1-1Antibody Biosynthesis, Structure and Stability
  • Track 1-2Bi-Specific Antibody Engineering
  • Track 1-3New Ways to Extend the in vivo Half-Life of Antibodies
  • Track 1-4Fc Engineered Antibodies and Special Antibody Engineering
  • Track 1-5Unique Therapeutic Vistas in Antibody Engineering: Bispecifics
  • Track 1-6Antibodies in a Complex Environment

The evidence indicates that two simian immunodeficiency viruses (SIV), one from Chimpanzees (SIVcpz) and the other from sooty mangabeys (SIVsm), crossed the species barrier to humans, generating HIV-1 and HIV-2, respectively. The importance of characterizing the prevalence, geographic distribution, and genetic diversity of naturally occurring SIV infections to investigate whether humans continue to be exposed to SIV and if such exposure could lead to additional zoonotic transmissions. Through vigorous efforts made in the past two centuries, public health workers have succeeded in developing vaccines, antibiotics, and chemotherapeutics, and as a result most infectious diseases have been brought under control in industrialized countries. However, in developing countries, infectious diseases have been harder to contain, and the increase in migration and movement of populations in the last two decades has made national boundaries disappear as far as the transmission of infection is concerned. Some diseases, such as malaria, have been eradicated from industrialized countries mainly through extensive work on vector control, but their presence in developing countries has increased because of neglect or drug resistance. One of these proteases (Histidine Aspartic Protease, HAP) is homologous to three other aspartic proteases involved in hemoglobin metabolism but has a histidine in place of one of the two aspartic acids involved in catalysis.

  • Track 2-1The Growing Challenge of Antibiotic Resistance
  • Track 2-2Antibodies for Pediatrics Diseases
  • Track 2-3Therapeutic Antibodies for Neurological Disease and other Neglected Diseases
  • Track 2-4Novel Therapeutic Antibodies for Infectious and Inflammatory Diseases
  • Track 2-5Novel Therapeutic Antibodies for Allergy, Asthma and Autoimmune Diseases

Auto-antibody is an antibody formed in response to, and reacting against, an antigenic constituent of the individual's own tissues. Several mechanisms may trigger the production of autoantibodies: an antigen, formed during fetal development and then sequestered, may be released as a result of infection, chemical exposure or trauma, as occurs in autoimmune thyroiditis, sympathetic uveitis and aspermia; there may be disorders of immune regulatory or surveillance function.     

  • Track 3-1Remarkable Findings of Auto-antibodies: Testing and Treatment
  • Track 3-2Antibody Fingerprinting: A Novel Method for Identifying Individual People and Animals
  • Track 3-3Current insights into the
  • Track 3-4Autoantibodies in infertility: current opinion
  • Track 3-5The origin and pathogenic role of anti-DNA autoantibodies

Monoclonal antibody therapy is a process in which monoclonal antibodies (mAb) are used to bind monospecifically to certain cells or proteins. This may then stimulate the patient's immune system to attack those cells. Alternatively, in radioimmunotherapy a radioactive dose localizes on a target cell line, delivering lethal chemical doses. More recently antibodies have been used to bind to molecules involved in T-cell regulation to remove inhibitory pathways that block T-cell responses, known as immune checkpoint therapy. It is possible to create a mAb specific to almost any extracellular/ cell surface target. Research and development is underway to create antibodies for diseases (such as rheumatoid arthritis, multiple sclerosis, Alzheimer's disease, Ebola and different types of cancers).    

  • Track 4-1Advantages in Monoclonal Antibody Therapy
  • Track 4-2Monoclonal and Polyclonal antibodies
  • Track 4-3Monoclonal Antibody Therapy for Cancer
  • Track 4-4Monoclonal Antibodies Applications
  • Track 4-5Hybridoma Technology and Monoclonal Antibody Preparation
  • Track 4-6Antibody Research in Agriculture, Aquaculture, Veterinary and Food Sciences
  • Track 4-7Antibody Technology in New Protein Discovery

Antibodies are used extensively as diagnostic tools in many different formats. The term applied for antibody based diagnostic tests is “immunoassay”. Antibody-based immunoassays are the most commonly used confirmatory diagnostic assays and is the fastest growing technologies for the analysis of biomolecules. Trends in antibody based diagnosis show advances in assay specificity, detection technologies and sensitivity. Sensitivity and specificity is ensured depending on whether or not the antigen to be quantified competes with labeled antigen for a limited number of antibody binding sites. Monoclonal antibodies are now widely used in all areas of biological and medical research as well as in clinical diagnostic tests and in therapy. This review concentrates on the clinical use of antibodies in therapy particularly with regard to the properties of the antibodies which seem most relevant to their usefulness. In-vitro tests using human effector systems and in-vivo animal models have demonstrated the importance of the antibody isotype and valency for antigen as well as the specificity of binding.

  • Track 5-1Novel Antibodies in Medical Treatment and Diagnosis
  • Track 5-2Antibodies for Immuno histochemistry and Immunofluorescence
  • Track 5-3Antibody for Western Blot Analyses, ELISA and ELISPOT Techniques

Antibody therapies are the most successful immunotherapy, treating a wide range of cancers. Antibodies are proteins produced by the immune system that bind to a target antigen on the cell surface. In normal physiology the immune system uses them to fight pathogens. Each antibody is specific to one or a few proteins. Those that bind to cancer antigens are used to treat cancer. Cell surface receptors are common targets for antibody therapies. Among the most promising approaches to activating therapeutic antitumor immunity is the blockade of immune checkpoints. Immune checkpoints refer to a plethora of inhibitory pathways hardwired into the immune system that are crucial for maintaining self-tolerance and modulating the duration and amplitude of physiological immune responses in peripheral tissues in order to minimize collateral tissue damage. It is now clear that tumors co-opt certain immune-checkpoint pathways as a major mechanism of immune resistance, particularly against T cells that are specific for tumor antigens. Because many of the immune checkpoints are initiated by ligand–receptor interactions, they can be readily blocked by antibodies or modulated by recombinant forms of ligands or receptors. Cytotoxic T-lymphocyte-associated antigen 4 (CTLA4) antibodies were the first of this class of immunetherapeutics to achieve US Food and Drug Administration (FDA) approval.

  • Track 6-1Combinations of Check-Point Inhibitors
  • Track 6-2In Vivo Discovery of Immunotherapy Targets
  • Track 6-3Immune Suppression and Microenvironment
  • Track 6-4Sequencing or Combination Approaches in Immunotherapy
  • Track 6-5Chimeric Antigen Receptor Re-Directed T Cell Therapy

Conventional anticancer therapeutics often suffer from lack of specificity, resulting in toxicities to normal healthy tissues and poor therapeutic index. Antibody-drug conjugates (ADCs) constitute a therapeutic modality in which a cytotoxic agent is chemically linked to an antibody (Ab) that recognizes a tumor-associated antigen. The basic strategy underlying ADC technology is to combine the target selectivity of mAbs with the potency of cytotoxic agents, such as certain natural products and synthetic molecules, with the goal of generating therapeutic drugs that are highly efficacious but also safe. The ADC platform currently includes a growing repertoire of cytotoxic payloads, linker technologies and conjugation methods. Two ADCs have recently received FDA approval and more than 30 are in clinical development. This meeting aims to highlight advances in ADC research, clinical development and regulatory perspectives.

  • Track 7-1Innovative Antibody drugs in Clinical Trail and Development
  • Track 7-2Antibody-drug Conjugate (ADC) Discovery and Development
  • Track 7-3Bispecific and Multispecific antibody discovery and development
  • Track 7-4Challenges and New Opportunities with Antibody and Protein Drugs
  • Track 7-5Novel Antibody and Biologic Drug Targets

Anticancer antibodies  has created great interest in antibody-based therapeutics for hematopoietic malignant neoplasms and solid tumors. Given the likelihood of lower toxic effects of antibodies that target tumor cells and have limited impact on nonmalignant bystander organs vs small molecules, the potential increased efficacy by conjugation to radioisotopes and other cellular toxins, and the ability to characterize the target with clinical laboratory diagnostics to improve the drug's clinical performance, current and future antibody therapeutics are likely to find substantial roles alone and in combination therapeutic strategies for treating patients with cancer. Therapeutic antibodies have become a major strategy in clinical oncology owing to their ability to bind specifically to primary and metastatic cancer cells with high affinity and create antitumor effects by complement-mediated cytolysis and antibody-dependent, cell-mediated cytotoxicity (naked antibodies) or by the focused delivery of radiation or cellular toxins (conjugated antibodies).

  • Track 8-1Antibodies in Treatment of Cancer
  • Track 8-2Characterizing and clinical use of Circulating Tumor Cells (CTCS)
  • Track 8-3Advances in tumor modeling, cancer drug discovery and development
  • Track 8-4Translational approaches in cancer immunotherapy development
  • Track 8-5Innovative in vitro and in vivo modeling

Cancer immunotherapy—treatments that harness and enhance the innate powers of the immune system to fight cancer—represents the most promising new cancer treatment approach since the development of the first chemotherapies in the late 1940s. Because of the immune system’s extraordinary power, its capacity for memory, its exquisite specificity, and its central and universal role in human biology, these treatments have the potential to achieve complete, long-lasting remissions and cancer cures, with few or no side effects, and for any cancer patient, regardless of their cancer type. Immunotherapy is treatment that uses your body's own immune system to help fight cancer. These treatment modalities are all based on destroying cancer cells by burning them (irradiation), poisoning them (chemotherapy) or removing them (surgery). While they can effectively kill or remove cancer cells, the use of these treatments often is limited because large numbers of healthy cells also tend to be destroyed. This often results in extreme morbidity and/or disfigurement of the patients treated with them.

  • Track 9-1Evolving Concepts in Cancer Immunology
  • Track 9-2Anti-tumor activity of immunomodulatory antibodies
  • Track 9-3Tumor Antigens for Targeting: Insights from Genomics
  • Track 9-4Impact of the immunogenic landscape of cancers on immunotherapy
  • Track 9-5Cancer neo-antigens

The first monoclonal antibodies were typically made entirely from mouse cells. One problem with this is that the human immune system will see these antibodies as foreign (because they’re from a different species) and will mount a response against them. In the short term, this can sometimes cause an immune response. In the long term, it means that the antibodies may only work the first time they are given; after that, the body’s immune system is primed to destroy them before they can provide treatment. This study presents a technology that generates stable, soluble, ultra-humanized antibodies via single-step CDR redundancy minimization. Lead clones demonstrated high stability, with affinity and specificity equivalent to, or better than, the parental immunoglobulin. This significantly lowered non-human sequence content, minimized t- and b-cell epitope risk in the final molecules and provided a heat map for the essential non-human CDR residue content of antibodies from disparate sources. Antibody humanization uses multiple sequence segments derived from variable (V) regions of unrelated human antibodies, unlike other technologies that typically use a single human V region framework as acceptors for complementarity determining regions (CDRs) from starting antibodies (typically rodent). Through careful selection of human sequence segments and the application of in silico tools, CD4+ T cell epitopes are avoided so the risk of immunogenicity is reduced compared to standard humanized antibodies whilst antibody affinity and specificity is maintained. Immunogenicity assessment technology is used to confirm T cell epitopes have been removed.

  • Track 10-1Rapid Humanization and Affinity Improvement of a Murine Antibody
  • Track 10-2IMGT Databases and Tools for Antibody Engineering and Humanization
  • Track 10-3Immunogenicity Assessment Strategies to Support the Development of Biological Therapeutics
  • Track 10-4Therapeutic Antibody Discovery and Development Using Humanized RabMAbs
  • Track 10-5Humanized Antibodies and Their Therapeutic Value
  • Track 10-6Non-human Primate Immune Libraries Combined with Germline Humanization
  • Track 10-7A Novel Fully Human Monoclonal Antibody Platform Using Transgenic Rats

Antibiotics are a type of antimicrobials used in treatment and prevention of bacterial infections. They may inhibit or kill the growth of bacteria. Many antibiotics are also effective against protozoans and fungi; some are toxic to animals and humans also, even when given in therapeutic dosage. Antibiotics are not effective against viruses such as influenza or common cold, and may be harmful when taken inappropriately. Physicians must ensure the patient has a bacterial infection before prescribing antibiotics.

  • Track 11-1Antibiotics: Uses and Challenges
  • Track 11-2Antibiotics: Basic Principles for prescribing
  • Track 11-3Antibiotics: Mechanism of bacteriostatic action
  • Track 11-4Antibiotics: Mechanism of bacteriostatic action

New diseases are arising worldwide and old diseases are re-emerging as Infectious agents evolve or spread, and as changes occur in ecology, socioeconomic conditions, and population patterns. Likewise, many diseases thought to be adequately controlled appear to be making a comeback. In developed countries, public health measures such as sanitation, sewage treatment, vaccination programs, and access to good medical care-including a wide range of antibiotics-have virtually eliminated “traditional” diseases such as diphtheria, whooping cough, and tuberculosis.

  • Track 12-1Resistance and re-emerging theories
  • Track 12-2Medication procedures
  • Track 12-3New drugs for emerging diseases
  • Track 12-4Molecular mechanism of resistance

Antibacterial action generally falls within one of four mechanisms, three of which involve the inhibition or regulation of enzymes involved in cell wall biosynthesis, nucleic acid metabolism and repair, or protein synthesis, respectively. The fourth mechanism involves the disruption of membrane structure. Many of these cellular functions targeted by antibiotics are most active in multiplying cells. Since there is often overlap in these functions between prokaryotic bacterial cells and eukaryotic mammalian cells, it is not surprising that some antibiotics have also been found to be useful as anticancer agents.

Antibiotics, also known as antibacterials, are types of medications that destroy or slow down the growth of bacteria. In 1929, Alexander Fleming identified penicillin, the first chemical compound with antibiotic properties. Some of the common antibiotics are Penicillins, Cephalosporins, Carbapenems, Macrolides, Aminoglycosides, Quinolones, Sulfonamides and, Tetracyclines etc. General principles of antibiotic prescribing are use: First-line antibiotics first, Reserve broad spectrum antibiotics for indicated conditions only, prescribe antibiotics for bacterial infections if Symptoms are significant or severe.

The antibiotics market generated sales of US$42 billion in 2009 globally, representing 46% of sales of anti-infective agents (which also include antiviral drugs and vaccines) and 5% of the global pharmaceutical market1. However, the antibiotics market is maturing; it showed an average annual growth of 4% over the past 5 years, compared with a growth of 16.7% and of 16.4% for antiviral drugs and vaccines, respectively.

The cephalosporin class of antibiotics is the largest in terms of sales, generating $11.9 billion in 2009, led by sales of the latest generation of drugs in this class (cefcapene (Flomox; Shionogi), ceftriaxone (Rocephin; Roche) and cefuroxime (Zinnat; GlaxoSmithKline). This class represents 28% of the total antibiotic market, and sales showed a growth of 3.4% over the past 5 years. With sales of $7.9 billion and 19% of the antibiotic market share in 2009, the second largest drug class is the broad-spectrum penicillins, which showed a growth of 5% between 2005 and 2009. The third largest drug class — the fluoroquinolones — had sales of $7.1 billion in 2009, accounting for 17% of the antibiotic market in 2009, and also showed an average growth of 5% between 2005 and 2009. By contrast, as generic versions of an increasing number of macrolides — which had $4.8 billion in sales in 2009 — became available, sales of this class declined by 5% between 2007 and 2009. Overall, the rate of patent expiry of leading antibiotics in the market is set to increase, with several of the current top-selling products facing patent expiry between 2010 and 2016. These include levofloxacin (Levaquin; Johnson & Johnson), moxifloxacin (Avelox; Bayer/Merck) and linezolid (Zyvox; Pfizer), which are expected to lose patent protection in 2011, 2014 and 2016, respectively.

  • Track 14-1Antibacterial Agents
  • Track 14-2Antifungal Agents
  • Track 14-3Penicillin
  • Track 14-4Anti-Migraine Agents
  • Track 14-5Immunosuppressive agents

Prescribing doctors are, increasingly, using clinical trial data as a major source of information for evidence-based medicine for the treatment of infectious diseases, as in other clinical disciplines. However, it may be difficult to extract from these data the information that is needed for the management of the individual patient. At the same time, clinical trial data have been used, apparently satisfactorily, in the process of drug registration, and the pharmaceutical industry has spent increasingly large sums of money to satisfy the needs of this process. In the face of all these problems, changes in the way antibiotic clinical trials are designed and performed are clearly necessary, although this must not tip the balance so far as to render them less useful for those who currently derive greatest benefit from them.

  • Track 15-1Evaluation of Efficacy
  • Track 15-2Evaluation of Safety
  • Track 15-3Clinical Microbiology
  • Track 15-4Clinical Bio-Chemistry

Although the field of antibiotics is developing so rapidly as to render it almost kaleidoscopic, during recent months several topics of general interest have come into sharp focus. The subject of first consideration will include not only new drugs but also new forms of old drugs, as well as old drugs. Beyond this, I shall confine my discussion to two main items, bacterial resistance to antibiotics, and antibiotic toxicity. A variety of biological solutions have yet to be fully explored.

  • Track 16-1Randomized Control Trails
  • Track 16-2Empiric Antibiotic Therapy
  • Track 16-3Bacterial Biofilms
  • Track 16-4Intra-Adhesive Antibiotics
  • Track 16-5Oral Antibiotic Therapy

Antibiotics must be used judiciously in humans and animals because both uses contribute to the emergence, persistence, and spread of resistant bacteria. Resistant bacteria in food-producing animals are of particular concern. Food animals serve as a reservoir of resistant pathogens and resistance mechanisms that can directly or indirectly result in antibiotic resistant infections in humans. For example, resistant bacteria may be transmitted to humans through the foods we eat. Some bacteria have become resistant to more than one type of antibiotic, which makes it more difficult to treat the infections they cause. Preserving the effectiveness of antibiotic drugs is vital to protecting human and animal health.

  • Track 17-1Antibiotics in Food Industry
  • Track 17-2Antibiotics in Agriculture
  • Track 17-3Antibiotics in Veterinary
  • Track 17-4Antibiotics in Aquaculture

Biopharmaceuticals is one of the fastest growing segments in the pharmaceutical industry. They have a vital use in the treatment of chronic diseases and also result in high profit margins for the drug developers. There are several therapeutic areas for which biopharmaceuticals are being investigated; these include oncology, metabolic disorders, viral infections, genetic disorders and immunological disorders

  • Track 18-1Formulation and delivery issues for monoclonal antibody therapeutics
  • Track 18-2Microneedles for drug and vaccine delivery
  • Track 18-3Biopharmaceutical formulations for pre-filled delivery devices
  • Track 18-4Nanotechnological advances for the delivery of CNS therapeutics
  • Track 18-5Biotherapeutics in Orthopaedic Medicine

Antibody conjugates are a diverse class of therapeutics consisting of a cytotoxic agent linked covalently to an antibody or antibody fragment directed toward a specific cell surface target expressed by tumor cells. The notion that antibodies directed toward targets on the surface of malignant cells could be used for drug delivery is not new.

  • Track 19-1Antibody-Drug Conjugates Design And Development
  • Track 19-2Bispecific antibody conjugates in therapeutics
  • Track 19-3Strategies and Advancement in Antibody-Drug Conjugate Optimization for Targeted Cancer Therapeutics
  • Track 19-4New Developments in Antibody-Drug Conjugates

Next-generation sequencing (NGS) provides a quantitative approach to measuring the diversity and distribution of antibody libraries. It will introduce and enable researchers on how to design, analyze, and perform antibody NGS studies and how these can be applied for discovery and engineering of monoclonal antibodies from both synthetic and immune libraries. 

  • Track 20-1Nanoengineering Strategies for Antibody- Nanoparticle Conjugates
  • Track 20-2Next-Generation Sequencing Of Antibody Libraries
  • Track 20-3Biologics for Autoimmune Diseases
  • Track 20-4Biologics and Vaccines for Infectious Diseases
  • Track 20-5NGS Clinical Applications & Diagnostics

Therapeutic proteins and monoclonal antibodies (MAb) have transformed biotechnology and the pharmaceutical industry. Together they form the largest part of the rapidly growing biologics drug market. Protein-based drugs include blood factors, thrombolytic agents, hormones, hematopoietic growth factors, interferons, interleukins, tumor necrosis factor, and therapeutic enzymes. 

  • Track 21-1Protein Particulate Detection Issues in Biotherapeutics Development
  • Track 21-2Analytical Development For Emerging Biotherapeutics
  • Track 21-3Automation And Emerging Analytical Methods
  • Track 21-4Bioanalytical Considerations Of Multi-Domain Biotherapeutics: Preclinical And Clinical Development

New biotherapeutics formats are flooding the discovery and development pipelines and with this comes an increasing need for better and faster characterization tools and strategies, and improved biomolecular and biophysical assays for the new biotherapeutics.

  • Track 22-1Assay development
  • Track 22-2High throughput analytical development
  • Track 22-3Early stage bioassay development
  • Track 22-4Analytical development for novel biotherapeutics

Protein Engineering is the process of creating helpful or profitable proteins and it research happens into the comprehension of collapsing and acknowledgment for protein plan standards. Analysts will have further point by point learning on In vitro development of proteins, Aspects of Biocatalysis, Advances in designing proteins for biocatalysis, Protein Engineered Biomaterials and many subjects. Computational Protein Engineering, Constructing practical biocatalysts and Growth of manufactured science are likewise normally utilized themes as a part of protein designing. The protein engineering business sector is estimated to develop at a CAGR of 15.7% to reach $1,463.0 million by 2020. There are very nearly 3000 individuals from 60-65 colleges in USA working for Protein Engineering and there are a few meetings & workshops like biomolecular designing gatherings, sub-atomic cell science workshops, protein engineering meetings, antibody engineering 2015 are conducting throughout the year globally.

  • Track 23-1Advances in Protein Engineering
  • Track 23-2Applications: Protein Engineering
  • Track 23-3Advances in Genetic Engineering
  • Track 23-4Applications: Genetic Engineering