Scientific Program

Conference Series Ltd invites all the participants across the globe to attend 2nd International conference on Flu San Francisco, California, USA .

Day 1 :

Keynote Forum

Donald Pinkston Francis

Global Solutions for Infectious Diseases, USA

Keynote: WHO technology transfer activities for influenza vaccines

Time : 09:35 - 10:10

Conference Series Flu-2016 International Conference Keynote Speaker Donald Pinkston Francis photo
Biography:

Donald Pinkston Francis is a Physician trained in Pediatrics and Infectious Diseases. He has received his Doctorate in Science (in Virology) from Harvard School of Public Health. He has also served 21 years for the Centers for Disease Control and Prevention including assignments with WHO’s Smallpox Eradication Program in Sudan, India and Bangladesh. Beginning in 1993, he focused on vaccine development at Genentech and VaxGen before starting a not-for-profit vaccine development organization in 2004, Global Solutions for Infectious Diseases. He regularly consults for ‘WHO’ and other international organizations on vaccine development for influenza and dengue and on WHO’s eradication program for polio.

Abstract:

The World Health Organization states that, “Though the world is better prepared for the next pandemic than ever before, it remains highly vulnerable, especially to a pandemic that causes severe disease. Nothing about influenza is predictable, including where the next pandemic might emerge and which virus might be responsible.” WHO’s preparations to minimize the impact of influenza virus related disease cover a wide variety of activities that include surveillance of circulating viruses (in both birds and humans), supplying current circulating virus reagents for clinical diagnosis of cases, efforts to expand influenza vaccine production in lower and middle income countries and collaboration and support of trials to evaluate vaccines in people. The surveillance efforts are essential to identifying circulating viruses so subsequent control activities can be matched to the strain and clinical severity of the disease. In 2014, 142 laboratories in 112 countries joined together in what is known as the Global Influenza Surveillance and Response System. The laboratories in that system tested more than 1.9 million clinical specimens. To speed the diagnostic capabilities for this rapidly changing infection, WHO supplies reagents capable of identifying the latest circulating viruses; for example for the 2009 when the H1N1 outbreak was declared a public health emergency, WHO shipped out diagnostic reagents to laboratories within seven days. More recently WHO has put in place influenza specific systems to speed new vaccine production technologies and set systems to speed each country’s regulatory approval system for influenza vaccines. These activities are crucial to protecting the public’s health against the constant onslaught of new and emerging influenza viruses.

Keynote Forum

Ananda M Chakrabarty

University of Illinois College of Medicine, USA

Keynote: Drug development from bacterial pathogens to fight infections and cancer

Time : 10:10 - 10:45

Conference Series Flu-2016 International Conference Keynote Speaker Ananda M Chakrabarty photo
Biography:

Ananda M Chakrabarty is a Distinguished University Professor at the University of Illinois, College of Medicine at Chicago. His research interest involves development of promiscuous bacterial protein/peptide drugs with anticancer, anti-viral and anti-parasitic activities. He is the Co-Founder of two start-up companies, CDG Therapeutics Inc., in Chicago and Amrita Therapeutics in India.

Abstract:

It is now well-known that many pathogenic bacteria with long term residence in the human body as biofilms consider the human body as their habitat and try to protect it from outside invaders such as cancers, viruses and parasites through secretion of protein weapons. In one instance, Pseudomonas aeruginosa, an opportunistic pathogen, secretes a protein azurin on contact with cancer cells. Upon release, azurin enters preferentially to cancer cells and interferes in cancer cell growth through multiple mechanisms involving complex formation with various cellular proteins in cancer cells that promote cancer cell growth. Such complex formation then leads to loss of function of such cancer growth promoting proteins. Thus azurin is known to induce apoptosis in cancer cells, as well as interfere in rapid cancer cell growth, through stabilization of tumor suppressor protein p53. Azurin also forms complexes with vascular endothelial growth factor receptor (VEGFR) and cell surface associated receptor tyrosine kinases such as EphB2 to inhibit angiogenesis and cell signaling in cancer cells to inhibit their growth. A chemically-synthesized 28 amino acid fragment (Azurin 50-77), termed p28, has completed a phase I trial in 15 stage IV cancer patients with metastatic tumors that were resistant to all conventional drugs and these patients had a life expectancy of about 6 months. P28 not only showed very little toxicity but also significant beneficial effects including partial and complete regression of the tumors in four patients, significantly prolonging their lives. P28 has also shown similar lack of toxicity but good efficacy in several pediatric brain tumor patients. The University of Illinois at Chicago holds many patents on azurin/p28 as anticancer and anti-infective agents and the patent eligibility issues on such products of nature will be discussed.

Keynote Forum

Marek Malecki

Phoenix Biomolecular Engineering Foundation, USA

Keynote: Universal therapeutic vaccine against influenza

Time : 10:45- 11:20

Conference Series Flu-2016 International Conference Keynote Speaker Marek Malecki photo
Biography:

Marek Malecki MD PhD is President of the Phoenix Foundation, Visiting Professor at the University of Wisconsin, Principal Investigator for the National Institutes of Health. He earned the MD degree at the Medical Academy, Poznan followed by Residency/Fellowships in Molecular Medicine in Rigshospitalet, Copenhagen, Cancer Center, Vienna, Cancer Center, Amsterdam, NL, and Cancer Center, Warsaw. He earned the PhD degree at the Polish Academy of Sciences, Warsaw followed by the postdoctoral fellowships in molecular biology at the Austrian Academy of Sciences, Salzburg, ETH, Zurich, Utrecht University, Utrecht, Cancer Center, Amsterdam, Biozentrum, Basel. Over the last 20 years, he held faculty positions in Oncology, Molecular Medicine, and Pharmacology, at the top teaching hospitals and medical universities in the USA. There, he acquired solid experience in streamlining advances in genomics and proteomics into novel strategies of therapy, as well as in teaching medical students, residents, and fellows. He was elected by his students and fellow faculty to the Honor Society for Excellence in Teaching and as a Faculty Role Model. His research, as the Principal Investigator, was continuously funded by the grants from the National Science Foundation and the National Institutes of Health since 1989. He is the first or senior author on the peer-reviewed publications in the high impact journals, which are indexed on the PubMed and justified  hisrecommendation to the Faculty of 1000 Prime. He is the inventor of the gene therapies, therapeutic vaccines, and regeneration of tissues, which are published at the USPTO and WIPO with the sequences available through the NCBI. He was elected to serve as the Editor in Chief in peer-reviewed, open-access journals in Science, Technology, and Medicine, as well as Editorial Board Member and Reviewer at many others.

Abstract:

The CDC reports 3697 deaths due to influenza in 2013. First line therapeutics recommended for the patients who are already suffering from influenza are systemic therapeutics: oseltamivir (oral), zanamivir (inhaled), peramivir (intravenous) for patients who present within first 48 hours from the initial symptoms and for people who are at the high risk of exposure. Additionally recommended are: amantadine and rimantadine. Meanwhile, the preventive vaccination of patients 6 months – 17 years of age reached only 49.9%. However, these measures promoted by the CDC are not meant for the general populations. Importantly, the main problem with these medications is that efficacy of prophylactic and therapeutic vaccinations is hindered by the quickly changing genotypes of new strains of viruses. This leads to resistance to therapies of the new strains. Furthermore, with the changing genotype, surface displayed molecular profiles may also change. The changed phenotypes lead to ineffective therapies or absence of immune responses. Moreover, the development of the immune response takes time. It takes approximately 5-7 days for the 1st round of antibody generation. The booster shot or prolonged / secondary exposure is needed to yield the amplified response in 45-55 days. The sick patients cannot wait that long. Last but not least, the aforementioned therapeutics may inflict very serious iatrogenic adverse effects. To address these problems, by computer modeling and statistical analysis, we have designed and bioengineered universal therapeutic vaccine for influenza as the solution. It is capable of mounting an immediate, amplified, and reliable response, while avoiding  entirely adverse effects reported by the currently recommended therapeutics.

Conference Series Flu-2016 International Conference Keynote Speaker Anthony S Gilbert photo
Biography:

Anthony S Gilbert has obtained his Bachelor of Medicine and Bachelor of Surgery degree from the University of the Witwatersrand, South Africa. He is a Member of the Institute of Clinical Research. He has served as an expert Member and Vice Chair of a National Research Ethics Service (NRES) committee, having been appointed by the Health Research Authority in the U.K. As a Principal Investigator, he has supervised and conducted viral challenge studies in order to further the quest to bring safer and more effective vaccines and antivirals to the global community. His research has been published in several medical and scientific journals, including Nature Medicine.

Abstract:

Since the beginning of the millennium, human viral challenge studies have successfully been conducted at hVIVO to develop a series of well-characterized virus stocks, whilst demonstrating that the Human Viral Challenge Model (HVCM) could be effective in offering clients a faster and cost effective route to market for their therapeutics. The Human Viral Challenge Model enables global pharmaceutical and biotechnology companies, as well as leading academic groups and government institutions, to undertake scientific research, accelerate the drug development timeline and reduce the cost of bringing antiviral drugs, vaccines and diagnostics to market. The HVCM also enables fundamental research into the human response to infection and crucial research into modes of infection and transmission between individuals in the community. As hVIVO has grown and developed, the HVCM has become widely accepted as an alternative to traditional early stage field trials to show the efficacy of antiviral and vaccine therapeutics in Influenza, Respiratory Syncytial Virus (RSV) and Human Rhinovirus (HRV). By monitoring the entire disease lifecycle as subjects move from healthy to sick and recover back to healthy again, we can obtain high quality, longitudinal data from the before, during and after phases of disease. The model can be used to study the efficacy of new therapies and also to study the target disease itself.

Keynote Forum

Palayakotai Raghavan

Nanorx Inc, USA

Keynote: Metadichol® a novel ICAM-1 inhibitor

Time : 12:10-12:45

Conference Series Flu-2016 International Conference Keynote Speaker Palayakotai Raghavan  photo
Biography:

Palayakotai Raghavan is CEO and Founder of Nanorx Inc has a Ph.D. in Organic Chemistry from Oregon State University (1979) and a M.S in Chemistry (1972) from I.I.T Mumbai, India. He has worked on drug discovery for over 25 years at Columbia University, Max-Planck Institute, Germany, Ciba-Geigy (now Novartis) and Boehringer Ingelheim. He has over 12 patents and another 15 pending patent applications 

Abstract:

Metadichol (US patent 8,722,093) is a Nano emulsion of long-chain alcohols found in many foods. It is commonly called Policosanol and is present in foods such as rice, sugar cane, wheat, peanuts. Metadichol acts on Nuclear Vitamin D receptors (VDR) (US patent 9,006,292) that are present in cells throughout the body to stimulate the immune system and inhibit a variety of disease processes, resulting from viral infections.

Studies with Zucker diabetic rats showed it was an effective ICAM-1 and TNF alpha and NFKB-1 inhibitor. ICAM-1 is the same receptor molecule used by the vast majority of viruses that cause the common cold, We tested for antiviral activity of Metadichol in Vero and MDCK cells infected with Influenza A, H1N1, Human Respiratory Syncytial viruses. Metadichol, showed no cytotoxicity and strongly inhibited cell death caused by each of the viruses tested.

Metadichol is a safe and effective inhibitor of enveloped viruses in humans. Since it is known to bind to the vitamin D receptor (VDR) (US patent 9,006,292), its mechanism of action likely involves the competitive displacement of virus particles from VDR’s on host cell membranes. Because it consists of natural components of common foods and has no known negative side effects, Metadichol has the potential to serve as a safe and novel, broad-spectrum antiviral treatment for enveloped viruses. 

Conference Series Flu-2016 International Conference Keynote Speaker Manon M J Cox photo
Biography:

Manon M.J. Cox, MBA is President and Chief Executive Officer of Protein Sciences. Dr. Cox has received many honours and awards recognizing her stature as a leader in innovation and influenza including receiving a Doctorate in Humane Letters honoris causa from St. Joseph University and in 2015 elected fellow of the International Society of Vaccines. Dr. Cox holds a Doctorate from the University of Wageningen, received her MBA with distinction from the University of Nijenrode and the University of Rochester, NY and holds a Doctorandus degree in Molecular Biology, Genetics and Biochemistry from the University of Nijmegen, The Netherlands.

Abstract:

Flublok is FDA approved for the prevention of influenza in adults 18 and older. Flublok is the first recombinant hemagglutinin influenza vaccine and is produced using the baculovirus-insect cell technology. This production platform provides an attractive alternative to the current egg-based influenza vaccine manufacturing process for a multitude of reasons including speed, scale-ability, cost and independence on eggs. The key advantage of this recombinant protein manufacturing platform is that a universal “plug and play” process enables manufacturing of new vaccine candidates within a matter of months while offering the potential for low manufacturing costs. Globally large scale mammalian cell culture facilities previously established for the manufacturing of monoclonal antibodies could be deployed for the manufacturing of vaccines in the event of an emergency or alternatively, manufacturing capacity could be established in geographic regions that do not have any vaccine production capability. Dependent on health care priorities, different vaccines could be manufactured in such facilities while maintaining the ability to rapidly convert to producing pandemic influenza vaccine when the need arise. The speaker will provide an update on the global manufacturing capacity established specifically for Flublok and present recently obtained comparative efficacy data demonstrating that Flublok is more effective than tradional vaccine in protecting older adults against mismatched influenza viruses. Acknowledgment: Protein Sciences Corporation was awarded a contract in June 2009 from the U.S. Department of Health and Human Services to further develop this technology for the production of recombinant influenza vaccines for pandemic preparedness.

  • Track 1: Flu Vaccines: Current and Novel Approach Track 4: Flu Vaccine: Manufactures & Market Analysis Track 7: Flu Vaccines and Vaccination: Opportunities and Challenges for All Age Groups Track 8 : Flu Virus -Advancements in Detection & Differentiation Track 9: Flu: Immunology & Genetics
Location: San Francisco, USA
Speaker
Biography:

Anthony S Gilbert has obtained his Bachelor of Medicine and Bachelor of Surgery degree from the University of the Witwatersrand, South Africa. He is a Member of the Institute of Clinical Research. He has served as an expert Member and Vice Chair of a National Research Ethics Service (NRES) committee, having been appointed by the Health Research Authority in the U.K. As a Principal Investigator, he has supervised and conducted viral challenge studies in order to further the quest to bring safer and more effective vaccines and antivirals to the global community. His research has been published in several medical and scientific journals, including Nature Medicine.

Abstract:

Since the beginning of the millennium, human viral challenge studies have successfully been conducted at hVIVO to develop a series of well-characterized virus stocks, whilst demonstrating that the Human Viral Challenge Model (HVCM) could be effective in offering clients a faster and cost effective route to market for their therapeutics. The Human Viral Challenge Model enables global pharmaceutical and biotechnology companies, as well as leading academic groups and government institutions, to undertake scientific research, accelerate the drug development timeline and reduce the cost of bringing antiviral drugs, vaccines and diagnostics to market. The HVCM also enables fundamental research into the human response to infection and crucial research into modes of infection and transmission between individuals in the community. As hVIVO has grown and developed, the HVCM has become widely accepted as an alternative to traditional early stage field trials to show the efficacy of antiviral and vaccine therapeutics in Influenza, Respiratory Syncytial Virus (RSV) and Human Rhinovirus (HRV). By monitoring the entire disease lifecycle as subjects move from healthy to sick and recover back to healthy again, we can obtain high quality, longitudinal data from the before, during and after phases of disease. The model can be used to study the efficacy of new therapies and also to study the target disease itself.

Speaker
Biography:

Manon M.J. Cox, MBA is President and Chief Executive Officer of Protein Sciences. Dr. Cox has received many honours and awards recognizing her stature as a leader in innovation and influenza including receiving a Doctorate in Humane Letters honoris causa from St. Joseph University and in 2015 elected fellow of the International Society of Vaccines. Dr. Cox holds a Doctorate from the University of Wageningen, received her MBA with distinction from the University of Nijenrode and the University of Rochester, NY and holds a Doctorandus degree in Molecular Biology, Genetics and Biochemistry from the University of Nijmegen, The Netherlands.

Abstract:

Flublok is FDA approved for the prevention of influenza in adults 18 and older. Flublok is the first recombinant hemagglutinin influenza vaccine and is produced using the baculovirus-insect cell technology. This production platform provides an attractive alternative to the current egg-based influenza vaccine manufacturing process for a multitude of reasons including speed, scale-ability, cost and independence on eggs. The key advantage of this recombinant protein manufacturing platform is that a universal “plug and play” process enables manufacturing of new vaccine candidates within a matter of months while offering the potential for low manufacturing costs. Globally large scale mammalian cell culture facilities previously established for the manufacturing of monoclonal antibodies could be deployed for the manufacturing of vaccines in the event of an emergency or alternatively, manufacturing capacity could be established in geographic regions that do not have any vaccine production capability. Dependent on health care priorities, different vaccines could be manufactured in such facilities while maintaining the ability to rapidly convert to producing pandemic influenza vaccine when the need arise. The speaker will provide an update on the global manufacturing capacity established specifically for Flublok and present recently obtained comparative efficacy data demonstrating that Flublok is more effective than tradional vaccine in protecting older adults against mismatched influenza viruses. Acknowledgment: Protein Sciences Corporation was awarded a contract in June 2009 from the U.S. Department of Health and Human Services to further develop this technology for the production of recombinant influenza vaccines for pandemic preparedness.

Tracey Goldstein

One Health Institute, University of California, USA

Title: Diverse hosts contributing to the diversity of influenza A virus

Time : 14:05-14:30

Speaker
Biography:

Tracey Goldstein, PhD, is Associate Director and Professor at the University of California Davis, One Health Institute, where she developed and oversees the One Health Institute Laboratory and the Marine Ecosystem Health Diagnostic and Surveillance Laboratory. She is also the Co-PI and Pathogen Diagnostics Co-Lead for the viral emergence early warning project PREDICT, developed with the US Agency for International Development’s Emerging Pandemic Threats (EPT) Program.

Abstract:

Surveillance has shown aquatic birds, particularly migrating waterfowl, to be reservoirs for nearly all of the currently recognized Influenza A viruses. Recently traditional and diverse Influenza A viruses have been detected in bats and other species such as seals, suggesting multiple species may play a role in transmission and emergence. A broader surveillance approach has led to an increased detection of animal-origin infections in humans following animal contact. We performed a retrospective analysis of available influenza A sequences examining high-priority animal-origin subtypes between 2000 and 2011 from 32 different countries to identify trends in evolutionary dynamics to better understand where novel strains may next emerge and to identify data gaps in knowledge due to reporting effort. As part of ongoing surveillance under the USAID funded Predict project, we also performed global screening of samples from diverse hosts including bats and rodents globally, and through joint surveillance with NOAA and NIAID we screened for influenza A viruses in marine mammals off the Pacific coast of the United States from Alaska to California. Retrospective analyses indicated that mutation rates were higher in Asian countries, particularly in East Asia. Diverse influenzas were detected in new hosts and locations, including in bats in South America and Africa, rodents in Asia, and human and avian-origin influenzas were detected in seals off California. Finally, we identified major global gaps in influenza reporting and in broad subtype testing which will continue to hinder efforts to track the evolution and diversity of Influenza A viruses around the world.

K Kai McKinstry

University of Central Florida, USA

Title: IL-15-dependent generation of lung tissue-resident memory CD4 T cells

Time : 14:30-14:55

Speaker
Biography:

K. Kai McKinstry completed his PhD at the University of Saskatchewan in 2005 and pursued postdoctoral studies at the Trudeau Institute in Saranac Lake, NY. He joined the Faculty of the Department of Pathology at the University of Massachusetts Medical School in 2010. In 2015, Dr. McKinstry was recruited to the Burnett School of Biomedial Sciences at the University of Central Florida.

Abstract:

Tissue-resident memory T cells (TRM) are thought to play a key role in protective recall responses against pathogens. TRM subsets that persist at sites of previous infection may be especially important for immunity against pathogens such as influenza A virus (IAV) that can evade neutralizing antibodies. How TRM cells form during immune responses is not fully understood, especially for CD4+ T cells. We recently found that virtually all memory CD4 T cells that develop in secondary lymphoid organs following IAV priming require autocrine IL-2 signals. Here, we describe a unique role for IL-15 in supporting the generation of a subset of IL-2-independent lung CD4+ TRM cells formed during IAV priming. Our results demonstrate that this TRM subset is highly functional and can more efficiently elicit pro-inflammatory responses from dendritic cells presenting cognate peptide antigen than can conventional splenic memory CD4 T cells expressing the same T cell receptor. We have previously shown that this function correlates with initial control of viral titers during the first few days following IAV challenge. These studies, identifying a novel role for IL-15 in specifically supporting the priming of a subset of lung CD4 TRM cells with specialized function are highly relevant to vaccine design.

Speaker
Biography:

Thomas Voss, Ph.D., is a world-renowned thought leader in infectious disease research, and has deep expertise in virology, immunology, biodefense, and vaccine and antiviral development. His research interests include cell-virus interactions and their role in the pathogenesis of acute viral infections, as well as therapeutic and vaccine development. Voss joined SRI in 2013 from the Tulane School of Medicine’s Department of Microbiology and Immunology, where he was an associate professor from 2005 to 2012. From 1998 to 2005, he worked in various roles at Southern Research Institute, ultimately as vice president of the Homeland Security and Emerging Infectious Disease Division. Previously, Voss held roles at Carl Zeiss, Inc. (Advanced Imaging Microscopy group), the U.S. Centers for Disease Control and Prevention (Measles Virus Section, Respiratory and Enteric Virus Branch), and University Hospital in Zurich, Switzerland (Internal Medicine Division).

Abstract:

TBA

Giulio Filippo Tarro

Foundation de Beaumont Bonelli for Cancer Research, Italy

Title: Workshop : Early diagnosis and prompt therapy for emerging flu viruses
Speaker
Biography:

Giulio Filippo Tarro has graduated from Medicine School, Naples University in 1962. He is the President Foundation de Beaumont Bonelli for Cancer Research. His basic researches have been concerned with antigens induced early during the replication cycle of human herpes viruses. Another study has involved the identification, isolation and characterization of specific virus-induced tumor antigens, which were the fingerprints left behind in human cancer. He is the recipient of the Sbarro Health Research Organization Lifetime Achievement Award in 2010. His achievements include patents in field of discovery of Respiratory Syncytial Virus in infant deaths in Naples and of tumor liberated protein as a tumor associated antigen, 55 kilo Dalton protein overexpressed in lung tumors and other epithelial adenocarcinomas.

Abstract:

Since the emergence of a novel aquatic bird Flu agent in humans may be detected in near future, approaches to early diagnosis and prompt therapy are welcome. The swine-origin influenza virus (S-OIV) detected in April 2009 in Mexico, Canada and USA exhibited an unique genome composition not shown before. The emerging new Flu agent can cause outbreaks of febrile respiratory infection from mild to severe diseases throughout the world. This summary has the purpose to emphasize the possibility of tracking the new influenza virus in the most affected regions of the world and to avoid a sad toll flu-related deaths that might occurre. The possible causes of high incidence and mortality rates are going to be discussed as well as their implications on the public opinion and the prevention campaign.

Jun Wang

University of Arizona, USA

Title: Targeting the influenza A virus M2 proton channel to combat drug resistance

Time : 15:20-15:45

Speaker
Biography:

Jun Wang is Assistant Professor in Pharmacology & Toxicology department, University of Arizona, Arizona. He is awarded with PhD in Chemistry from University of Pennsylvania in 2010. He completed his MS in Chemistry from National University of Singapore, Singapore in 2006 followed by BS Chemistry from Wuhan University, China in 2003.

Abstract:

Each year more people die of flu-related illness than breast cancer, which places influenza virus infection the top ten leading causes of deaths in the U.S. In addition, sporadic and unpredictable influenza pandemics cause even more devastating damages to public health and global economy. Despite this grand challenge, we are limited by the therapeutic countermeasures against flu infection. The only class of drugs that remains effective is the viral neuraminidase inhibitors (NAs), such oseltamivir, zanamivir, and peramivir. However, like many other drugs used to treat infectious diseases, NAs are not an exception in terms of drug resistance, and flu viruses that are resistant to NAs have been continuously reported. Thus this is a pressing need to develop antivirals to combat multi-drug resistant influenza A viruses. Towards this goal, we explore the influenza A virus M2 proton channel as the drug target. Specifically, we focused on the M2-S31N mutant, which is the predominant drug-resistant mutant among current circulating influenza A viruses. Using an integrated approach involving molecular dynamics simulations, NMR, X-ray, electrophysiology, virology, and medicinal chemistry, we were able to design the first-in-class M2-S31N inhibitors. These lead compounds are not only potential drug candidates, but also invaluable chemical tools which enable us to address fundamental questions such as: Are M2-S31N inhibitors active against multi-drug resistant influenza A viruses? how is the genetic barrier of drug resistance of M2-S31N inhibitors compared to amantadine? Will mutant viruses similarly evolve resistance to M2-S31N inhibitors? If resistance were to emerge, what is the mechanism? Answers to these questions are pivotal to advance the drug discovery program forward. 

Speaker
Biography:

Tara M. Strutt received her PhD from the Department of Microbiology and Immunology at the University of Saskatchewan. Her studies focused on the signals required to activate CD4 T cells during immune responses. Dr. Strutt continued to study CD4 T cells during her postdoctoral studies at the Trudeau Institute in the laboratory of Dr. Susan L. Swain. While at the Trudeau Institute, Dr. Strutt uncovered novel protective functions mediated by memory T cells during influenza virus infection and that different protective functions are mediated by memory cells in different organs. In 2010, Dr. Strutt relocated to the University of Massachustts Medical School as an Instructor faculty member of the Department of Pathology. Her research focus centered on understanding how the tissue environment dictates T cell function, and on defining how the adaptive immune system can control innate inflammatory responses. In 2015, she started her own independent research laboratory within the Immunity and Pathogenesis Division of the Burnett School of Biomedical Sciences in the College of Medicine at the University of Central Florida where she is continuing to study how adaptive immune responses regulate innate immunity.

Abstract:

The primary goal of vaccination is to protect individuals from the morbidity and mortality of pathogen exposure by generating immunological memory. Our current understanding is that memory CD4 T cells, commonly regarded as the regulators of memory cytotoxic CD8 T cells and/or antibody-producing B cells, provide protection by hastening pathogen clearance through ‘faster, bigger, and better’ immune responses. It is now clear, however, that many different subsets of CD4 T cells, many with specialized functions other than providing ‘help’, are generated during immune responses against pathogens. In studies designed to ascertain the full functional potential of memory CD4 T cells responding against influenza A virus (IAV), we found that virus induced TH1-like, as well as in vitro polarized TH1 or TH17 memory CD4 T cells, enhance early innate inflammatory responses that correlate with better and earlier control of IAV in infected lungs. In further studies, we found innate lymphoid cells (ILC), which are not normally associated with anti-viral responses but rather associated with healing and wound repair, are also mobilized by memory CD4 T cells during the early stage of the response against IAV. Unexpectedly, the protection afforded by memory CD4 T cells against lethal IAV challenge is significantly compromised when ILCs are depleted. These findings reveal a previously unappreciated beneficial role of memory CD4 T cells in regulating tissue homeostasis during recall responses against pathogens through the regulation of ILC subsets.

Tara M. Strutt

University of Central Florida, USA

Title: Regulation of acute inflammation by memory CD4 T cells during IAV infection

Time : 15:45 - 16:15

Speaker
Biography:

Tara M. Strutt received her PhD from the Department of Microbiology and Immunology at the University of Saskatchewan. Her studies focused on the signals required to activate CD4 T cells during immune responses. Dr. Strutt continued to study CD4 T cells during her postdoctoral studies at the Trudeau Institute in the laboratory of Dr. Susan L. Swain. While at the Trudeau Institute, Dr. Strutt uncovered novel protective functions mediated by memory T cells during influenza virus infection and that different protective functions are mediated by memory cells in different organs. In 2010, Dr. Strutt relocated to the University of Massachustts Medical School as an Instructor faculty member of the Department of Pathology. Her research focus centered on understanding how the tissue environment dictates T cell function, and on defining how the adaptive immune system can control innate inflammatory responses. In 2015, she started her own independent research laboratory within the Immunity and Pathogenesis Division of the Burnett School of Biomedical Sciences in the College of Medicine at the University of Central Florida where she is continuing to study how adaptive immune responses regulate innate immunity.

Abstract:

It is generally accepted that the flow of immunologically relevant information during the early stages of responses against pathogens is one-way, - that inflammation induced upon pattern recognition by highly conserved receptors of the innate immune dramatically impacts subsequent antigen-specific T and B cell responses.  We asked if the reverse occurs, and if cells of the adaptive immune system can influence the character and magnitude of innate inflammatory responses.  We show here that resting, antigen-specific memory CD4 T cells can dramatically alter innate inflammatory responses within 36 hours of viral infection in a manner independent of other T cells and TLR signaling.  Virus-specific memory CD4 T cells transferred to naïve mice that are then challenged with influenza induce greater expression of multiple inflammatory mediators both at the site of infection and systemically upon cognate recognition of antigen in an IFN-gindependent fashion.  Our results show that the adaptive immune system can profoundly influence the character of inflammation following pathogen challenge, demonstrating a novel role for memory CD4 T cells in controlling virus titers during protective.