Scientific Program

Conference Series Ltd invites all the participants across the globe to attend Annual Congress on Antibiotics and Antimicrobial Resistance
Hotel Hyatt Place Amsterdam Airport, Netherlands
.

Day 1 :

Keynote Forum

Geert C. Mudde

Chief Scinetific Officer, OncoQR ML GmbH, Austria

Keynote: Next generation immuno-therapy: tumour specific control of immune checkpoints

Time : 09:55 A.M -10:45 A.M

Conference Series Antibiotics Summit 2018 International Conference Keynote Speaker Geert C. Mudde photo
Biography:

Dr. Geert C. Mudde received a Ph.D. in immunology from the University of Utrecht in 1985 and started his international professional career at the Swiss Institute for Asthma and Allergy Research in Davos in 1989. In 1992 he joined the pharmaceutical/biotech industry, where he held several senior management positions at the Novartis Research Institute in Vienna, Austria, the Parke Davis Research Institute in Fresnes, France, Ingenium Pharmaceuticals, Martinsried, Germany, and at igeneon AG, Vienna, Austria. Finally, in 2006, while joining Baxter BioScience in Vienna as interim manager, Dr. Mudde co-founded the biotech company f-star Biotechnology, where he served as “Chief Scientific Officer” from 2007 to 2009. In 2009, together with Christof Langer, he started to develop the S-TIR™ technology platform for human specific therapeutic vaccines which led to the foundation of S-TARget therapeutics GmbH in 2010. Since then he serves as CSO and managing director for S-TARget therapeutics as well as for the S-TIR™ technology spin-off companies OncoQR ML GmbH and TYG oncology Ltd., which were both founded in 2013.

Abstract:

Using the S-TIR™ technology platform for human specific therapeutic vaccines OncoQR ML has developed two prototype vaccines for treatment of pancreatic cancer (TYG100) and breast cancer (OQR200). Vaccines derived from this platform consist of 2 modules, the disease specific module, “immunogen” and the generic module, “warhead”, which directs the vaccines to CD32 on antigen presenting cells, especially pDCs and B cells. The immunogen in oncology is a tumour associated auto-antigen, against which under normal conditions no clinically relevant immune responses can be induced. We will present conclusive proof that it is finally possible to overcome all the tricks of cancer cells to prevent therapeutic immune responses. No more need for bulk infusion of very expensive and artificial monoclonal antibodies, which either try to mimic tumour specific B cell responses (e.g. Herceptin and Perjeta) OR try to activate cytotoxic T cells, that by chance may also kill tumours (e.g. Opdivo, Yervoy, Keytruda). S-TIR™ vaccines fully activate both arms of the patient’s own immune system resulting in tumour specific polyclonal IgG responses simultaneously with the generation and activation of tumour specific cytotoxic T cells. The responses are reversible and boostable, thus allowing fine-tuning of the clinical responses on a patient to patient basis. S-TIR™ vaccines in contrast to the current checkpoint inhibitors do not induce autoimmune disease and are tumours specific

 

Conference Series Antibiotics Summit 2018 International Conference Keynote Speaker Fariba Ahmadizar photo
Biography:

Mario Rodriguez has his expertise in Virology and Bioprocess Development. Working with recombinants and their aplications against viral diseases as Influenza, Ebola and members of the Polyomaviride family like the newly discovered MCPyV. Scientist and Innovation manager with expertise in prototype technology projects that results in commercial technology (i.e. High fidelity DNA polymerase under commercialization, vaccines and Mononoclonal antibodies, licensed), advisor of technological companies in USA and Mexico. He is part in Mexico from SNI (National Researchers System) from CONACYT.

 

Abstract:

Currently, production of vaccines and diagnostic systems for infectious diseases have failed to provide a systematic vision that merges state-of-the-art technologies with industry to provide an effective commercial solution. Infectious and rapidly transmitted diseases, such as Cancer, Ebola and Influenza, should be a focus of interest for these prospects.

While technological advances of recent years have been revolutionizing the life sciences industry, specifically in the biopharma field, these advances have been disproportional in terms of their applications towards infectious diseases.

Working on the development of recombinant technology is needed for the production of chimeric proteins using mammalian, yeast, and bacterial cells modified for those purposes. Proteins developed through a process of molecular engineering, which begins with in silico bioinformatic processes, using validations and algorithms, subsequently through synthetic biology, molecular biology, genetic engineering, and BioProcess development. The aim being, the scaling efforts towards pilot plant levels. The primary goal of these proteins is the development of integrated solutions that can be used as antigens or antibodies in diagnostic systems, as well treatments and vaccines.

The main challenge is in the final application that results in the free exposure of epitopes for recognition between the antibody and the antigen of interest, which implies their effectiveness in terms of use. A secondary challenge is productivity rates in bio-production systems, which vary greatly depending on the platform used and the quality of the bioprocess developed.

The recombinant proteins HA-RBD, tAg, ScFv-13F6, ScFv-13C6 and FAb-KZ52 were designed, developed, expressed and characterized by the integral use of molecular engineering and bioprocess engineering. The expressed proteins showed biological antibodies (HA-RBD and tAg) and antigen (ScFv-13F6 and ScFv-13C6) recognition, recognizing specific epitopes. Significantly tAg production occurred with a yield of 50 mg L-1 and HA-RBD protein was produced in 120 mg L-1.

  • Vaccines | Therapeutic Cancer Vaccines | Antimicrobials | Recombinanat Vaccines | Antibiotics
Speaker
Biography:

Carlie Skellington is a MPH candidate at Columbia University Mailman School of Public Health studying epidemiology and humanitarian assistance. She graduated with honors from Lehigh University in 2015 with a B.S. in Biology, during which she worked in Dr. Jill Schneider’s neuroendocrinology laboratory and completed her independent thesis research project on asesssing environmentally-induced variability of morphological, ingestive, and reproductive traits in the male Syrian hamster based on living conditions (group-housed vs. singly-housed). Carlie has also worked within a sustainable development lab, during which she helped design and develop a phytopurifier that degrades common indoor home air pollutants within Pennsylvania, USA.

Abstract:

From misunderstanding to misuse of antimicrobials, Uganda experiences high rates of resistance to antimicrobials that are heavily relied upon to treat prevalent infections, including pneumonias, bacterial meningitis, and respiratory infections. Prior literature suggests seasonal periodicity of bacterial infections, typically with increased prevalence when higher temperature and lower humidity. Unfortunately, Uganda has yet to implement a national, systematic AMR surveillance system. The aim of this retrospective cohort study was to identify AMR seasonality trends (rainy vs. dry season) among isolates collected by MNRH’s Microbiology Laboratory in Kampala, Uganda. Demographic variables and antibiotic sensitivity (S/I/R) for positive cultures were assessed via laboratory chart review of blood specimen (n=552), CSF isolates (n=742), and culture swabs (n=508) from patients treated between January and December, 2017. Controls were selected via systematic sampling of negative cultures. Two-tailed chi-square tests compared frequencies of antibiotic use and sensitivities. Average monthly rainfall and humidity were retrieved from IAMAT. The most commonly prescribed antibiotics included SXT, TE, CTX, AMP, and C. Blood isolates and culture swabs did not show differences in AMR according to season at the 5% significance level. Resistance within CSF cultures, however, differed significantly by season, showing a 22% greater risk of resistant strains identified during rainy months than during dry months (c2=4.19, p=0.04, RR=1.22). Based on preliminary analysis, AMR trends within CSF cultures appear to mirror Kampala’s seasonal patterns. Multivariate regression analysis may account for potential selection bias and confounding. Results warrant additional AMR research in order to improve surveillance and better inform practitioner prescription habits.

Giulio F. Tarro

Foundation de Beaumont Bonelli for Cancer Research, Italy

Title: Tumor associated antigen as potential vaccine for cancer patients
Speaker
Biography:

Giulio Tarro graduated from Medicine School, Naples University (1962). Research Associate, Division of Virology and Cancer Research, Children’s Hospital (1965-1968), Assistant Professor of Research Pediatrics, College Medicine (1968-1969), Cincinnati University, Ohio. Oncological Virology Professor, Naples University (1972-1985). Chief Division Virology (1973-2003), Head Department Diagnostic Laboratories, (2003-2006). D. Cotugno Hospital for Infectious Diseases, Naples; Emeritus, 2006 -. Since 2007 Chairman Committee of Biotechnologies and VirusSphere, World Academy Biomedical Technologies, UNESCO, Adjunct Professor Department Biology, Temple University, College of Science and Technology, Philadelphia, recipient of the Sbarro Health Research Organization lifetime achievement award (2010). His researches have been concerned with the characterization of specific virus-induced tumour antigens, which were the "finger-prints" left behind in human cancer. Achievements include patents in field; discovery of Respiratory Syncytial Virus in infant deaths in Naples and of tumor liberated protein as a tumor associated antigen, 55 kilodalton protein overexpressed in lung tumors and other epithelial adenocarcinomas.

Abstract:

Tumor liberated protein (TLP) has been previously described as a TAA (complex) present in the sera from lung cancer patients with early stage disease.

Since early detection improves overall survival in lung cancer, identification of screening biomarkers for patients at risk for the development of this disease represents an important target. Starting from the peptide epitope RTNKEASI previously isolated from TLP complexes, we generated a rabbit anti-RTNKEASI serum. This antiserum detected and immunoprecipitated a 55kDa protein band in the lysate of the lung cancer cell line A549. This protein band was identified as aldehyde dehydrogenase isoform 1A1 through mass spectrometry, revealing the molecular nature of at least one component of the previously described TLP complex. Next, we screened a cohort of 29 lung cancer patients (all histologies), 17 patients with non-neoplastic lung pathologies and 9 healthy donors for the presence of serum ALDH1A1 and global serum ALDH by enzyme-linked immunosorbent assay. This analysis indicated that the presence of ALDH was highly restricted to patients with lung cancer. Interestingly, the global ALDH test detected more lung cancer patients compared to the ALDH1A1-specific test, suggesting that other ALDH isoforms might add to the sensitivity of the assay. Our data suggest that ALDH levels may therefore be evaluated as part of a marker panel for lung cancer screening.

Finally, the ability of the immune system to recognize a TAA, enables the development of a vaccine approach for preventive and therapeutic application and represents a main target of this field of research.

 

Biography:

Abstract:

Biography:

Abstract:

Biography:

Abstract:

Ali Khamesipour

Tehran University of Medical Sciences, Iran

Title: Vaccination via natural infection - Leishmanization
Biography:

Abstract:

Samer Ellahham

Cleveland Clinic Abu Dhabi, UAE

Title: Antibiotic stewardship: A necessity not a luxury
Speaker
Biography:

Certified Professional in Healthcare Quality (CPHQ) and in Medical Quality (CMQ) by The American Board of Medical Quality (ABMQ). Lean Six Sigma Master Black Belted. Recipient of the Quality Leadership Award from the World Quality Congress and Awards and the Business Leadership Excellence Award from World Leadership Congress in 2015.

Abstract:

Antimicrobial resistance is increasing, and antimicrobial stewardship is of the utmost importance to optimize the use of antimicrobials to prevent the development of resistance and improve patient outcomes. The World Health Organization (WHO) declared antimicrobial resistance a global health issue. The U.S. Centers for Disease Control and Prevention (CDC) reports that each year in the United States approximately 2 million people develop an infection that is resistant to antibiotics and about 23,000 of these people die because of the infection. The program for antibiotic stewardship should involve infection prevention professionals, physicians, nurses, pharmacists, trainees, patients, and families. Patients and families are included as part of the program to ensure they understand the importance of compliance with antibiotic treatment. Included as part of the program is the need for proper use of antibiotics for prophylaxis in addition to properly prescribing antibiotics for infections. Implementing an antibiotic stewardship program takes time and resources. To gain hospital wide acceptance and increase success for the program, formal implementation is recommended as well as leadership support for staffing, technical, and financial resources. Antimicrobial stewardship incorporation in caregiver daily practice, can improve patient safety and care, reduce the unnecessary use of valuable resources, and reduce resistance.

Speaker
Biography:

Dr. Omar F. Khan is Chief Scientist of Tiba Biotech and the lead inventor of Tiba’s disruptive RNA delivery technology.  His expertise includes nanomaterial-based nucleic acid delivery systems, vaccination nanotechnology, organic chemistry and manufacturing. He is equally skilled in biomaterial synthesis, tissue engineering, reactor design, process engineering, translational research models, and scale-up production.  Omar is widely published on topics at the interface of materials and life science and has been cited over 1000 times. He holds a B.A.Sc. and Ph.D. in Chemical Engineering from the University of Toronto, where he was cross-appointed with the Institute of Biomaterials and Biomedical Engineering. Prior to founding Tiba, Omar conducted his postdoctoral research in the labs of Robert Langer and Daniel Anderson at MIT.  Omar leads delivery platform development and manufacturing design, drawing upon a wide breadth of scientific and leadership experience from multiple sectors, including academia, biotechnology, pharmaceutical, petrochemical and water treatment.

Abstract:

To combat orphaned indications, sudden outbreaks, evolving pathogens and biosecurity threats, Tiba Biotech has developed a fully synthetic replicon mRNA platform for the rapid design and scalable manufacture of vaccines that generate protective cellular (T cell) and humoral (antibody) responses against a range of diseases. The technology consists of two major components: an engineered antigen-expressing replicon mRNA payload capable of finite, controlled self-replication that induces potent interferon responses, and a chemically-defined modified dendrimer-based delivery material.  The engineering advances embodied in this vaccine platform delivery technology include: (1) thermostable components that facilitate a straightforward self-assembly process.  This capability reproducibly yields synthetic adjuvant-free mRNA nanoparticles of uniform size and shape, thus enabling rapid formulation of the drug product; (2) creation of stable nanoparticles that do not expose or release the replicon mRNA extracellularly, which would prematurely activate innate immunity and shut down the ability to express the exogenous mRNA; (3) a large, flexible payload capacity for multiplexing, which is the co-delivery of multiple large nucleic acid molecules in a single nanoparticle, thus enabling the simultaneous transport of complex or multiple subunit antigens and potentially the induction of cross-protective immunity to multiple strain sequences; and (4) an ionizable nature to prevent cytotoxicity and a systemic increase in inflammatory cytokines.  Demonstrative of this platform’s broad utility, it has been used to generate protective immunity in multiple lethal challenge models, including Ebola virus, Venezuelan Equine Encephalitis, H1N1, Toxoplasma gondii and HPV-induced cancer. Additionally, candidate vaccines have been developed against Zika virus and parasitic flatworms.  Furthermore, early delivery material safety studies in nonhuman primates and livestock animals showed no reactogenicity, highlighting its tolerability and potential for both clinical translation and animal health.